Next Article in Journal
The Trypanosoma brucei-Derived Ketoacids, Indole Pyruvate and Hydroxyphenylpyruvate, Induce HO-1 Expression and Suppress Inflammatory Responses in Human Dendritic Cells
Next Article in Special Issue
Mitochondrial Calcium: Effects of Its Imbalance in Disease
Previous Article in Journal
Peroxide-Mediated Oxygenation of Organic Compounds by Fungal Peroxygenases
Previous Article in Special Issue
The cAMP Inducers Modify N-Acetylaspartate Metabolism in Wistar Rat Brain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Strategies Targeting Mitochondrial Calcium Signaling: A New Hope for Neurological Diseases?

by
Laura R. Rodríguez
1,2,3,*,
Tamara Lapeña-Luzón
1,2,3,
Noelia Benetó
1,2,3,
Vicent Beltran-Beltran
1,
Federico V. Pallardó
1,2,3,
Pilar Gonzalez-Cabo
1,2,3,*,† and
Juan Antonio Navarro
4,5,*,†
1
Department of Physiology, Faculty of Medicine and Dentistry, Universitat de València-INCLIVA, 46010 Valencia, Spain
2
Associated Unit for Rare Diseases INCLIVA-CIPF, 46010 Valencia, Spain
3
Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
4
Department of Genetics, Universitat de València-INCLIVA, 46100 Valencia, Spain
5
INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2022, 11(1), 165; https://doi.org/10.3390/antiox11010165
Submission received: 14 December 2021 / Revised: 12 January 2022 / Accepted: 13 January 2022 / Published: 15 January 2022

Abstract

:
Calcium (Ca2+) is a versatile secondary messenger involved in the regulation of a plethora of different signaling pathways for cell maintenance. Specifically, intracellular Ca2+ homeostasis is mainly regulated by the endoplasmic reticulum and the mitochondria, whose Ca2+ exchange is mediated by appositions, termed endoplasmic reticulum–mitochondria-associated membranes (MAMs), formed by proteins resident in both compartments. These tethers are essential to manage the mitochondrial Ca2+ influx that regulates the mitochondrial function of bioenergetics, mitochondrial dynamics, cell death, and oxidative stress. However, alterations of these pathways lead to the development of multiple human diseases, including neurological disorders, such as amyotrophic lateral sclerosis, Friedreich’s ataxia, and Charcot–Marie–Tooth. A common hallmark in these disorders is mitochondrial dysfunction, associated with abnormal mitochondrial Ca2+ handling that contributes to neurodegeneration. In this work, we highlight the importance of Ca2+ signaling in mitochondria and how the mechanism of communication in MAMs is pivotal for mitochondrial maintenance and cell homeostasis. Lately, we outstand potential targets located in MAMs by addressing different therapeutic strategies focused on restoring mitochondrial Ca2+ uptake as an emergent approach for neurological diseases.

1. Introduction

Calcium (Ca2+) is the most ubiquitous secondary messenger in intracellular signaling of most living cells, acting as a key connection between extracellular signals and intracellular responses [1]. The most remarkable property of Ca2+ is that such a simple bivalent ion is involved in a plethora of different signaling pathways. Its versatility is achieved by its rich dynamics in concentration changes, which can be caused either by Ca2+ entry from the extracellular space or Ca2+ release from intracellular storage compartments [2] or by other side pumping Ca2+ out of the cell or to intracellular organelles. The main Ca2+ storage in mammal cells is, depending on the cell type, the sarcoplasmic–endoplasmic reticulum (SR/ER) [3]. Intracellular concentration of Ca2+ is in the range of nM, whereas extracellular Ca2+ is in the range of mM [1]. Changes in intracellular Ca2+ levels are required for different structures, cell compartments, receptors, channels, Ca2+-binding proteins, pumps, transporters, enzymes, and transcription factors [4]. In addition, when intracellular levels rise above physiological concentration, a number of deleterious cellular processes can be triggered [5].
In non-excitable cells, the pathways regulated by these Ca2+ signals encompass a wide variety of processes, including from gene expression to fertilization, secretion, protein folding, energy metabolism, and cell cycle regulation [6,7,8]. In excitable cells, the signal depends on Ca2+ entry through voltage or ligand-operated channels, which regulates muscle contraction, postsynaptic potentials, memory formation in neurons (long term potentiation), and insulin secretion from beta cells [9].
Due to the huge amount of Ca2+-dependent events occurring in cells, alteration of its signaling pathways contributes to the development of multiple human disorders. Therefore, the study of Ca2+ signaling is essential for understanding the pathophysiology of many diseases, including diabetes, carcinogenesis, cardio- and cerebrovascular diseases including endothelial dysfunction, as well as neurodegenerative disorders [4,10,11,12,13,14,15,16].
In this review, we describe the importance of Ca2+ signaling in mitochondria and how the mechanism of communication between the ER and the mitochondria is pivotal to the mitochondria. Lately, we address different therapeutic strategies targeting mitochondrial Ca2+ uptake as an emergent therapeutic approach for neurological disorders.

2. MAMs’ Composition and Function

Mitochondria and the ER are structures that experience continuous remodeling to coordinate complex mechanisms of signal transduction and gene expression, generating physical interactions that facilitate a fast and efficient regulation of these processes [17]. Termed endoplasmic reticulum–mitochondria-associated membranes (MAMs), the contact sites between the two compartments are dynamic structures that are highly sensitive to the physiological changes of the cell [18].
The association between the ER and the mitochondria was described in the 1950s, when Copeland and Dalton observed a precise orientation of the ER with respect to the mitochondria [19]. The distance between membranes in this region is 10–30 nm depending on the cell type and cell conditions [20]. Besides, it is estimated that, in physiological conditions, 5–20% of the mitochondrial surface is transiently connected to the ER and these contacts are signaling-dependent [21].
MAMs encompass an extensive variety of different proteins. The first independent proteomic studies identified 911 and 1212 proteins [22,23] localized in the tethers, but only 44% of them were common. During the last decade, different authors have contributed to increment the list by different molecular approaches, such as microscopy or subcellular fractionation [24,25,26]. The development of new techniques has facilitated the proteomic analysis of subcellular domains in-depth. The group of Alice Y Ting has recently identified more than 100 new proteins located in MAMs by means of TurboID technique. This approach was developed to study the interactome of a protein of interest in a specific cell compartment [27]. This emphasizes the complexity of these structures, specialized in each cell type and organism. Indeed, the set of proteins involved in MAMs provides important information about the functions regulated in this domain. As proteins involved in essential cellular processes belong to both the ER and the mitochondrial membranes, the contacts between the organelles enable a coordinated regulation of events, such as lipid biosynthesis [28,29], mitochondrial biogenesis [30,31,32,33], mitochondrial dynamics [34,35], and Ca2+ transfer [21,36].
Ca2+ exchange between the ER and the mitochondria requires the formation of a protein bridge composed by proteins of both compartments [37]. In particular, the formation of microdomains localized in the ER–mitochondria contact sites promotes a rapid and efficient exchange of Ca2+, fundamental for mitochondrial function, dynamics, and the regulation of apoptosis [38]. In 1993, Rosario Rizzuto and colleagues reported the increase in mitochondrial Ca2+ upon the cation mobilization through the ER channel IP3R (inositol 1,4,5-trisphosphate receptor). Recently, the spatial relation between the ER and the mitochondria was described by the same group [39]. They observed numerous close appositions between these two organelles that contributed to Ca2+ entry into the mitochondria in Hela cells [21].
The lumen of the ER is one of the main storages of free Ca2+ in the cell (about 100–500 µM) compared to the cytosol (~100 nM). Ca2+ is released to the cytosolic space upon the input signals from the ER through the IP3R and through the RyR (ryanodine receptor) in the case of the SR [40]. Furthermore, Sig-1R (Sigma non-opioid intracellular receptor 1 or shortly Sigma 1R), located in the ER, is also involved in Ca2+ signaling regulation. Sig-1R is enriched in MAMs and stabilizes activated IP3R, promoting Ca2+ influx into the mitochondria [41,42].
For the mitochondria, Ca2+ must cross both mitochondrial membranes. The outer mitochondrial membrane (OMM) is Ca2+ permeable due to VDAC (voltage-dependent anion channel), which enables different metabolites (succinate, malate, pyruvate, NADH, ATP, and phosphate) to cross from the cytosol to the mitochondria [36]. In connection with the inner mitochondrial membrane (IMM), Ca2+ enters the mitochondrial matrix through the mitochondrial calcium uniporter (MCU) since this layer is ion-impermeable [43].
In addition, another key protein that stabilizes the connections of both compartments is glucose-regulated protein 75 (GRP75), which chaperones IP3R and VDAC, maintaining the junction and ensuring an efficient transfer of Ca2+ to the mitochondria [36,44]. Altogether, all these attributes highlight MAMs as a coordinated domain that requires an optimal communication between the ER and the mitochondria.

3. Ca2+ Regulates Mitochondrial Functions

IP3R-GRP75-VDAC-MCU is one of the complexes in MAMs that is not only essential for the regulation of Ca2+ homeostasis, but also for the control of mitochondrial function in the regulation of bioenergetics, mitochondrial dynamics, and cell death [45,46]. Mitochondria are considered the powerhouse of the cell, providing at least 90% of energy in most cell types. In this context, energy requirements and, thus, mitochondrial function depend on the function of each tissue, as well as developmental and physiological conditions. Mitochondria mainly orchestrate the metabolic profile of tissues with high energy demand, such as heart, liver, kidney skeletal muscle, and brain [47]. In particular, neurons consume 70–80% of the total energy of the brain, being the remaining spent by glial cells [48]. Thus, disruption of bioenergetic pathways compromise mitochondrial function, contributing to pathological features displayed in neurological disorders.
The mechanism of ATP production depends on oxidative phosphorylation, and it is dynamically and promptly regulated by mitochondrial Ca2+ levels. Enzymes of the tricarboxylic acid cycle and the electron transport chain require an increase in mitochondrial Ca2+ uptake to promote ATP synthesis [49,50]. For instance, isocitrate dehydrogenase and oxoglutarate dehydrogenase are activated upon Ca2+ increase in the mitochondrial matrix [51,52]. Furthermore, pyruvate dehydrogenase is a key complex of oxidative metabolism that links glycolysis with the tricarboxylic acid cycle and is influenced by mitochondrial Ca2+ concentration. In this way, when energy demand increases, different neurotransmitter and hormone receptors increase mitochondrial Ca2+ through IP3R [17,53].
Mitochondrial Ca2+ uptake is also influenced by mitochondrial dynamics. Several properties of mitochondria, in terms of network, orientation, and shape, regulate the amounts of Ca2+ that reach the mitochondrial matrix and, thus, the subsequent functions regulated by these events [54].
  • The first is distribution, because clustered mitochondria are able to buffer Ca2+ more efficiently than disperse mitochondria. Mitochondrial fusion/fission events require elevated amounts of cytosolic Ca2+ to be transferred to the mitochondria [33,55]. In this way, Ca2+ can activate the cytosolic GTPase dynamin-related protein (Drp-1), which is recruited to form a ring around mitochondria to promote mitochondrial fission [31]. On the other hand, mitofusin 2 (MFN2), the GTPase responsible for the OMM fusion, also participates as a key regulator of MAMs, contributing to intracellular Ca2+ homeostasis [33].
  • Connectivity, because elongated mitochondria are better Ca2+ conductors, distributing the cation along the fused network. It has been described that, while fragmented mitochondria buffer Ca2+ from the ER in a heterogeneous manner, tubular mitochondria incorporate Ca2+ in an equilibrated and connected way [54,56].
  • Vicinity is a dynamic property that also affects Ca2+ buffering, since elevated concentrations of Ca2+ near mitochondria are required to promote mitochondrial Ca2+ uptake. In this context, Csordás et al. demonstrated the importance of spacing distance in MAMs for an efficient Ca2+ transfer, outlining that it is essential that the tethered bridge is properly assembled to ensure Ca2+ influx into the mitochondria [44,57].
  • Last but not least, the volume of mitochondria is also crucial for mitochondrial Ca2+ buffering, as forced mitochondrial expansion [57] and fragmentation reduce Ca2+ uptake capacity [58].
Furthermore, abnormal Ca2+ accumulation in the mitochondria normally precedes cell death driven by necrosis and apoptosis [59,60]. Under physiological and pathological conditions, impaired Ca2+ handling can lead to mitochondrial Ca2+ overload, thus activating the opening mitochondrial permeability transition pore. This causes mitochondrial swelling, which leads to release of cytochrome c and caspase cofactors into the cytosol [60,61].

4. Ca2+ and Oxidative Stress

Mitochondria is considered the main source of reactive oxygen species (ROS) in cells with high metabolic rates. In circumstances of mitochondrial dysfunction, an uncontrolled production of ROS would lead to an imbalance in the cellular redox state which, in turn, might likely contribute to pathogenesis [62]. Oxidative stress occurs when ROS production exceeds detoxification, causing cell damage. Thus, a balance between ROS production and antioxidant systems is crucial to maintain cell homeostasis and survival [63,64]. The electron transport chain located in mitochondrial cristae generates ROS, such as the superoxide anion (O2), which are converted to the diffusible redox signaling molecule hydrogen peroxide (H2O2) by superoxide dismutase 2 (SOD2). There is mounting evidence about the role of H2O2 as a messenger located in redox nanodomains in MAMs [65]. H2O2, mainly generated in the mitochondria, can modulate the activity of IP3R and RYR channels, promoting the release of Ca2+ from the ER to the mitochondria. This event can, in turn, induce redox signaling through the activation of mitochondrial metabolism, further inducing the accumulation of additional H2O2. This positive feedback mechanism attenuates when Ca2+ returns from the mitochondrial matrix to the ER [66,67]. This process can be beneficial or detrimental, depending on the cellular context and the levels of ROS generated. Indeed, excessive amounts of mitochondrial Ca2+ lead to high ROS levels, which may trigger cell death [65]. In addition, it has been reported that mitochondrial Ca2+ overload can inhibit H2O2 clearance, promoting its accumulation in the mitochondria [68].
Hence, this outstands the necessity of an equilibrate Ca2+ exchange ER–mitochondria through proper contacts between the two compartments. Nonetheless, a growing knowledge and a better understanding about the translational and clinical role of Ca2+ homeostasis and oxidative stress in the physiopathology of neurological diseases are required to find novel therapeutic strategies.

5. MAMs’ Communication and Neurological Diseases

As it has been discussed above, the regulation of MAMs is crucial to maintain a proper Ca2+ exchange and regulate key mechanisms in cell homeostasis. These processes (lipid metabolism, Ca2+ homeostasis, mitochondrial dynamics, and axonal maintenance) are usually involved in the physiopathology of neurodegeneration. Mitochondrial dysfunction results in abnormal Ca2+ handling, leading to alterations in axonal transport, bioenergetics, redox status, contractility, and cell viability [69,70,71]. It is clear that ER–mitochondria communication is very important for axonal survival and degeneration. In fact, MAMs are functionally implicated in axons, dendrites, and neuronal soma, modulating and maintaining synaptic activity [72,73,74,75]. For this reason, ER–mitochondria assembly has been proposed as a common mechanism in neurodegenerative disorders [76,77,78,79].
The first proteomic description of MAMs detected proteins involved in mitochondrial dysfunction as well as in neuromuscular and degenerative diseases, including Huntington’s disease (HD), Parkinson’s disease (PD), and Alzheimer’s disease (AD) [23]. Nowadays, it is well described that the proteins involved in these diseases are resident in MAMs [80,81,82,83]. Specifically, these diseases exhibit increased ER–mitochondria contacts, leading to toxic mitochondrial Ca2+ uptake, reduced cell viability, and dysfunction in the subsequent mechanisms regulated in this domain [77,78,79].
Conversely, several neuropathies exhibit reduced ER–mitochondria connections and Ca2+ accumulation in the cytosol, impairing the communication between the two compartments and displaying mitochondrial alterations. Several forms of amyotrophic lateral sclerosis (ALS) are characterized by mitochondrial disruption and abnormal mitochondrial Ca2+ handling. This includes ALS forms with mutations in superoxide dismutase 1 (SOD1), in which mouse models exhibit mitochondrial defects and deficits in mitochondrial Ca2+ uptake [84,85]. Homozygous mutations in the ER protein Sig-1R are the cause of the juvenile form of ALS16 [86], showing ER–mitochondria dissociation in motor neurons, as well as a reduction in mitochondrial Ca2+ influx via IP3R, and lower ATP production. These defects lead to neuron vulnerability, directly associated with the physiopathology of the disease [87].
Accordingly, dominant cerebellar ataxias are multifactorial and progressive diseases with common mechanisms of mitochondrial dysregulation, Ca2+ handling defects, and oxidative stress that contribute to neurodegeneration [88]. Specifically, spinocerebellar ataxias type 2 and 3 (SCA2/3) have been found to exhibit mutations in the IP3R channel, which leads to the abnormal Ca2+ release to the cytoplasm, potentially inducing Ca2+ buffering defects in mitochondria [89]. Interestingly, different models of the recessive neuromuscular disorder Friedreich’s ataxia (FRDA) exhibit cytosolic Ca2+ accumulation, as well as impaired mitochondrial Ca2+ uptake and decreased inter-organelle interactions in MAMs. The protein involved in the disease, frataxin, has recently been found as a member of the protein network of MAMs, interacting with GRP75 and IP3R [90,91,92]. Charcot–Marie–Tooth (CMT) is an inherited neuropathy caused by mutations in an important number of proteins related to mitochondrial function. Specifically, the causative genes for CMT type 2 are ganglioside-induced associated protein 1 (GDAP1) and MFN2 [93], which encode proteins located in the OMM that contribute to MAM’s function. Deficiency in GDAP1 leads to neuronal Ca2+ and mitochondrial defects, coupled with altered interplay between ER–mitochondria and Ca2+ accumulation in the cytosol [71,94,95]. Hereditary spastic paraplegia is associated with alterations in genes related to the ER that also affect the axonal transport of mitochondria, including IP3R, suggesting impaired ER–mitochondria communication [96,97,98].

6. Therapeutic Approaches Targeting MAMs

Mitochondrial Ca2+ modulation is fundamental to maintain the physiological mechanisms that regulate metabolism, mitochondrial dynamics, and cell death. Therefore, MAMs emerge as potential therapeutic targets of neurological disorders. In this review we will focus on different therapeutic approaches (see Figure 1 for further information [99]) aimed to stabilize the ER–mitochondria assembly and, thus, promote Ca2+ influx into the mitochondria.

6.1. Sigma-1 Receptor as a Therapeutic Target

Sig-1R is a Ca2+-sensitive chaperone located in the ER membrane, specifically in MAMs, and regulates Ca2+ homeostasis, lipid dynamics, MAMs’ stability, and the ER stress response [41,100]. Sig-1Rs of different species share a very high sequence identity (>93%), but share no sequence homology with any other mammalian protein. Interestingly, the Sig-1R displays a 30% identity, a 67% homology, and a similar ligand profile to the yeast sterol isomerase encoded by the ERG2 gene [101,102]. This conservation points towards Sig-1R as a fundamental protein for cell functioning. It is highly expressed in the central nervous system, playing a key role in physiological functions, such as cell differentiation, axon formation, microglial activation, and astrocyte regulation [103].
Sig-1R resides specifically on ceramide and cholesterol-rich lipid microdomains at MAMs, acting as an inter-organelle Ca2+ signaling modulator and exerting a pivotal role in neuroprotection and neuroplasticity [104]. In physiological conditions, Sig-1R forms a complex with the chaperone binding immunoglobulin protein/glucose response protein 78 (BiP/GRP78), in a dormant, Ca2+-dependent state. Once activated by agonists or ER Ca2+-depletion, Sig-1R dissociates from BiP and reallocates within the ER membrane, interacting with different proteins, such as IP3R. Then, IP3R is prevented from degradation and promotes mitochondrial Ca2+ uptake (see Figure 1 for further information [99]) [42]. Thus, Sig-1R is a key element for maintaining the structure and function of MAMs. Besides, during ER stress response, Sig-1R expression increases, which prevents cells from apoptosis triggered under such conditions [105]. Sig-1R has been recently identified as indispensable for mitochondrial bioenergetics during early ER stress, which gives rise to an increased ER–mitochondrial Ca2+ exchange [106]. Hence, targeting Sig-1R may regulate ER stress, a common mechanism displayed in neurodegeneration [107]. Interestingly, Sig-1R ligands have been found to potentially offer protection against the most severe symptoms of SARS-CoV-2, providing mitochondrial protection, activating mitophagy, preventing ER-stress, managing Ca2+ transport, and inducing autophagy to prevent cell death in response to infection [108].
Recently, the basic structural pharmacophore of Sig-1R has been identified, which is critical for drug development. There is evidence about different shifting monomerization–oligomerization states of Sig-1R modulated by its ligands. In this sense, agonists and antagonists regulate the association between Sig-1R and BiP, controlling the interactome of Sig-1R [105,109,110]. The regulation of these mechanisms has a pivotal role in the context of MAMs and neurodegenerative diseases [111]. Transcriptional upregulation of Sig-1R induces its neuroprotective properties. For instance, it has been reported that Sig-1R upregulates the expression of the antiapoptotic mitochondrial protein Bcl-2 (B-cell lymphoma 2), preventing neuronal cell death [112]. On the other hand, Sig-1R has been involved in the protection of cellular oxidative stress and the activation of antioxidant response elements [113]. In this line, overexpression of Sig-1R enhanced resistance to oxidative stress in Drosophila melanogaster [114]. Conversely, knockdown of Sig-1R leads to increased ROS and decreased expression and activity of nuclear factor erythroid 2-related factor 2 (NRF2), the protein implicated in the activation of the intracellular antioxidant response [115]. These data point Sig-1R as a potential therapeutic target against oxidative stress-related diseases.
While Sig-1R deficiency exacerbates progression of neurological disorders and symptoms commonly associated to neurodegenerative disorders [116,117,118], many Sig-1R agonists exert anti-amnestic, synaptogenesis, and neuroprotective effects under neuronal stress conditions [119,120]. Indeed, the absence of Sig-1R leads to motor neuron degeneration, associated with reduced ER–mitochondria contacts, disturbed mitochondrial dynamics, and intracellular Ca2+ dyshomeostasis [121]. Remarkably, pharmacological modulation of Sig-1R has been demonstrated to mitigate disease and symptoms in different models of ALS, AD, PD, and HD (reviewed in [105]). Given the chaperone nature of Sig-1R, its activity in targeting conformationally misfolded proteins occurs only under such conditions. Indeed, it has been described that Sig-1R activation only exhibits therapeutic effects under pathological conditions and has no effect in control animals [122,123]. This provides evidence about the specificity of Sig-1R ligands in pharmacotherapy.

6.1.1. Pridopidine

Pridopidine (ACR16) is a selective Sig-1R agonist and a dopamine stabilizer [124,125]. Great focus has been given to pridopidine in recent years, especially in HD, where it has demonstrated to improve motor function in both animal models and patients, protecting neurons from mutant huntingtin toxicity [126]. For instance, Ryskamp and colleagues evaluated the relevance of Sig-1R as a therapeutic target of pridopidine in HD. Action of pridopidine is mediated by Sig-1R, leading to restoration of IP3R-dependent Ca2+ release and upregulation of key Ca2+-regulating genes [126]. Furthermore, early treatment of pridopidine prior to the appearance of disease phenotypes improved motor coordination and reduced anxiety and depressive-like phenotypes in the YAC128 HD mice model, whereas late treatment only rescued depressive-like symptoms [126]. Interestingly, the research carried out by Naia et al. using different models of HD has highlighted the effects of pridopidine in mitochondrial function, contributing to neuroprotection mediated by Sig-1R. Authors reported that pridopidine prevented the disruption of ER–mitochondria contacts, also improving the colocalization of IP3R and Sig-1R with mitochondria in YAC128 neurons. Accordingly, this compound increased mitochondrial activity, elongation, and motility. In both HD human neural stem cells and YAC128 neurons, pridopidine increased mitochondrial respiration, rescued antioxidant response, and decreased mitochondrial ROS levels caused by Sig-1R knockdown. Additionally, apart from the improvement in motor coordination, YAC128 mice treated at early/pre-symptomatic age with pridopidine showed a reduction in mitochondrial ROS levels. Overall, these results highlighted the effects of pridopidine in mitochondrial function, contributing to neuroprotection mediated by Sig-1R [126].
Regarding clinical evaluation, pridopidine has demonstrated some improvements in motor symptoms. A phase-III randomized, double-blind, multicenter trial study (NCT00665223, MermaiHD) failed to provide evidence about the effects of pridopidine in the primary outcome (changes in modified Motor score). Despite this, some parameters related to motor scales improved significantly in the 90 mg/day group [127]. According to these results, a randomized, double-blind, placebo-controlled, multicenter trial (HART study, NCT00724048) could not reach its primary endpoint after 12 weeks of treatment. Nevertheless, patients treated with the highest doses improved in secondary motor evaluation, suggesting modest beneficial effects of pridopidine in HD [127]. Altogether, these results suggest the reproducible positive effects of pridopidine on motor symptoms in HD. Currently, a phase-III, randomized, double-blind, placebo-controlled study is recruiting patients to evaluate the efficacy and safety of pridopidine in patients with early-stage HD (NCT04556656) [128].
The efficacy of pridopidine has been tested in in vitro and in vivo models of ALS, due to the fact that different forms of ALS are caused by mutations in the Sig-1R gene [87,129]. Authors demonstrated beneficial effects of pridopidine-targeting Sig-1R on axonal transport perturbations, neuromuscular junction disruption, and motor neuron death. In addition, pridopidine slowed the progression of the disease in an ALS mouse model [130]. There is currently an ongoing phase-II clinical trial on the efficacy of pridopidine in ALS patients (NCT04615923) [131].
Recently, pridopidine has demonstrated neuroprotective and neurorestorative effects in nigrostriatal dopamine neurons via Sig-1R in an animal model of PD [132]. A phase-II, double-blind, parallel-group study started in 2019 with the aim of assessing two doses of pridopidine in levodopa-induced dyskinesia patients with PD (gLIDE study, NCT03922711) [133]. Despite the pending results, it seems that the study was cancelled early due to COVID-19 pandemic.

6.1.2. SA4503

SA4503 or cutamesine is an orally available, potent, and selective Sig-1R agonist that exhibits antiarrhythmic and antidepressant effects [134]. It has been reported to alleviate mitochondrial dysfunction, recovering ATP production in a dose-dependent manner and mobilizing intracellular Ca2+ into the mitochondria. These effects attenuated neuronal apoptosis [135] and ameliorated cardiac hypertrophy [136]. In addition, SA4503 promoted the survival of cortical neurons from oxidative-stress-induced cell death [137]. In ALS models, this compound has shown to suppress motor neuron degeneration and symptom progression [137]. In this line, SA4503 also enhanced the cytosolic Ca2+ clearance in motoneurons and IP3R-mediated ER Ca2+ release in ALS mice [138]. In alpha-thalassemia X-linked intellectual disability, SA4503 reversed axonal development and dendritic spine abnormalities in cultured cortical neurons, as well as cognitive deficits exhibited in the mice model [139].
In the clinical field, SA4503 has been evaluated in two trials. In 2008, the safety and efficacy of SA4503 was assessed in subjects with major depressive disorder (NCT00551109) [140]. Results and outcomes are pending. Furthermore, a phase-II, double-blind, placebo-controlled, ascending dose study evaluated the safety and motor function restoration in subjects with acute ischemic stroke (NCT00639249). Even though it was safe and well tolerated, no significant improvements were found [139].

6.1.3. Blarcamesine

Blarcamesine (ANAVEX2-73) is a safe Sig-1R agonist and muscarinic receptor modulator with preliminary efficacy evidence in patients with AD and Rett syndrome [141]. Preclinically, blarcamesine exerted anticonvulsant, anti-amnesic, neuroprotective, and antidepressant effects in various animal models of Rett syndrome [141], fragile X syndrome [141], AD [142,143], and amnesia [144]. These data suggest its potential in neurodegenerative and neurodevelopmental diseases.
After demonstrating good safety, bioavailability, and tolerability in AD patients (NCT02244541) [145], blarcamesine has been tested in a phase-II, placebo-controlled study with Rett Syndrome patients (NCT03758924) [146]. Even though the trial is completed, results are still pending. Currently, different phase-II/III trials focused on PD (NCT04575259) [147], AD (NCT04314934) [148], and Rett Syndrome (NCT03941444, NCT04304482) [149,150], which are recruiting patients to evaluate the tolerability and efficacy of blarcamesine.

6.1.4. PRE-084

PRE-084 is a selective Sig-1R agonist that has demonstrated promising effects against oxidative stress and modulating intracellular Ca2+ levels in preclinical studies using different disease models. For instance, PRE-084 exerted protective action against oxidation and improved viability in human retinal cells [151]. In a HD cell model, pre-treatment with PRE-084 resulted in the prevention of caspase 3-cleavage, stimulation of cellular antioxidants, and a reduction in ROS in mutant huntingtin-expressing neuronal cells [152].
Furthermore, in a model of AD, treatment with PRE-084 restored mitochondrial respiratory dysfunction in mouse hippocampus and prevented increases in lipid peroxidation levels and apoptosis markers [143]. In this line, Watanabe S. et al. evaluated a Ca2+ influx into the mitochondria and ATP levels after incubating motor neurons of ALS with PRE-084. In all the experiments, this compound restored the function of IP3R impaired in the model, suggesting that Sig-1R activation by PRE-084 prevented the disruption of Sig-1R-IP3R interaction. In addition, intraperitoneal administration of PRE-084 in pre-symptomatic ALS mice successfully restored co-localization of Sig-1R and IP3R analyzed in neurons of the lumbar spinal cord. These data indicate that Sig-1R activation is crucial to prevent MAMs disruption and regulate the Ca2+ exchange between the ER and the mitochondria via IP3R [87]. PRE-084 improved locomotor function and motor neuron survival in pre-symptomatic and early symptomatic mutant ALS mice. Authors reported a promising strategy of pharmacological manipulation of Sig-1R, pointing to an increased availability of growth factors, as well as modulation of astrocytosis and macrophage–microglia as part of the mechanisms involved in Sig-1R-mediated neuroprotection [87]. Nevertheless, one of the studies aforementioned about the treatment of ALS motor neurons with SA4503 also tested PRE-084. Contrary to SA4503, PRE-084 did not reduce the cytosolic Ca2+ levels [138]. These findings indicate that different Sig-1R ligands may have different effects on MAMs-regulated Ca2+ homeostasis.

6.1.5. Fluvoxamine

Fluvoxamine is a selective serotonin reuptake inhibitor with high affinity for Sig-1R that has been widely used in clinical practice as an antidepressant. Fluvoxamine has shown to increase Sig-1R expression, inducing neuroprotection and protecting cells from ER-stress [153]. This compound has also been found to rescue impaired mitochondrial Ca2+ uptake and ATP production in hypertrophic cardiomyocytes [154] and protect against cardiac dysfunction [155]. Furthermore, activation of Sig-1R by fluvoxamine has been demonstrated to activate several antioxidant pathways [155,156]. For instance, in brain and liver of oxidative stress-induced mice, fluvoxamine alleviated lipid peroxidation and oxidative stress by reducing malondialdehyde and nitric oxide levels and increasing reduced glutathione (GSH) [157]. Accordingly, this drug exerted anti-inflammatory and antioxidant properties by enhancing GSH levels and reducing the nitric oxide-dependent oxidative marker 3-nitrotyrosine [158]. Special attention has been paid lately to fluvoxamine regarding COVID-19. Clinically, this compound is well tolerated and widely available. Its mechanisms of action are involved with the hallmarks of severe COVID-19 (reviewed in [159]). At the moment, fluvoxamine has not been used in the clinical field beyond antidepressant or COVID-19 purposes.

6.2. Mitochondrial Calcium Uniporter as a Therapeutic Target

The mitochondrial calcium uniporter (MCU) is one of the most important and highly selective Ca2+ transporting complexes [160]. MCU is a crucial element of MAMs, acting as a gatekeeper of Ca2+ and controlling the mitochondrial Ca2+ influx. In fact, it has been determined that MCU and IP3R must have optimal distance to ensure mitochondrial Ca2+ signaling in physiological conditions [161]. Located in the IMM, this pore-protein is composed of several subunits. While the pore-forming and Ca2+-conducting subunit of the MCU complex is named Mcu, the regulatory subunit encompasses MICU1; MICU2; mitochondrial calcium uptake protein 1, 2, 3 (MICU3); essential MCU regulator (EMRE); mitochondrial calcium uniporter regulator 1 (MCUR1); and mitochondrial calcium uniporter regulatory subunit (MCUb) [162,163].
Specifically, MICU1 is an important gatekeeper located towards the mitochondrial matrix, next to the IMM. MICU1 controls the mitochondrial Ca2+ influx by interacting with Mcu, so when binding to Ca2+, its conformation changes from hexamers to oligomers and activates MCU [164,165]. Knock out of MICU1 in mice causes significant mortality, marked ataxia, and muscle weakness. Furthermore, patients with mutations in MICU1 exhibit brain and muscle disorders, proximal myopathy, learning difficulties, and a progressive extrapyramidal movement disorder [166]. Besides, MICU2 is also a gatekeeper and exhibits an inhibitor role of Ca2+ uptake, preventing Ca2+ to cross the IMM unless it is above the threshold. In fact, elevated Ca2+ levels in the intermembrane space are required to reach the mitochondrial matrix through MCU due to its low Ca2+ affinity [167]. MICU3 displays the same role as MICU2 but, in comparison, has a different tissue-dependent expression pattern [168]. EMRE is a single-pass transmembrane protein that functions as a positive regulator of MCU, and its interaction with MCU is essential for mitochondrial Ca2+ uptake, acting as a gatekeeper and preventing mitochondrial Ca2+ overload [169,170]. MCUR1 exerts a scaffold role, binding Mcu and EMRE [171,172]. MCUb is the negative regulatory subunit of MCU since its overexpression leads to a decreased Ca2+ intake by the MCU [173].
In the last decade, special interest has been taken in the modulation of MCU activity as a novel therapeutic target. Most current therapeutic approaches are focused on the inhibition of ER–mitochondria Ca2+ exchange, mainly in cancer [174,175] and neurodegenerative disorders [176] (such as AD [177] and PD [178]).
However, diseases such as type 2 diabetes point towards MCU activation to alleviate mitochondrial dysfunction associated with dysregulation in intracellular Ca2+ homeostasis, MAMs disruption, and defects in several functions regulated in this domain [179,180]. Targeting the MCU has been demonstrated to be beneficial in models with impaired ER–mitochondria inter-organelle communication. For instance, MCU activation by spermine increased cytosolic Ca2+ clearance by promoting mitochondrial Ca2+ buffering capability in cardiomyocytes with cardiac hypertrophy. In this cellular model, SR–mitochondria connections were decreased by the inhibition of Mfn2, leading to inhibition of ATP synthesis and contributing to pathogenesis [181].
Besides, genetic modulation of MCU expression has proved to modulate mitochondrial Ca2+ uptake [182]. In this line, either MCU overexpression or MCU activation by spermine reversed Pb2+-induced oxidative stress and inhibition of mitochondrial Ca2+ uptake in SH-SY5Y human neuroblastoma cells [183]. The promotion of mitochondrial Ca2+ import through MCU overexpression in glia recovered degenerative phenotypes and ATP production in a FRDA Drosophila melanogaster model [90].

Kaempferol

The most relevant MCU enhancer described until now is Kaempferol, which has demonstrated to enhance MCU, promoting Ca2+ import into the mitochondria and exhibiting neuro and cardioprotective properties [184,185,186,187,188]. In 2004, Montero and collaborators identified different flavonoids with the effect of stimulating mitochondrial Ca2+ entry through MCU activation. The most active compound was Kaempferol, which increased the rate of mitochondrial Ca2+ uptake by 85-fold [189]. In addition, Kaempferol modulates autophagy to protect cells from malfunction and regulates ER stress [190,191]. Special attention has been paid to Kaempferol recently due to its anti-inflammatory and antioxidant properties. For instance, Kaempferol was able to induce NRF2 expression in brain tissues [184]. A recent study suggests that Kaempferol augmented the phosphorylation of PIK3 and Akt, thus allowing Keap-1 to release NRF2 and promote antioxidant response in the nucleus [192].
Accordingly, in cerebellar granule cells, Kaempferol prevented cells from apoptosis, exerting potent effects by blocking ROS production [193]. In ischemia–reperfusion injury models, Kaempferol reduced mitochondrial dysfunction and reduced oxidative stress by decreasing ROS and malondialdehyde levels, whereas GSH and GSH peroxidase levels increased significantly [194,195,196]. In a D. melanogaster model of PD, kaempferol delayed degenerative phenotype onset in a dose-dependent manner, accompanied by a reduction in oxidative stress markers [197]. Similar effects on phenotype and oxidative stress were reported in a D. melanogaster model of AD [198]. Indeed, a study conducted with 921 participants concluded that kaempferol and other flavonoids are associated with lower risk of developing AD [199]. Kaempferol was proposed as a bone-fide candidate for the design of therapeutic approaches against familial ALS from a computational perspective through molecular docking, quantum chemical studies, and molecular dynamics [200].

6.3. Other Approaches

In addition to the aforementioned approaches, other compounds may be suitable to increase mitochondrial Ca2+ uptake without a specific ER–mitochondria target.

6.3.1. Taurine

Taurine is a sulfur-containing amino acid present in abundance in many excitable tissues, including the brain, skeletal, and cardiac muscles. Physiological actions of taurine include membrane stabilization, neurotransmission, and modulation of cellular Ca2+ levels [201]. El Idrissi and Trenkner evaluated the neuroprotective role of taurine in the regulation of mitochondrial Ca2+ buffering in cerebellar granule cells. They demonstrated an active role in the regulatory mechanisms of Ca2+ homeostasis, suggesting an enhancement in mitochondrial function and regulation of intracellular Ca2+ [202,203]. Besides the suggestion of a selective mechanism of taurine in mitochondrial Ca2+ uptake enhancement, though the specific receptor of taurine is not known.

6.3.2. Nerve Growth Factor

Nerve growth factor (NGF) is a secreted neurotrophin involved in survival, maintenance, and regeneration of specific types of neurons in the central and peripheral nervous system [204]. It has been highlighted as a potential therapeutic option for neurodegeneration due its role in apoptosis prevention [205], mitochondrial dysfunction protection [206], mitochondrial remodeling, and intracellular Ca2+ mobilization [207,208]. In the context of Ca2+ signaling, NGF has been demonstrated to increase cytosolic free Ca2+ concentration in C6-2B glioma cells and PC12 cells [209], as well as mitochondrial Ca2+ [207]. NGF was assessed in an open-label, dose-escalation study of encapsulated cell biodelivery of NGF in AD patients. Authors reported that preliminary data of the NGF treatment seemed to slow the rate of atrophy depending on the subtype of AD [210]. For this reason, NGF treatment should be further investigated for neuronal support.

6.3.3. MiCUps

Mitochondrial Ca2+ uptake enhancers (MiCUps) are a group of compounds recently identified to increase mitochondrial Ca2+ import, especially in cardiomyocytes. Using molecular ligand–protein docking and mutational analysis, a recent study has determined Efsevin as a MiCUp that shifts the opening of the VDAC2 channel, promoting Ca2+ entry to the mitochondria [211]. It has been mostly used in cardiac models, due to its ability to regulate cardiac rhythmicity [187,212]. Ezetimibe and disulfiram have been recently identified as MiCUps, proving to efficiently suppress arrhythmogenesis in different experimental models. The identification of such compounds underscores mitochondrial Ca2+ uptake as a pharmacological target [213].

6.3.4. Antioxidants

Targeting the mitochondrial redox state could be a suitable therapeutic strategy to recover ER–mitochondria communication. In this context, compounds may modulate mitochondrial Ca2+ signaling to stabilize its redox state or directly target mitochondrial ROS [214]. The first strategy has been discussed before; many of the aforementioned compounds exert antioxidant properties in addition to its main mechanism of action, which can readjust the ER–mitochondria Ca+2 flux and, thus, ensure a correct balance in the ROS–antioxidant system. In the context of the second strategy, treatment with antioxidants has been demonstrated to recover ER–mitochondria communication and Ca2+ exchange between the two compartments. Vitamin E restored mitochondrial Ca2+ uptake in a cardiomyocyte model of FRDA [91]. Accordingly, trolox (mimic of vitamin E) and N-acetylcysteine were able to recover both function and structure of MAMs in a neuronal model of FRDA [90], suggesting an oxidative environment in MAMs is implied in the pathophysiology of the disease. In essence, the use of antioxidants may be a good strategy to both reduce redox environment in MAMs and potentiate Ca2+ influx in the mitochondria.
On the other hand, when mitochondrial function is impaired, it is realistic to assume the involvement of different effectors and mechanisms. For this reason, some authors point towards the use of a combination of antioxidants also known as ‘mitochondrial cocktails’, due to its irrelevant toxicity, providing relevant benefits by increasing the spectrum of action [215,216,217,218].
SS-31 or elamipretide is an aromatic–cationic tetrapeptide that readily penetrates cell membranes and transiently localizes to the inner mitochondrial membrane. This mitochondrial-targeted agent was found to interact with cardiolipin, an anionic phospholipid located in the IMM and required for cristae formation. In addition, SS-31 accelerates ATP recovery and increases the enzymatic activities of Fe-S enzymes, including aconitase and complex II and III of the respiratory chain [219,220]. Another study suggested that SS-31 could have therapeutic potential effects in preventing damage from oxidative stress in neurocognitive disorders [221]. SS-31 has demonstrated to be safe and well tolerated. It has been tested in a crossover clinical trial evaluating its efficacy in primary mitochondrial myopathy for 4 weeks. Patients experienced a clinically meaningful change in the primary endpoint, which was not significant [222]. Nonetheless, these and other relevant results provided efficacy to support the initiation of a 6-month-long, phase-III study. Another clinical trial (NCT04689360) is currently recruiting patients with genetically confirmed rare diseases with known mitochondrial dysfunction [223].
Mitoquinone or MitoQ is an orally active mitochondria-targeted antioxidant that mimics the role of the endogenous mitochondrial antioxidant coenzyme Q10 [224]. In addition, MitoQ has demonstrated to substantially increase the antioxidant capacity of coenzyme Q10 by modulating oxidative stress via activating the NRF2 pathway [225,226] and increasing GSH levels [227]. Treatment in vitro and in vivo with MitoQ has provided evidence about its beneficial effects in neuroprotection, as well as in restoring mitochondrial dynamics, bioenergetics, and the redox state [228,229]. MitoQ is currently being evaluated in several clinical trials, including its assessment in AD (NCT03514875) [230], vascular function (NCT02966665) [231], and multiple sclerosis (NCT04267926) [232], among others.
These and other antioxidants targeting mitochondrial dysfunction in neurodegeneration have been extensively reviewed by several authors, including us [218,233,234,235,236,237,238].

7. Conclusions and Future Perspectives

Since mitochondria are intracellular dynamic compartments involved in multiple mechanisms, the in-depth study of these mitochondrial-dependent pathways is crucial to understand the pathophysiology of neurological and neuromuscular disorders. Particularly, we highlight the importance of crosstalk communication between the ER and the mitochondria in intracellular Ca2+ homeostasis and, therefore, in cell physiology. Since MAMs are the structures carrying out such communication, their disruption involves dramatic consequences that not only affect mitochondrial mechanisms, but also a plethora of intracellular signaling pathways. An example of such disarrangement is exerted in neurological disorders, as the proteins involved in these diseases are part of the protein network of MAMs [86,90,239].
On the other hand, proteins belonging to MAMs are a growing list, which contributes to the idea of the complexity and dynamism of these structures and, thus, the mechanisms involved in its regulation. For this reason, it is also important to determine the properties of MAMs in different cell types under distinct cellular conditions. The elucidation of these pathways will provide valuable information about the physiopathology of diseases that present impaired ER–mitochondria communication, opening new fields of research to identify adequate treatments for patients.
We believe that restoration of MAMs communication may be a suitable strategy to reverse this impairment. In addition, some patients suffering from neuromuscular diseases usually undergo heart conditions [240,241,242]. Interestingly, the activation of targets, such as Sig-1R, has demonstrated to have both neuro and cardioprotective effects. As compounds such as pridopidine and blacarmesine are currently being evaluated in clinical trials, the results obtained may be applicable to other diseases with common impaired mechanisms. Furthermore, oxidative stress is a common hallmark in neurological disorders [234,243,244], so the activation of antioxidant mechanisms has always been a common therapeutic strategy. The fact that many compounds targeting mitochondrial Ca2+ uptake can, therefore, exert antioxidant properties, making them more versatile in restoring the molecular defects involved in these diseases.
In terms of future therapeutic approaches, special attention is being paid to miRNAs. These small molecules regulate genetic expression by binding to its target mRNA and can be detected in biological fluids. The identification of miRNA signatures could provide valuable information about diagnosis or prognosis, even in the early stages of the disease. Besides, this should address the nature of targets and the extent of the biological regulation of the occurring pathways. For instance, miR-20b has been found to mediate MFN2 signaling, which can impair ER–mitochondria Ca2+ crosstalk and contribute to cardiac hypertrophy [181]. This opens a new field of therapy development in MAMs communication.
Neurological diseases, such as ALS, CMT, and FRDA, have no cure, so it is essential to find therapeutic approaches that can improve patient’s wellbeing by either slowing the onset of symptoms and/or delaying the progression of the disease, in addition to physical therapy, occupational therapy, and surgery. The repurposing of compounds outlined in this review could be a good strategy to test in vitro and in vivo models of disease. Furthermore, the use of new in silico approaches, such as pharmacophore modeling and molecular docking, are useful to ensure optimal molecular interactions with a specific biological target. This may contribute to identifying new targets, drug discovery, and optimization for specific treatments [245,246,247].

Author Contributions

L.R.R., T.L.-L., N.B., V.B.-B., F.V.P., P.G.-C. and J.A.N. participated in the preparation of the manuscript and read and approved the final document. L.R.R., P.G.-C. and J.A.N. coordinated and supervised the production of the final document. P.G.-C. and J.A.N. participated in project administration and funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the Ministerio de Economía y Competitividad de España [Grant no. PID2020-115190RB-100] within the framework of the National R+D+I Plan and co-funded by the Instituto de Salud Carlos III (ISCIII)-Subdirección General de Evaluación y Fomento de la Investigación and FEDER funds (PI19/01084); the Generalitat Valenciana (PROMETEO/2018/135); ACCI-2019-22 and ACCI-2020 from (CIBERer-ISCIII). CIBERER is an initiative developed by the Instituto de Salud Carlos III in cooperative and translational research on rare diseases. Conselleria de Sanitat Universal i Salut Pública de la Generalitat Valenciana (Plan GenT CDEI-04/20-C).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AD—Alzheimer’s diseaseALS—Amyotrophic lateral sclerosis
Bcl-2—B-cell lymphoma 2BiP/GRP78—Binding immunoglobulin protein/Glucose response protein 78
Ca2+—CalciumCMT—Charcot–Marie–Tooth
Drp-1—Dynamin -related proteinEMRE—Essential MCU regulator
FRDA—Friedreich’s ataxiaGDAP1—Ganglioside-induced associated protein 1
GRP75—Glucose-regulated protein 75GSH—Glutathione
H2O2—Hydrogen peroxideHD—Huntington’s disease
IMM—Inner mitochondrial membraneIP3R—Inositol 1,4,5-trisphosphate receptor
MAMs—Endoplasmic reticulum–mitochondria-associated membranesMCU/Mcu—Mitochondrial calcium uniporter
MCUb—Mitochondrial calcium uniporter regulatory subunitMCUR1—Mitochondrial calcium uniporter regulator 1
MFN2—Mitofusin 2MICU1/2/3—Mitochondrial calcium uptake protein 1, 2, 3
MiCUps—Mitochondrial Ca2+ uptake enhancersNGF—Nerve Growth Factor
NRF2—Nuclear factor erythroid 2-related factor 2OMM—Outer mitochondrial membrane
PD—Parkinson’s diseaseROS—Reactive oxygen species
RyR—Ryanodine receptorSCA2/3—Spinocerebellar ataxias type 2 and 3
Sig-1R—Sigma non-opioid intracellular receptor 1SOD1—Superoxide dismutase 1
SOD2—Superoxide dismutase 2SR/ER—Sarcoplasmic/endoplasmic reticulum
VDAC—Voltage-dependent anion channel

References

  1. Skupin, A.; Thurley, K. Calcium signaling: From single channels to pathways. Adv. Exp. Med. Biol. 2012, 740, 531–551. [Google Scholar] [CrossRef]
  2. Berridge, M.J.; Lipp, P.; Bootman, M.D. The versatility and universality of calcium signalling. Nat. Rev. Mol. Cell Biol. 2000, 1, 11–21. [Google Scholar] [CrossRef]
  3. Taylor, C.W. Controlling calcium entry. Cell 2002, 111, 767–769. [Google Scholar] [CrossRef] [Green Version]
  4. Islam, M.S. Calcium Signaling: From Basic to Bedside. In Advances in Experimental Medicine and Biology; Springer: Berlin/Heidelberg, Germany, 2020; Volume 1131, pp. 1–6. [Google Scholar]
  5. Farber, J.L. The role of calcium ions in toxic cell injury. In Proceedings of the Environmental Health Perspectives. Env. Health Perspect. 1990, 84, 107–111. [Google Scholar] [CrossRef] [PubMed]
  6. Rizzuto, R.; Duchen, M.R.; Pozzan, T. Flirting in little space: The ER/mitochondria Ca2+ liaison. Sci. STKE 2004, 2004, re1. [Google Scholar] [CrossRef]
  7. Berridge, M.J. Inositol trisphosphate and calcium signalling mechanisms. Biochim. Biophys. Acta Mol. Cell Res. 2009, 1793, 933–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Mikoshiba, K. Role of IP3 receptor signaling in cell functions and diseases. Adv. Biol. Regul. 2015, 57, 217–227. [Google Scholar] [CrossRef] [Green Version]
  9. Berridge, M.J. The inositol trisphosphate/calcium signaling pathway in health and disease. Physiol. Rev. 2016, 96, 1261–1296. [Google Scholar] [CrossRef] [Green Version]
  10. Bezprozvanny, I. Role of inositol 1,4,5-trishosphate receptors in pathogenesis of Huntington’s disease and spinocerebellar ataxias. Neurochem. Res. 2011, 36, 1186–1197. [Google Scholar] [CrossRef] [Green Version]
  11. Landstrom, A.P.; Dobrev, D.; Wehrens, X.H.T. Calcium Signaling and Cardiac Arrhythmias. Circ. Res. 2017, 120, 1969–1993. [Google Scholar] [CrossRef] [PubMed]
  12. Smani, T.; Gallardo-Castillo, I.; Ávila-Médina, J.; Jimenez-Navarro, M.F.; Ordoñez, A.; Hmadcha, A. Impact of Diabetes on Cardiac and Vascular Disease: Role of Calcium Signaling. Curr. Med. Chem. 2017, 26, 4166–4177. [Google Scholar] [CrossRef] [PubMed]
  13. Izquierdo-Torres, E.; Hernández-Oliveras, A.; Fuentes-García, G.; Zarain-Herzberg, Á. Calcium signaling and epigenetics: A key point to understand carcinogenesis. Cell Calcium 2020, 91, 102285. [Google Scholar] [CrossRef]
  14. Bezprozvanny, I. Calcium signaling and neurodegenerative diseases. Trends Mol. Med. 2009, 15, 89–100. [Google Scholar] [CrossRef] [Green Version]
  15. Zhang, X.; Connelly, J.; Levitan, E.S.; Sun, D.; Wang, J.Q. Calcium/Calmodulin–Dependent Protein Kinase II in Cerebrovascular Diseases. Transl. Stroke Res. 2021, 12, 513–529. [Google Scholar] [CrossRef] [PubMed]
  16. Thakore, P.; Earley, S. Transient receptor potential channels and endothelial cell calcium signaling. Compr. Physiol. 2019, 9, 1249–1277. [Google Scholar] [CrossRef] [PubMed]
  17. Hayashi, T.; Rizzuto, R.; Hajnoczky, G.; Su, T.P. MAM: More than just a housekeeper. Trends Cell Biol. 2009, 19, 81–88. [Google Scholar] [CrossRef] [Green Version]
  18. Perrone, M.; Caroccia, N.; Genovese, I.; Missiroli, S.; Modesti, L.; Pedriali, G.; Vezzani, B.; Vitto, V.A.M.; Antenori, M.; Lebiedzinska-Arciszewska, M.; et al. The role of mitochondria-associated membranes in cellular homeostasis and diseases. Int. Rev. Cell Mol. Biol. 2020, 350, 119–196. [Google Scholar] [CrossRef]
  19. Copeland, D.E.; Dalton, A.J. An association between mitochondria and the endoplasmic reticulum in cells of the pseudobranch gland of a teleost. J. Biophys. Biochem. Cytol. 1959, 5, 393–396. [Google Scholar] [CrossRef]
  20. Herrera-Cruz, M.S.; Simmen, T. Of yeast, mice and men: MAMs come in two flavors. Biol. Direct 2017, 12, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Rizzuto, R.; Pinton, P.; Carrington, W.; Fay, F.S.; Fogarty, K.E.; Lifshitz, L.M.; Tuft, R.A.; Pozzan, T. Close contacts with the endoplasmic reticulum as determinants of mitochondrial Ca2+ responses. Science 1998, 280, 1763–1766. [Google Scholar] [CrossRef]
  22. Zhang, A.; Williamson, C.D.; Wong, D.S.; Bullough, M.D.; Brown, K.J.; Hathout, Y.; Colberg-Poley, A.M. Quantitative proteomic analyses of human cytomegalovirus-induced restructuring of endoplasmic reticulum-mitochondrial contacts at late times of infection. Mol. Cell. Proteomics 2011, 10, M111.009936. [Google Scholar] [CrossRef] [Green Version]
  23. Poston, C.N.; Krishnan, S.C.; Bazemore-Walker, C.R. In-depth proteomic analysis of mammalian mitochondria-associated membranes (MAM). J. Proteomics 2013, 79, 219–230. [Google Scholar] [CrossRef]
  24. Cieri, D.; Vicario, M.; Giacomello, M.; Vallese, F.; Filadi, R.; Wagner, T.; Pozzan, T.; Pizzo, P.; Scorrano, L.; Brini, M.; et al. SPLICS: A split green fluorescent protein-based contact site sensor for narrow and wide heterotypic organelle juxtaposition. Cell Death Differ. 2018, 25, 1131–1145. [Google Scholar] [CrossRef] [Green Version]
  25. Tubbs, E.; Rieusset, J. Study of endoplasmic reticulum and mitochondria interactions by in situ proximity ligation assay in fixed cells. J. Vis. Exp. 2016, 2016, e54899. [Google Scholar] [CrossRef] [PubMed]
  26. Wieckowski, M.R.M.R.; Giorgi, C.; Lebiedzinska, M.; Duszynski, J.; Pinton, P. Isolation of mitochondria-associated membranes and mitochondria from animal tissues and cells. Nat. Protoc. 2009, 4, 1582–1590. [Google Scholar] [CrossRef]
  27. Cho, K.F.; Branon, T.C.; Rajeev, S.; Svinkina, T.; Udeshi, N.D.; Thoudam, T.; Kwak, C.; Rhee, H.W.; Lee, I.K.; Carr, S.A.; et al. Split-TurboID enables contact-dependent proximity labeling in cells. Proc. Natl. Acad. Sci. USA 2020, 117, 12143–12154. [Google Scholar] [CrossRef] [PubMed]
  28. Stone, S.J.; Vance, J.E. Phosphatidylserine synthase-1 and -2 are localized to mitochondria-associated membranes. J. Biol. Chem. 2000, 275, 34534–34540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Voelker, D.R. Interorganelle transport of aminoglycerophospholipids. Biochim. Biophys. Acta 2000, 1486, 97–107. [Google Scholar] [CrossRef]
  30. Ingerman, E.; Perkins, E.M.; Marino, M.; Mears, J.A.; McCaffery, J.M.; Hinshaw, J.E.; Nunnari, J. Dnm1 forms spirals that are structurally tailored to fit mitochondria. J. Cell Biol. 2005, 170, 1021–1027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Smirnova, E.; Griparic, L.; Shurland, D.L.; van der Bliek, A.M. Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol. Biol. Cell 2001, 12, 2245–2256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Otera, H.; Wang, C.; Cleland, M.M.; Setoguchi, K.; Yokota, S.; Youle, R.J.; Mihara, K. Mff is an essential factor for mitochondrial recruitment of Drp1 during mitochondrial fission in mammalian cells. J. Cell Biol. 2010, 191, 1141–1158. [Google Scholar] [CrossRef] [Green Version]
  33. De Brito, O.M.; Scorrano, L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 2008, 456, 605–610. [Google Scholar] [CrossRef]
  34. Pilling, A.D.; Horiuchi, D.; Lively, C.M.; Saxton, W.M. Kinesin-1 and Dynein are the primary motors for fast transport of mitochondria in Drosophila motor axons. Mol. Biol. Cell 2006, 17, 2057–2068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Glater, E.E.; Megeath, L.J.; Stowers, R.S.; Schwarz, T.L. Axonal transport of mitochondria requires milton to recruit kinesin heavy chain and is light chain independent. J. Cell Biol. 2006, 173, 545–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Szabadkai, G.; Bianchi, K.; Várnai, P.; De Stefani, D.; Wieckowski, M.R.; Cavagna, D.; Nagy, A.I.; Balla, T.; Rizzuto, R. Chaperone-mediated coupling of endoplasmic reticulum and mitochondrial Ca2+ channels. J. Cell Biol. 2006, 175, 901–911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Rowland, A.A.; Voeltz, G.K. Endoplasmic reticulum-mitochondria contacts: Function of the junction. Nat. Rev. Mol. Cell Biol. 2012, 13, 607–615. [Google Scholar] [CrossRef] [Green Version]
  38. Rutter, G.A. Moving Ca2+ from the endoplasmic reticulum to mitochondria: Is spatial intimacy enough? Biochem. Soc. Trans. 2006, 34, 351–355. [Google Scholar] [CrossRef] [Green Version]
  39. Rizzuto, R.; Brini, M.; Murgia, M.; Pozzan, T. Microdomains with high Ca2+ close to IP3-sensitive channels that are sensed by neighboring mitochondria. Science 1993, 262, 744–747. [Google Scholar] [CrossRef]
  40. Berridge, M.J. The endoplasmic reticulum: A multifunctional signaling organelle. Cell Calcium 2002, 32, 235–249. [Google Scholar] [CrossRef]
  41. Su, T.P.; Hayashi, T.; Maurice, T.; Buch, S.; Ruoho, A.E. The sigma-1 receptor chaperone as an inter-organelle signaling modulator. Trends Pharmacol. Sci. 2010, 31, 557–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hayashi, T.; Su, T.P. Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca2+ signaling and cell survival. Cell 2007, 131, 596–610. [Google Scholar] [CrossRef] [Green Version]
  43. Marchi, S.; Patergnani, S.; Pinton, P. The endoplasmic reticulum-mitochondria connection: One touch, multiple functions. Biochim. Biophys. Acta 2014, 1837, 461–469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Csordás, G.; Renken, C.; Várnai, P.; Walter, L.; Weaver, D.; Buttle, K.F.; Balla, T.; Mannella, C.A.; Hajnóczky, G. Structural and functional features and significance of the physical linkage between ER and mitochondria. J. Cell Biol. 2006, 174, 915–921. [Google Scholar] [CrossRef] [Green Version]
  45. Drago, I.; Pizzo, P.; Pozzan, T. After half a century mitochondrial calcium in- and efflux machineries reveal themselves. EMBO J. 2011, 30, 4119–4125. [Google Scholar] [CrossRef] [Green Version]
  46. Boyman, L.; Karbowski, M.; Lederer, W.J. Regulation of Mitochondrial ATP Production: Ca2+ Signaling and Quality Control. Trends Mol. Med. 2020, 26, 21–39. [Google Scholar] [CrossRef] [PubMed]
  47. Moreno-Loshuertos, R.; Fern Andez-Silva, P. Tissue specificity of energy metabolism in mitochondria. Clin. Bioenerg. 2021, 3–60. [Google Scholar] [CrossRef]
  48. Harris, J.J.; Jolivet, R.; Attwell, D. Synaptic energy use and supply. Neuron 2012, 75, 762–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Szabadkai, G.; Duchen, M.R. Mitochondria: The hub of cellular Ca2+ signaling. Physiology 2008, 23, 84–94. [Google Scholar] [CrossRef] [Green Version]
  50. Jouaville, L.S.; Pinton, P.; Bastianutto, C.; Rutter, G.A.; Rizzuto, R. Regulation of mitochondrial ATP synthesis by calcium: Evidence for a long-term metabolic priming. Proc. Natl. Acad. Sci. USA 1999, 96, 13807–13812. [Google Scholar] [CrossRef] [Green Version]
  51. Denton, R.M.; Richards, D.A.; Chin, J.G. Calcium ions and the regulation of NAD+-linked isocitrate dehydrogenase from the mitochondria of rat heart and other tissues. Biochem. J. 1978, 176, 899–906. [Google Scholar] [CrossRef]
  52. McCormack, J.G.; Denton, R.M. The effects of calcium ions and adenine nucleotides on the activity of pig heart 2-oxoglutarate dehydrogenase complex. Biochem. J. 1979, 180, 533–544. [Google Scholar] [CrossRef] [PubMed]
  53. Cárdenas, C.; Miller, R.A.; Smith, I.; Bui, T.; Molgó, J.; Müller, M.; Vais, H.; Cheung, K.-H.; Yang, J.; Parker, I.; et al. Essential Regulation of Cell Bioenergetics by Constitutive InsP3 Receptor Ca2+ Transfer to Mitochondria. Cell 2010, 142, 270–283. [Google Scholar] [CrossRef] [Green Version]
  54. Bravo-Sagua, R.; Parra, V.; López-Crisosto, C.; Díaz, P.; Quest, A.F.G.; Lavandero, S. Calcium Transport and Signaling in Mitochondria. Compr. Physiol. 2017, 7, 623–634. [Google Scholar] [CrossRef]
  55. Tilokani, L.; Nagashima, S.; Paupe, V.; Prudent, J. Mitochondrial dynamics: Overview of molecular mechanisms. Essays Biochem. 2018, 62, 341–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Frieden, M.; James, D.; Castelbou, C.; Danckaert, A.; Martinou, J.C.; Demaurex, N. Ca2+ homeostasis during mitochondrial fragmentation and perinuclear clustering induced by hFis1. J. Biol. Chem. 2004, 279, 22704–22714. [Google Scholar] [CrossRef] [Green Version]
  57. Csordás, G.; Várnai, P.; Golenár, T.; Roy, S.; Purkins, G.; Schneider, T.G.; Balla, T.; Hajnóczky, G. Imaging interorganelle contacts and local calcium dynamics at the ER-mitochondrial interface. Mol. Cell 2010, 39, 121–132. [Google Scholar] [CrossRef] [PubMed]
  58. Eisner, V.; Lenaers, G.; Hajnóczky, G. Mitochondrial fusion is frequent in skeletal muscle and supports excitation-contraction coupling. J. Cell Biol. 2014, 205, 179–195. [Google Scholar] [CrossRef] [Green Version]
  59. Wozniak, A.L.; Wang, X.; Stieren, E.S.; Scarbrough, S.G.; Elferink, C.J.; Boehning, D. Requirement of biphasic calcium release from the endoplasmic reticulum for Fas-mediated apoptosis. J. Cell Biol. 2006, 175, 709–714. [Google Scholar] [CrossRef]
  60. Marchi, S.; Patergnani, S.; Missiroli, S.; Morciano, G.; Rimessi, A.; Wieckowski, M.R.; Giorgi, C.; Pinton, P. Mitochondrial and endoplasmic reticulum calcium homeostasis and cell death. Cell Calcium 2018, 69, 62–72. [Google Scholar] [CrossRef]
  61. Baumgartner, H.K.; Gerasimenko, J.V.; Thorne, C.; Ferdek, P.; Pozzan, T.; Tepikin, A.V.; Petersen, O.H.; Sutton, R.; Watson, A.J.M.; Gerasimenko, O.V. Calcium elevation in mitochondria is the main Ca2+ requirement for mitochondrial permeability transition pore (mPTP) opening. J. Biol. Chem. 2009, 284, 20796–20803. [Google Scholar] [CrossRef] [Green Version]
  62. Lin, M.T.; Beal, M.F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 2006, 443, 787–795. [Google Scholar] [CrossRef]
  63. Nickel, A.; Kohlhaas, M.; Maack, C. Mitochondrial reactive oxygen species production and elimination. J. Mol. Cell. Cardiol. 2014, 73, 26–33. [Google Scholar] [CrossRef] [PubMed]
  64. Aon, M.A.; Cortassa, S.; O’Rourke, B. Redox-optimized ROS balance: A unifying hypothesis. Biochim. Biophys. Acta Bioenerg. 2010, 1797, 865–877. [Google Scholar] [CrossRef] [Green Version]
  65. Booth, D.M.; Enyedi, B.; Geiszt, M.; Várnai, P.; Hajnóczky, G. Redox Nanodomains Are Induced by and Control Calcium Signaling at the ER-Mitochondrial Interface. Mol. Cell 2016, 63, 240–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hopper, R.K.; Carroll, S.; Aponte, A.M.; Johnson, D.T.; French, S.; Shen, R.-F.; Witzmann, F.A.; Harris, R.A.; Balaban, R.S. Mitochondrial matrix phosphoproteome: Effect of extra mitochondrial calcium. Biochemistry 2006, 45, 2524–2536. [Google Scholar] [CrossRef] [Green Version]
  67. Gerasimenko, J.V.; Gerasimenko, O.V.; Palejwala, A.; Tepikin, A.V.; Petersen, O.H.; Watson, A.J.M. Menadione-induced apoptosis: Roles of cytosolic Ca2+ elevations and the mitochondrial permeability transition pore. J. Cell Sci. 2002, 115, 485–497. [Google Scholar] [CrossRef]
  68. Zoccarato, F.; Cavallini, L.; Alexandre, A. Respiration-dependent Removal of Exogenous H2O2 in Brain Mitochondria. Inhibition by Ca2+. J. Biol. Chem. 2004, 279, 4166–4174. [Google Scholar] [CrossRef] [Green Version]
  69. Stepanova, A.; Magrané, J. Mitochondrial dysfunction in neurons in Friedreich’s ataxia. Mol. Cell. Neurosci. 2020, 102, 103419. [Google Scholar] [CrossRef] [PubMed]
  70. Obrador, E.; Salvador-Palmer, R.; López-Blanch, R.; Jihad-Jebbar, A.; Vallés, S.L.; Estrela, J.M. The link between oxidative stress, redox status, bioenergetics and mitochondria in the pathophysiology of als. Int. J. Mol. Sci. 2021, 22, 6352. [Google Scholar] [CrossRef]
  71. Bernard-Marissal, N.; van Hameren, G.; Juneja, M.; Pellegrino, C.; Louhivuori, L.; Bartesaghi, L.; Rochat, C.; El Mansour, O.; Médard, J.J.; Croisier, M.; et al. Altered interplay between endoplasmic reticulum and mitochondria in Charcot-Marie-Tooth type 2A neuropathy. Proc. Natl. Acad. Sci. USA 2019, 116, 2328–2337. [Google Scholar] [CrossRef] [Green Version]
  72. Park, S.H.; Zhu, P.P.; Parker, R.L.; Blackstone, C. Hereditary spastic paraplegia proteins REEP1, spastin, and atlastin-1 coordinate microtubule interactions with the tubular ER network. J. Clin. Investig. 2010, 120, 1097–1110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Villegas, R.; Martinez, N.W.; Lillo, J.; Pihan, P.; Hernandez, D.; Twiss, J.L.; Court, F.A. Calcium release from intra-axonal endoplasmic reticulum leads to axon degeneration through mitochondrial dysfunction. J. Neurosci. 2014, 34, 7179–7189. [Google Scholar] [CrossRef]
  74. Ellisman, M.H.; Porter, K.R. Microtrabecular structure of the axoplasmic matrix: Visualization of cross-linking structures and their distribution. J. Cell Biol. 1980, 87, 464. [Google Scholar] [CrossRef]
  75. Mironov, S.L.; Symonchuk, N. ER vesicles and mitochondria move and communicate at synapses. J. Cell Sci. 2006, 119, 4926–4934. [Google Scholar] [CrossRef] [Green Version]
  76. Manfredi, G.; Kawamata, H. Mitochondria and endoplasmic reticulum crosstalk in amyotrophic lateral sclerosis. Neurobiol. Dis. 2016, 90, 35–42. [Google Scholar] [CrossRef] [Green Version]
  77. Rodríguez-Arribas, M.; Yakhine-Diop, S.M.S.; Pedro, J.M.B.S.; Gómez-Suaga, P.; Gómez-Sánchez, R.; Martínez-Chacón, G.; Fuentes, J.M.; González-Polo, R.A.; Niso-Santano, M. Mitochondria-Associated Membranes (MAMs): Overview and Its Role in Parkinson’s Disease. Mol. Neurobiol. 2017, 54, 6287–6303. [Google Scholar] [CrossRef] [Green Version]
  78. Eysert, F.; Kinoshita, P.F.; Mary, A.; Vaillant-Beuchot, L.; Checler, F.; Chami, M. Molecular dysfunctions of mitochondria-associated membranes (Mams) in alzheimer’s disease. Int. J. Mol. Sci. 2020, 21, 9521. [Google Scholar] [CrossRef]
  79. Veeresh, P.; Kaur, H.; Sarmah, D.; Mounica, L.; Verma, G.; Kotian, V.; Kesharwani, R.; Kalia, K.; Borah, A.; Wang, X.; et al. Endoplasmic reticulum–mitochondria crosstalk: From junction to function across neurological disorders. Ann. N. Y. Acad. Sci. 2019, 1457, 41–60. [Google Scholar] [CrossRef] [PubMed]
  80. Guardia-Laguarta, C.; Area-Gomez, E.; Rüb, C.; Liu, Y.; Magrané, J.; Becker, D.; Voos, W.; Schon, E.A.; Przedborski, S. α-synuclein is localized to mitochondria-associated ER membranes. J. Neurosci. 2014, 34, 249–259. [Google Scholar] [CrossRef]
  81. Calì, T.; Ottolini, D.; Negro, A.; Brini, M. α-Synuclein controls mitochondrial calcium homeostasis by enhancing endoplasmic reticulum-mitochondria interactions. J. Biol. Chem. 2012, 287, 17914–17929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Cherubini, M.; Lopez-Molina, L.; Gines, S. Mitochondrial fission in Huntington’s disease mouse striatum disrupts ER-mitochondria contacts leading to disturbances in Ca2+ efflux and Reactive Oxygen Species (ROS) homeostasis. Neurobiol. Dis. 2020, 136, 104741. [Google Scholar] [CrossRef] [PubMed]
  83. Del Prete, D.; Suski, J.M.; Oulès, B.; Debayle, D.; Gay, A.S.; Lacas-Gervais, S.; Bussiere, R.; Bauer, C.; Pinton, P.; Paterlini-Bréchot, P.; et al. Localization and Processing of the Amyloid-β Protein Precursor in Mitochondria-Associated Membranes. J. Alzheimer’s Dis. 2017, 55, 1549–1570. [Google Scholar] [CrossRef] [Green Version]
  84. Zhou, J.; Yi, J.; Fu, R.; Liu, E.; Siddique, T.; Rios, E.; Deng, H.X. Hyperactive intracellular calcium signaling associated with localized mitochondrial defects in skeletal muscle of an animal model of amyotrophic lateral sclerosis. J. Biol. Chem. 2010, 285, 705–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Jaiswal, M.K.; Keller, B.U. Cu/Zn superoxide dismutase typical for familial amyotrophic lateral sclerosis increases the vulnerability of mitochondria and perturbs Ca2+ homeostasis in SOD1G93A mice. Mol. Pharmacol. 2009, 75, 478–489. [Google Scholar] [CrossRef] [Green Version]
  86. Wong, A.Y.C.; Hristova, E.; Ahlskog, N.; Tasse, L.A.; Ngsee, J.K.; Chudalayandi, P.; Bergeron, R. Aberrant subcellular dynamics of sigma-1 receptor mutants underlying neuromuscular diseases. Mol. Pharmacol. 2016, 90, 238–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Watanabe, S.; Ilieva, H.; Tamada, H.; Nomura, H.; Komine, O.; Endo, F.; Jin, S.; Mancias, P.; Kiyama, H.; Yamanaka, K. Mitochondria-associated membrane collapse is a common pathomechanism in SIGMAR 1—And SOD 1 -linked ALS. EMBO Mol. Med. 2016, 8, 1421–1437. [Google Scholar] [CrossRef]
  88. Matilla-Dueñas, A.; Ashizawa, T.; Brice, A.; Magri, S.; McFarland, K.N.; Pandolfo, M.; Pulst, S.M.; Riess, O.; Rubinsztein, D.C.; Schmidt, J.; et al. Consensus Paper: Pathological Mechanisms Underlying Neurodegeneration in Spinocerebellar Ataxias. Cerebellum 2014, 13, 269. [Google Scholar] [CrossRef] [Green Version]
  89. Chen, X.; Tang, T.S.; Tu, H.; Nelson, O.; Pook, M.; Hammer, R.; Nukina, N.; Bezprozvanny, I. Deranged calcium signaling and neurodegeneration in spinocerebellar ataxia type 3. J. Neurosci. 2008, 28, 12713–12724. [Google Scholar] [CrossRef] [Green Version]
  90. Rodríguez, L.R.; Calap-Quintana, P.; Lapeña-Luzón, T.; Pallardó, F.V.; Schneuwly, S.; Navarro, J.A.; Gonzalez-Cabo, P. Oxidative stress modulates rearrangement of endoplasmic reticulum-mitochondria contacts and calcium dysregulation in a Friedreich’s ataxia model. Redox Biol. 2020, 37, 101762. [Google Scholar] [CrossRef]
  91. Abeti, R.; Brown, A.F.; Maiolino, M.; Patel, S.; Giunti, P. Calcium Deregulation: Novel Insights to Understand Friedreich’s Ataxia Pathophysiology. Front. Cell. Neurosci. 2018, 12, 264. [Google Scholar] [CrossRef] [Green Version]
  92. Bolinches-Amorós, A.; Mollá, B.; Pla-Martín, D.; Palau, F.; González-Cabo, P. Mitochondrial dysfunction induced by frataxin deficiency is associated with cellular senescence and abnormal calcium metabolism. Front. Cell. Neurosci. 2014, 8, 124. [Google Scholar] [CrossRef] [Green Version]
  93. Zhou, Y.; Carmona, S.; Muhammad, A.K.M.G.; Bell, S.; Landeros, J.; Vazquez, M.; Ho, R.; Franco, A.; Lu, B.; Dorn, G.W.; et al. Erratum: Restoring mitofusin balance prevents axonal degeneration in a Charcot-marie-tooth type 2A model. J. Clin. Investig. 2021, 131, e147307. [Google Scholar] [CrossRef] [PubMed]
  94. Civera-Tregón, A.; Domínguez, L.; Martínez-Valero, P.; Serrano, C.; Vallmitjana, A.; Benítez, R.; Hoenicka, J.; Satrústegui, J.; Palau, F. Mitochondria and calcium defects correlate with axonal dysfunction in GDAP1-related Charcot-Marie-Tooth mouse model. Neurobiol. Dis. 2021, 152, 105300. [Google Scholar] [CrossRef]
  95. Pla-Martín, D.; Rueda, C.B.; Estela, A.; Sánchez-Piris, M.; González-Sánchez, P.; Traba, J.; de la Fuente, S.; Scorrano, L.; Renau-Piqueras, J.; Alvarez, J.; et al. Silencing of the Charcot-Marie-Tooth disease-associated gene GDAP1 induces abnormal mitochondrial distribution and affects Ca2+ homeostasis by reducing store-operated Ca2+ entry. Neurobiol. Dis. 2013, 55, 140–151. [Google Scholar] [CrossRef]
  96. Wagner, M.; Osborn, D.P.S.; Gehweiler, I.; Nagel, M.; Ulmer, U.; Bakhtiari, S.; Amouri, R.; Boostani, R.; Hentati, F.; Hockley, M.M.; et al. Bi-allelic variants in RNF170 are associated with hereditary spastic paraplegia. Nat. Commun. 2019, 10, 1–13. [Google Scholar] [CrossRef]
  97. Zhu, P.P.; Denton, K.R.; Pierson, T.M.; Li, X.J.; Blackstone, C. Pharmacologic rescue of axon growth defects in a human iPSC model of hereditary spastic paraplegia SPG3A. Hum. Mol. Genet. 2014, 23, 5638–5648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Lim, Y.; Cho, I.T.; Schoel, L.J.; Cho, G.; Golden, J.A. Hereditary spastic paraplegia-linked REEP1 modulates endoplasmic reticulum/mitochondria contacts. Ann. Neurol. 2015, 78, 679–696. [Google Scholar] [CrossRef]
  99. SMART-Servier Medical ART. Available online: https://smart.servier.com/ (accessed on 13 December 2021).
  100. Penke, B.; Fulop, L.; Szucs, M.; Frecska, E. The Role of Sigma-1 Receptor, an Intracellular Chaperone in Neurodegenerative Diseases. Curr. Neuropharmacol. 2018, 16, 97–116. [Google Scholar] [CrossRef] [PubMed]
  101. Mei, J.; Pasternak, G.W. Molecular cloning and pharmacological characterization of the rat sigma1 receptor. Biochem. Pharmacol. 2001, 62, 349–355. [Google Scholar] [CrossRef]
  102. Laurini, E.; Marson, D.; Fermeglia, M.; Pricl, S. 3D Homology Model of Sigma1 Receptor. Handb. Exp. Pharmacol. 2017, 244, 27–50. [Google Scholar] [CrossRef]
  103. Wu, N.-H.; Ye, Y.; Wan, B.-B.; Yu, Y.-D.; Liu, C.; Chen, Q.-J. Emerging Benefits: Pathophysiological Functions and Target Drugs of the Sigma-1 Receptor in Neurodegenerative Diseases. Mol. Neurobiol. 2021, 58, 5649–5666. [Google Scholar] [CrossRef]
  104. Maurice, T.; Grégoire, C.; Espallergues, J. Neuro(active)steroids actions at the neuromodulatory sigma1 (sigma1) receptor: Biochemical and physiological evidences, consequences in neuroprotection. Pharmacol. Biochem. Behav. 2006, 84, 581–597. [Google Scholar] [CrossRef] [PubMed]
  105. Ryskamp, D.A.; Korban, S.; Zhemkov, V.; Kraskovskaya, N.; Bezprozvanny, I. Neuronal Sigma-1 Receptors: Signaling Functions and Protective Roles in Neurodegenerative Diseases. Front. Neurosci. 2019, 13, 862. [Google Scholar] [CrossRef]
  106. Koshenov, Z.; Oflaz, F.E.; Hirtl, M.; Pilic, J.; Bachkoenig, O.A.; Gottschalk, B.; Madreiter-Sokolowski, C.T.; Rost, R.; Malli, R.; Graier, W.F. Sigma-1 Receptor Promotes Mitochondrial Bioenergetics by Orchestrating ER Ca2+ Leak during Early ER Stress. Metabolites 2021, 11, 442. [Google Scholar] [CrossRef] [PubMed]
  107. Hetz, C.; Saxena, S. ER stress and the unfolded protein response in neurodegeneration. Nat. Rev. Neurol. 2017, 13, 477–491. [Google Scholar] [CrossRef]
  108. Brimson, J.M.; Prasanth, M.I.; Malar, D.S.; Brimson, S.; Thitilertdecha, P.; Tencomnao, T. Drugs that offer the potential to reduce hospitalization and mortality from SARS-CoV-2 infection: The possible role of the sigma-1 receptor and autophagy. Expert Opin. Ther. Targets 2021, 25, 435–449. [Google Scholar] [CrossRef]
  109. Gromek, K.A.; Suchy, F.P.; Meddaugh, H.R.; Wrobel, R.L.; LaPointe, L.M.; Chu, U.B.; Primm, J.G.; Ruoho, A.E.; Senes, A.; Fox, B.G. The oligomeric states of the purified sigma-1 receptor are stabilized by ligands. J. Biol. Chem. 2014, 289, 20333–20344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Mishra, A.K.; Mavlyutov, T.; Singh, D.R.; Biener, G.; Yang, J.; Oliver, J.A.; Ruoho, A.; Raicu, V. The sigma-1 receptors are present in monomeric and oligomeric forms in living cells in the presence and absence of ligands. Biochem. J. 2015, 466, 263–271. [Google Scholar] [CrossRef] [Green Version]
  111. Ye, N.; Qin, W.; Tian, S.; Xu, Q.; Wold, E.A.; Zhou, J.; Zhen, X.-C. Small Molecules Selectively Targeting Sigma-1 Receptor for the Treatment of Neurological Diseases. J. Med. Chem. 2020, 63, 15187–15217. [Google Scholar] [CrossRef] [PubMed]
  112. Meunier, J.; Hayashi, T. Sigma-1 receptors regulate Bcl-2 expression by reactive oxygen species-dependent transcriptional regulation of nuclear factor kappaB. J. Pharmacol. Exp. Ther. 2010, 332, 388–397. [Google Scholar] [CrossRef] [Green Version]
  113. Pal, A.; Fontanilla, D.; Gopalakrishnan, A.; Chae, Y.-K.; Markley, J.L.; Ruoho, A.E. The sigma-1 receptor protects against cellular oxidative stress and activates antioxidant response elements. Eur. J. Pharmacol. 2012, 682, 12–20. [Google Scholar] [CrossRef] [Green Version]
  114. Couly, S.; Khalil, B.; Viguier, V.; Roussel, J.; Maurice, T.; Liévens, J.C. Sigma-1 receptor is a key genetic modulator in amyotrophic lateral sclerosis. Hum. Mol. Genet. 2020, 29, 529–540. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, J.; Shanmugam, A.; Markand, S.; Zorrilla, E.; Ganapathy, V.; Smith, S.B. Sigma 1 receptor regulates the oxidative stress response in primary retinal Müller glial cells via NRF2 signaling and system xc(−), the Na(+)-independent glutamate-cystine exchanger. Free Radic. Biol. Med. 2015, 86, 25–36. [Google Scholar] [CrossRef] [Green Version]
  116. Mavlyutov, T.A.; Nickells, R.W.; Guo, L.-W. Accelerated retinal ganglion cell death in mice deficient in the Sigma-1 receptor. Mol. Vis. 2011, 17, 1034–1043. [Google Scholar]
  117. Miki, Y.; Tanji, K.; Mori, F.; Wakabayashi, K. Sigma-1 receptor is involved in degradation of intranuclear inclusions in a cellular model of Huntington’s disease. Neurobiol. Dis. 2015, 74, 25–31. [Google Scholar] [CrossRef] [PubMed]
  118. Maurice, T.; Strehaiano, M.; Duhr, F.; Chevallier, N. Amyloid toxicity is enhanced after pharmacological or genetic invalidation of the σ1 receptor. Behav. Brain Res. 2018, 339, 1–10. [Google Scholar] [CrossRef]
  119. Antonini, V.; Prezzavento, O.; Coradazzi, M.; Marrazzo, A.; Ronsisvalle, S.; Arena, E.; Leanza, G. Anti-amnesic properties of (±)-PPCC, a novel sigma receptor ligand, on cognitive dysfunction induced by selective cholinergic lesion in rats. J. Neurochem. 2009, 109, 744–754. [Google Scholar] [CrossRef] [PubMed]
  120. Ruscher, K.; Shamloo, M.; Rickhag, M.; Ladunga, I.; Soriano, L.; Gisselsson, L.; Toresson, H.; Ruslim-Litrus, L.; Oksenberg, D.; Urfer, R.; et al. The sigma-1 receptor enhances brain plasticity and functional recovery after experimental stroke. Brain 2011, 134, 732–746. [Google Scholar] [CrossRef] [Green Version]
  121. Bernard-Marissal, N.; Médard, J.-J.; Azzedine, H.; Chrast, R. Dysfunction in endoplasmic reticulum-mitochondria crosstalk underlies SIGMAR1 loss of function mediated motor neuron degeneration. Brain 2015, 138, 875–890. [Google Scholar] [CrossRef] [Green Version]
  122. Hayashi, T.; Su, T.-P. Sigma-1 receptor ligands: Potential in the treatment of neuropsychiatric disorders. CNS Drugs 2004, 18, 269–284. [Google Scholar] [CrossRef]
  123. Nguyen, L.; Lucke-Wold, B.P.; Mookerjee, S.; Kaushal, N.; Matsumoto, R.R. Sigma-1 Receptors and Neurodegenerative Diseases: Towards a Hypothesis of Sigma-1 Receptors as Amplifiers of Neurodegeneration and Neuroprotection. Adv. Exp. Med. Biol. 2017, 964, 133–152. [Google Scholar] [CrossRef] [Green Version]
  124. Sahlholm, K.; Århem, P.; Fuxe, K.; Marcellino, D. The dopamine stabilizers ACR16 and (−)-OSU6162 display nanomolar affinities at the σ-1 receptor. Mol. Psychiatry 2013, 18, 12–14. [Google Scholar] [CrossRef]
  125. Sahlholm, K.; Sijbesma, J.W.A.; Maas, B.; Kwizera, C.; Marcellino, D.; Ramakrishnan, N.K.; Dierckx, R.A.J.O.; Elsinga, P.H.; van Waarde, A. Pridopidine selectively occupies sigma-1 rather than dopamine D2 receptors at behaviorally active doses. Psychopharmacology 2015, 232, 3443–3453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Eddings, C.R.; Arbez, N.; Akimov, S.; Geva, M.; Hayden, M.R.; Ross, C.A. Pridopidine protects neurons from mutant-huntingtin toxicity via the sigma-1 receptor. Neurobiol. Dis. 2019, 129, 118–129. [Google Scholar] [CrossRef] [PubMed]
  127. De Yebenes, J.G.; Landwehrmeyer, B.; Squitieri, F.; Reilmann, R.; Rosser, A.; Barker, R.A.; Saft, C.; Magnet, M.K.; Sword, A.; Rembratt, A.; et al. Pridopidine for the treatment of motor function in patients with Huntington’s disease (MermaiHD): A phase 3, randomised, double-blind, placebo-controlled trial. Lancet. Neurol. 2011, 10, 1049–1057. [Google Scholar] [CrossRef]
  128. PRidopidine’s Outcome on Function in Huntington Disease, PROOF- HD-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04556656?term=Pridopidine&draw=2&rank=1 (accessed on 5 October 2021).
  129. Al-Saif, A.; Al-Mohanna, F.; Bohlega, S. A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann. Neurol. 2011, 70, 913–919. [Google Scholar] [CrossRef] [PubMed]
  130. Ionescu, A.; Gradus, T.; Altman, T.; Maimon, R.; Saraf Avraham, N.; Geva, M.; Hayden, M.; Perlson, E. Targeting the Sigma-1 Receptor via Pridopidine Ameliorates Central Features of ALS Pathology in a SOD1G93A Model. Cell Death Dis. 2019, 10, 210. [Google Scholar] [CrossRef]
  131. HEALEY ALS Platform Trial-Regimen D Pridopidine-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04615923?term=Pridopidine&cond=Amyotrophic+Lateral+Sclerosis&draw=2&rank=1 (accessed on 6 October 2021).
  132. Francardo, V.; Geva, M.; Bez, F.; Denis, Q.; Steiner, L.; Hayden, M.R.; Cenci, M.A. Pridopidine Induces Functional Neurorestoration Via the Sigma-1 Receptor in a Mouse Model of Parkinson’s Disease. Neurotherapeutics 2019, 16, 465–479. [Google Scholar] [CrossRef] [Green Version]
  133. A Study to Assess the Safety and Effectiveness of Pridopidine Compared to Placebo in the Treatment of Levodopa-Induced Dyskinesia in Patients With Parkinson’s Disease-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/study/NCT03922711?term=Pridopidine&draw=2&rank=7 (accessed on 7 October 2021).
  134. Maurice, T.; Su, T.P. The pharmacology of sigma-1 receptors. Pharmacol. Ther. 2009, 124, 195–206. [Google Scholar] [CrossRef] [Green Version]
  135. Qin, J.; Wang, P.; Li, Y.; Yao, L.; Liu, Y.; Yu, T.; Lin, J.; Fang, X.; Huang, Z. Activation of Sigma-1 Receptor by Cutamesine Attenuates Neuronal Apoptosis by Inhibiting Endoplasmic Reticulum Stress and Mitochondrial Dysfunction in a Rat Model of Asphyxia Cardiac Arrest. Shock 2019, 51, 105–113. [Google Scholar] [CrossRef]
  136. Tagashira, H.; Zhang, C.; Lu, Y.M.; Hasegawa, H.; Kanai, H.; Han, F.; Fukunaga, K. Stimulation of σ1-receptor restores abnormal mitochondrial Ca2+ mobilization and ATP production following cardiac hypertrophy. Biochim. Biophys. Acta Gen. Subj. 2013, 1830, 3082–3094. [Google Scholar] [CrossRef]
  137. Tuerxun, T.; Numakawa, T.; Adachi, N.; Kumamaru, E.; Kitazawa, H.; Kudo, M.; Kunugi, H. SA4503, a sigma-1 receptor agonist, prevents cultured cortical neurons from oxidative stress-induced cell death via suppression of MAPK pathway activation and glutamate receptor expression. Neurosci. Lett. 2010, 469, 303–308. [Google Scholar] [CrossRef]
  138. Tadić, V.; Malci, A.; Goldhammer, N.; Stubendorff, B.; Sengupta, S.; Prell, T.; Keiner, S.; Liu, J.; Guenther, M.; Frahm, C.; et al. Sigma 1 receptor activation modifies intracellular calcium exchange in the G93AhSOD1 ALS model. Neuroscience 2017, 359, 105–118. [Google Scholar] [CrossRef]
  139. Yamaguchi, K.; Shioda, N.; Yabuki, Y.; Zhang, C.; Han, F.; Fukunaga, K. SA4503, A Potent Sigma-1 Receptor Ligand, Ameliorates Synaptic Abnormalities and Cognitive Dysfunction in a Mouse Model of ATR-X Syndrome. Int. J. Mol. Sci. 2018, 19, 2811. [Google Scholar] [CrossRef] [Green Version]
  140. SA4503 8-Week Study in Major Depressive Disorder (MDD)-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT00551109?term=cutamesine&draw=2&rank=1 (accessed on 8 October 2021).
  141. Goguadze, N.; Zhuravliova, E.; Morin, D.; Mikeladze, D.; Maurice, T. Sigma-1 Receptor Agonists Induce Oxidative Stress in Mitochondria and Enhance Complex I Activity in Physiological Condition but Protect Against Pathological Oxidative Stress. Neurotox. Res. 2019, 35, 1–18. [Google Scholar] [CrossRef] [PubMed]
  142. Lahmy, V.; Meunier, J.; Malmström, S.; Naert, G.; Givalois, L.; Kim, S.H.; Villard, V.; Vamvakides, A.; Maurice, T. Blockade of Tau hyperphosphorylation and Aβ1–42 eneration by the aminotetrahydrofuran derivative ANAVEX2-73, a mixed muscarinic and σ1 receptor agonist, in a nontransgenic mouse model of Alzheimer’s disease. Neuropsychopharmacology 2013, 38, 1706–1723. [Google Scholar] [CrossRef] [PubMed]
  143. Lahmy, V.; Long, R.; Morin, D.; Villard, V.; Maurice, T. Mitochondrial protection by the mixed muscarinic/σ1 ligand ANAVEX2-73, a tetrahydrofuran derivative, in Aβ25-35 peptide-injected mice, a nontransgenic Alzheimer’s disease model. Front. Cell. Neurosci. 2014, 8, 463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Villard, V.; Espallergues, J.; Keller, E.; Vamvakides, A.; Maurice, T. Anti-amnesic and neuroprotective potentials of the mixed muscarinic receptor/sigma 1 (σ1) ligand ANAVEX2-73, a novel aminotetrahydrofuran derivative. J. Psychopharmacol. 2011, 25, 1101–1117. [Google Scholar] [CrossRef] [PubMed]
  145. Phase 2a Dose Finding, PK/PD and 12 Month Exploratory Efficacy Study of ANAVEX2-73 in Patients with Alzheimer’s Disease -Full Text Vie-ClinicalTrials.gov. Available online: https://www.clinicaltrials.gov/ct2/show/NCT02244541 (accessed on 7 October 2021).
  146. Study of ANAVEX2-73 in Patients with Rett Syndrome-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03758924?term=anavex2&draw=2&rank=6 (accessed on 7 October 2021).
  147. OLE Study for Patients with Parkinson’s Disease With Dementia Enrolled in Study ANAVEX2-73-PDD-001-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04575259?term=anavex2&draw=2&rank=1 (accessed on 7 October 2021).
  148. OLE of Phase 2b/3 Study ANAVEX2-73-AD-004-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04314934?term=anavex2&draw=2&rank=2 (accessed on 7 October 2021).
  149. ANAVEX2-73 Study in Patients with Rett Syndrome-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03941444?term=anavex2&draw=2&rank=4 (accessed on 7 October 2021).
  150. ANAVEX2-73 Study in Pediatric Patients with Rett Syndrome-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04304482?term=anavex2&draw=2&rank=3 (accessed on 7 October 2021).
  151. Bucolo, C.; Drago, F.; Lin, L.R.; Reddy, V.N. Sigma receptor ligands protect human retinal cells against oxidative stress. Neuroreport 2006, 17, 287–291. [Google Scholar] [CrossRef] [PubMed]
  152. Hyrskyluoto, A.; Pulli, I.; Törnqvist, K.; Ho, T.H.; Korhonen, L.; Lindholm, D. Sigma-1 receptor agonist PRE084 is protective against mutant huntingtin-induced cell degeneration: Involvement of calpastatin and the NF-κB pathway. Cell Death Dis. 2013, 4, e646. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, X.; Wang, B.; Zeng, H.; Cai, C.; Hu, Q.; Cai, S.; Xu, L.; Meng, X.; Zou, F. Role of the mitochondrial Ca2+ uniporter in Pb2+-induced oxidative stress in human neuroblastoma cells. Brain Res. 2014, 1575, 12–21. [Google Scholar] [CrossRef]
  154. Omi, T.; Tanimukai, H.; Kanayama, D.; Sakagami, Y.; Tagami, S.; Okochi, M.; Morihara, T.; Sato, M.; Yanagida, K.; Kitasyoji, A.; et al. Fluvoxamine alleviates ER stress via induction of Sigma-1 receptor. Cell Death Dis. 2014, 5, e1332. [Google Scholar] [CrossRef]
  155. Tagashira, H.; Bhuiyan, M.S.; Shioda, N.; Fukunaga, K. Fluvoxamine rescues mitochondrial Ca2+ transport and ATP production through σ1-receptor in hypertrophic cardiomyocytes. Life Sci. 2014, 95, 89–100. [Google Scholar] [CrossRef]
  156. Tagashira, H.; Bhuiyan, S.; Shioda, N.; Hasegawa, H.; Kanai, H.; Fukunaga, K. Σ1-Receptor Stimulation With Fluvoxamine Ameliorates Transverse Aortic Constriction-Induced Myocardial Hypertrophy and Dysfunction in Mice. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1535–H1545. [Google Scholar] [CrossRef] [Green Version]
  157. Elsaed, W.M.; Alahmadi, A.M.; Al-Ahmadi, B.T.; Taha, J.A.; Tarabishi, R.M. Gastroprotective and antioxidant effects of fluvoxamine on stress-induced peptic ulcer in rats. J. Taibah Univ. Med. Sci. 2018, 13, 422–431. [Google Scholar] [CrossRef]
  158. Abdel-Salam, O.M.E.; Youssef Morsy, S.M.; Sleem, A.A. The effect of different antidepressant drugs on oxidative stress after lipopolysaccharide administration in mice. EXCLI J. 2011, 10, 290–302. [Google Scholar] [CrossRef]
  159. Almási, N.; Török, S.; Valkusz, Z.; Tajti, M.; Csonka, Á.; Murlasits, Z.; Pósa, A.; Varga, C.; Kupai, K. Sigma-1 receptor engages an anti-inflammatory and antioxidant feedback loop mediated by peroxiredoxin in experimental colitis. Antioxidants 2020, 9, 1081. [Google Scholar] [CrossRef]
  160. Sukhatme, V.P.; Reiersen, A.M.; Vayttaden, S.J.; Sukhatme, V.V. Fluvoxamine: A Review of Its Mechanism of Action and Its Role in COVID-19. Front. Pharmacol. 2021, 12, 652688. [Google Scholar] [CrossRef]
  161. De Stefani, D.; Raffaello, A.; Teardo, E.; Szabó, I.; Rizzuto, R. A forty-kilodalton protein of the inner membrane is the mitochondrial calcium uniporter. Nature 2011, 476, 336–340. [Google Scholar] [CrossRef]
  162. Qi, H.; Li, L.; Shuai, J. Optimal microdomain crosstalk between endoplasmic reticulum and mitochondria for Ca2+ oscillations. Sci. Rep. 2015, 5, 7984. [Google Scholar] [CrossRef] [Green Version]
  163. Baughman, J.M.; Perocchi, F.; Girgis, H.S.; Plovanich, M.; Belcher-Timme, C.A.; Sancak, Y.; Bao, X.R.; Strittmatter, L.; Goldberger, O.; Bogorad, R.L.; et al. Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature 2011, 476, 341–345. [Google Scholar] [CrossRef] [Green Version]
  164. Oxenoid, K.; Dong, Y.; Cao, C.; Cui, T.; Sancak, Y.; Markhard, A.L.; Grabarek, Z.; Kong, L.; Liu, Z.; Ouyang, B.; et al. Architecture of the mitochondrial calcium uniporter. Nature 2016, 533, 269–273. [Google Scholar] [CrossRef] [Green Version]
  165. Wang, L.; Yang, X.; Li, S.; Wang, Z.; Liu, Y.; Feng, J.; Zhu, Y.; Shen, Y. Structural and mechanistic insights into MICU1 regulation of mitochondrial calcium uptake. EMBO J. 2014, 33, 594–604. [Google Scholar] [CrossRef] [PubMed]
  166. Liu, J.C.; Liu, J.; Holmström, K.M.; Menazza, S.; Parks, R.J.; Fergusson, M.M.; Yu, Z.X.; Springer, D.A.; Halsey, C.; Liu, C.; et al. MICU1 Serves as a Molecular Gatekeeper to Prevent In Vivo Mitochondrial Calcium Overload. Cell Rep. 2016, 16, 1561–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Logan, C.V.; Szabadkai, G.; Sharpe, J.A.; Parry, D.A.; Torelli, S.; Childs, A.M.; Kriek, M.; Phadke, R.; Johnson, C.A.; Roberts, N.Y.; et al. Loss-of-function mutations in MICU1 cause a brain and muscle disorder linked to primary alterations in mitochondrial calcium signaling. Nat. Genet. 2014, 46, 188–193. [Google Scholar] [CrossRef]
  168. Kirichok, Y.; Krapivinsky, G.; Clapham, D.E. The mitochondrial calcium uniporter is a highly selective ion channel. Nature 2004, 427, 360–364. [Google Scholar] [CrossRef] [PubMed]
  169. Patron, M.; Granatiero, V.; Espino, J.; Rizzuto, R.; de Stefani, D. MICU3 is a tissue-specific enhancer of mitochondrial calcium uptake. Cell Death Differ. 2019, 26, 179–195. [Google Scholar] [CrossRef] [Green Version]
  170. Sancak, Y.; Markhard, A.L.; Kitami, T.; Kovács-Bogdán, E.; Kamer, K.J.; Udeshi, N.D.; Carr, S.A.; Chaudhuri, D.; Clapham, D.E.; Li, A.A.; et al. EMRE is an essential component of the mitochondrial calcium uniporter complex. Science 2013, 342, 1379–1382. [Google Scholar] [CrossRef] [Green Version]
  171. Yamamoto, T.; Yamagoshi, R.; Harada, K.; Kawano, M.; Minami, N.; Ido, Y.; Kuwahara, K.; Fujita, A.; Ozono, M.; Watanabe, A.; et al. Analysis of the structure and function of EMRE in a yeast expression system. Biochim. Biophys. Acta Bioenerg. 2016, 1857, 831–839. [Google Scholar] [CrossRef]
  172. Mallilankaraman, K.; Cárdenas, C.; Doonan, P.J.; Chandramoorthy, H.C.; Irrinki, K.M.; Golenár, T.; Csordás, G.; Madireddi, P.; Yang, J.; Müller, M.; et al. MCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolism. Nat. Cell Biol. 2012, 14, 1336–1343. [Google Scholar] [CrossRef] [Green Version]
  173. Tomar, D.; Dong, Z.; Shanmughapriya, S.; Koch, D.A.; Thomas, T.; Hoffman, N.E.; Timbalia, S.A.; Goldman, S.J.; Breves, S.L.; Corbally, D.P.; et al. MCUR1 Is a Scaffold Factor for the MCU Complex Function and Promotes Mitochondrial Bioenergetics. Cell Rep. 2016, 15, 1673–1685. [Google Scholar] [CrossRef] [Green Version]
  174. Hoffman, N.E.; Chandramoorthy, H.C.; Shamugapriya, S.; Zhang, X.; Rajan, S.; Mallilankaraman, K.; Gandhirajan, R.K.; Vagnozzi, R.J.; Ferrer, L.M.; Sreekrishnanilayam, K.; et al. MICU1 motifs define mitochondrial calcium uniporter binding and activity. Cell Rep. 2013, 5, 1576–1588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Bustos, G.; Cruz, P.; Lovy, A.; Cárdenas, C. Endoplasmic reticulum-mitochondria calcium communication and the regulation of mitochondrial metabolism in cancer: A novel potential target. Front. Oncol. 2017, 7, 199. [Google Scholar] [CrossRef] [Green Version]
  176. Liu, Y.; Jin, M.; Wang, Y.; Zhu, J.; Tan, R.; Zhao, J.; Ji, X.; Jin, C.; Jia, Y.; Ren, T.; et al. MCU-induced mitochondrial calcium uptake promotes mitochondrial biogenesis and colorectal cancer growth. Signal Transduct. Target. Ther. 2020, 5, 59. [Google Scholar] [CrossRef]
  177. Liao, Y.; Dong, Y.; Cheng, J. The function of the mitochondrial calcium uniporter in neurodegenerative disorders. Int. J. Mol. Sci. 2017, 18, 248. [Google Scholar] [CrossRef]
  178. Venugopal, A.; Iyer, M.; Balasubramanian, V.; Vellingiri, B. Mitochondrial calcium uniporter as a potential therapeutic strategy for Alzheimer’s disease. Acta Neuropsychiatr. 2020, 32, 65–71. [Google Scholar] [CrossRef] [PubMed]
  179. Soman, S.; Keatinge, M.; Moein, M.; da Costa, M.; Mortiboys, H.; Skupin, A.; Sugunan, S.; Bazala, M.; Kuznicki, J.; Bandmann, O. Inhibition of the mitochondrial calcium uniporter rescues dopaminergic neurons in pink1−/− zebrafish. Eur. J. Neurosci. 2017, 45, 528–535. [Google Scholar] [CrossRef] [PubMed]
  180. Tubbs, E.; Theurey, P.; Vial, G.; Bendridi, N.; Bravard, A.; Chauvin, M.A.; Ji-Cao, J.; Zoulim, F.; Bartosch, B.; Ovize, M.; et al. Mitochondria-associated endoplasmic reticulum membrane (MAM) integrity is required for insulin signaling and is implicated in hepatic insulin resistance. Diabetes 2014, 63, 3279–3294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Wang, C.H.; Wei, Y.H. Role of mitochondrial dysfunction and dysregulation of Ca2+ homeostasis in the pathophysiology of insulin resistance and type 2 diabetes. J. Biomed. Sci. 2017, 24, 70. [Google Scholar] [CrossRef] [PubMed]
  182. Qiu, Y.; Cheng, R.; Liang, C.; Yao, Y.; Zhang, W.; Zhang, J.; Zhang, M.; Li, B.; Xu, C.; Zhang, R. MicroRNA-20b Promotes Cardiac Hypertrophy by the Inhibition of Mitofusin 2-Mediated Inter-organelle Ca2+ Cross-Talk. Mol. Ther. Nucleic Acids 2020, 19, 1343–1356. [Google Scholar] [CrossRef]
  183. Drago, I.; de Stefani, D.; Rizzuto, R.; Pozzan, T. Mitochondrial Ca2+ uptake contributes to buffering cytoplasmic Ca2+ peaks in cardiomyocytes. Proc. Natl. Acad. Sci. USA 2012, 109, 12986–12991. [Google Scholar] [CrossRef] [Green Version]
  184. Hussein, R.M.; Mohamed, W.R.; Omar, H.A. A neuroprotective role of kaempferol against chlorpyrifos-induced oxidative stress and memory deficits in rats via GSK3β-Nrf2 signaling pathway. Pestic. Biochem. Physiol. 2018, 152, 29–37. [Google Scholar] [CrossRef]
  185. Feng, H.; Cao, J.; Zhang, G.; Wang, Y. Kaempferol Attenuates Cardiac Hypertrophy via Regulation of ASK1/MAPK Signaling Pathway and Oxidative Stress. Planta Med. 2017, 83, 837–845. [Google Scholar] [CrossRef] [Green Version]
  186. Guo, Z.; Liao, Z.; Huang, L.; Liu, D.; Yin, D.; He, M. Kaempferol protects cardiomyocytes against anoxia/reoxygenation injury via mitochondrial pathway mediated by SIRT1. Eur. J. Pharmacol. 2015, 761, 245–253. [Google Scholar] [CrossRef]
  187. Schweitzer, M.K.; Wilting, F.; Sedej, S.; Dreizehnter, L.; Dupper, N.J.; Tian, Q.; Moretti, A.; My, I.; Kwon, O.; Priori, S.G.; et al. Suppression of Arrhythmia by Enhancing Mitochondrial Ca2+ Uptake in Catecholaminergic Ventricular Tachycardia Models. JACC Basic to Transl. Sci. 2017, 2, 737–747. [Google Scholar] [CrossRef] [PubMed]
  188. Sanganahalli, B.G.; Herman, P.; Hyder, F.; Kannurpatti, S.S. Mitochondrial calcium uptake capacity modulates neocortical excitability. J. Cereb. Blood Flow Metab. 2013, 33, 1115–1126. [Google Scholar] [CrossRef] [Green Version]
  189. Montero, M.; Lobatón, C.D.; Hernández-Sanmiguel, E.; Santodomingo, J.; Vay, L.; Moreno, A.; Alvarez, J. Direct activation of the mitochondrial calcium uniporter by natural plant flavonoids. Biochem. J. 2004, 384, 19–24. [Google Scholar] [CrossRef] [Green Version]
  190. Ashrafizadeh, M.; Tavakol, S.; Ahmadi, Z.; Roomiani, S.; Mohammadinejad, R.; Samarghandian, S. Therapeutic effects of kaempferol affecting autophagy and endoplasmic reticulum stress. Phyther. Res. 2020, 34, 911–923. [Google Scholar] [CrossRef]
  191. Wang, H.; Chen, L.; Zhang, X.; Xu, L.; Xie, B.; Shi, H.; Duan, Z.; Zhang, H.; Ren, F. Kaempferol protects mice from D-GalN/LPS-induced acute liver failure by regulating the ER stress-Grp78-CHOP signaling pathway. Biomed. Pharmacother. 2019, 111, 468–475. [Google Scholar] [CrossRef]
  192. Rajendran, P.; Ammar, R.B.; Al-Saeedi, F.J.; Mohamed, M.E.; Elnaggar, M.A.; Al-Ramadan, S.Y.; Bekhet, G.M.; Soliman, A.M. Kaempferol Inhibits Zearalenone-Induced Oxidative Stress and Apoptosis via the PI3K/Akt-Mediated Nrf2 Signaling Pathway: In Vitro and In Vivo Studies. Int. J. Mol. Sci. 2020, 22, 217. [Google Scholar] [CrossRef]
  193. Samhan-Arias, A.K.; Martín-Romero, F.J.; Gutiérrez-Merino, C. Kaempferol blocks oxidative stress in cerebellar granule cells and reveals a key role for reactive oxygen species production at the plasma membrane in the commitment to apoptosis. Free Radic. Biol. Med. 2004, 37, 48–61. [Google Scholar] [CrossRef] [PubMed]
  194. Yang, C.; Yang, W.; He, Z.; He, H.; Yang, X.; Lu, Y.; Li, H. Kaempferol improves lung ischemia-reperfusion injury via antiinflammation and antioxidative stress regulated by SIRT1/HMGB1/NF-κB axis. Front. Pharmacol. 2020, 10, 1635. [Google Scholar] [CrossRef] [Green Version]
  195. Yang, C.; Yang, W.; He, Z.; Guo, J.; Yang, X.; Wang, R.; Li, H. Kaempferol Alleviates Oxidative Stress and Apoptosis Through Mitochondria-dependent Pathway During Lung Ischemia-Reperfusion Injury. Front. Pharmacol. 2021, 12, 12. [Google Scholar] [CrossRef] [PubMed]
  196. Wang, J.; Mao, J.; Wang, R.; Li, S.; Wu, B.; Yuan, Y. Kaempferol Protects Against Cerebral Ischemia Reperfusion Injury Through Intervening Oxidative and Inflammatory Stress Induced Apoptosis. Front. Pharmacol. 2020, 11, 424. [Google Scholar] [CrossRef] [Green Version]
  197. Naz, F.; Jyoti, S.; Siddique, Y.H. Effect of kaempferol on the transgenic Drosophila model of Parkinson’s disease. Sci. Rep. 2020, 10, 13793. [Google Scholar] [CrossRef]
  198. Beg, T.; Jyoti, S.; Naz, F.; Rahul; Ali, F.; Ali, S.K.; Reyad, A.M.; Siddique, Y.H. Protective Effect of Kaempferol on the Transgenic Drosophila Model of Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2018, 17, 421–429. [Google Scholar] [CrossRef] [PubMed]
  199. Holland, T.M.; Agarwal, P.; Wang, Y.; Leurgans, S.E.; Bennett, D.A.; Booth, S.L.; Morris, M.C. Dietary flavonols and risk of Alzheimer dementia. Neurology 2020, 94, E1749–E1756. [Google Scholar] [CrossRef] [PubMed]
  200. Srinivasan, E.; Rajasekaran, R. Comparative binding of kaempferol and kaempferide on inhibiting the aggregate formation of mutant (G85R) SOD1 protein in familial amyotrophic lateral sclerosis: A quantum chemical and molecular mechanics study. BioFactors 2018, 44, 431–442. [Google Scholar] [CrossRef]
  201. Foos, T.M.; Wu, J.Y. The role of taurine in the central nervous system and the modulation of intracellular calcium homeostasis. Neurochem. Res. 2002, 27, 21–26. [Google Scholar] [CrossRef] [PubMed]
  202. El Idrissi, A.; Trenkner, E. Taurine regulates mitochondrial calcium homeostasis. Adv. Exp. Med. Biol. 2003, 526, 527–536. [Google Scholar] [CrossRef] [PubMed]
  203. El Idrissi, A. Taurine increases mitochondrial buffering of calcium: Role in neuroprotection. Amino Acids 2008, 34, 321–328. [Google Scholar] [CrossRef] [PubMed]
  204. Allen, S.J.; Watson, J.J.; Shoemark, D.K.; Barua, N.U.; Patel, N.K. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacol. Ther. 2013, 138, 155–175. [Google Scholar] [CrossRef]
  205. Martinou, I.; Desagher, S.; Eskes, R.; Antonsson, B.; André, E.; Fakan, S.; Martinou, J.-C. The Release of Cytochrome c from Mitochondria during Apoptosis of NGF-deprived Sympathetic Neurons Is a Reversible Event. J. Cell Biol. 1999, 144, 883–889. [Google Scholar] [CrossRef] [PubMed]
  206. Carito, V.; Pingitore, A.; Cione, E.; Perrotta, I.; Mancuso, D.; Russo, A.; Genchi, G.; Caroleo, M.C. Localization of nerve growth factor (NGF) receptors in the mitochondrial compartment: Characterization and putative role. Biochim. Biophys. Acta 2012, 1820, 96–103. [Google Scholar] [CrossRef] [PubMed]
  207. Martorana, F.; Gaglio, D.; Bianco, M.R.; Aprea, F.; Virtuoso, A.; Bonanomi, M.; Alberghina, L.; Papa, M.; Colangelo, A.M. Differentiation by nerve growth factor (NGF) involves mechanisms of crosstalk between energy homeostasis and mitochondrial remodeling. Cell Death Dis. 2018, 9, 1–16. [Google Scholar] [CrossRef]
  208. Jiang, H.; Takeda, K.; Lazarovici, P.; Katagiri, Y.; Yu, Z.X.; Dickens, G.; Chabuk, A.; Liu, X.W.; Ferrans, V.; Guroff, G. Nerve Growth Factor (NGF)-induced Calcium Influx and Intracellular Calcium Mobilization in 3T3 Cells Expressing NGF Receptors. J. Biol. Chem. 1999, 274, 26209–26216. [Google Scholar] [CrossRef] [Green Version]
  209. De Bernardi, M.A.; Rabin, S.J.; Colangelo, A.M.; Brooker, G.; Mocchetti, I. TrkA mediates the nerve growth factor-induced intracellular calcium accumulation. J. Biol. Chem. 1996, 271, 6092–6098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. MacHado, A.; Ferreira, D.; Grothe, M.J.; Eyjolfsdottir, H.; Almqvist, P.M.; Cavallin, L.; Lind, G.; Linderoth, B.; Seiger, Å.; Teipel, S.; et al. The cholinergic system in subtypes of Alzheimer’s disease: An in vivo longitudinal MRI study. Alzheimers. Res. Ther. 2020, 12, 51. [Google Scholar] [CrossRef] [PubMed]
  211. Wilting, F.; Kopp, R.; Gurnev, P.A.; Schedel, A.; Dupper, N.J.; Kwon, O.; Nicke, A.; Gudermann, T.; Schredelseker, J. The antiarrhythmic compound efsevin directly modulates voltage-dependent anion channel 2 by binding to its inner wall and enhancing mitochondrial Ca2+ uptake. Br. J. Pharmacol. 2020, 177, 2947–2958. [Google Scholar] [CrossRef] [Green Version]
  212. Shimizu, H.; Schredelseker, J.; Huang, J.; Lu, K.; Naghdi, S.; Lu, F.; Franklin, S.; Fiji, H.D.G.; Wang, K.; Zhu, H.; et al. Mitochondrial Ca2+ uptake by the voltage-dependent anion channel 2 regulates cardiac rhythmicity. eLife 2015, 4, e04801. [Google Scholar] [CrossRef]
  213. Sander, P.; Feng, M.; Schweitzer, M.K.; Wilting, F.; Gutenthaler, S.M.; Arduino, D.M.; Fischbach, S.; Dreizehnter, L.; Moretti, A.; Gudermann, T.; et al. Approved drugs ezetimibe and disulfiram enhance mitochondrial Ca2+ uptake and suppress cardiac arrhythmogenesis. Br. J. Pharmacol. 2021, 178, 4518–4532. [Google Scholar] [CrossRef]
  214. Dietl, A.; Maack, C. Targeting Mitochondrial Calcium Handling and Reactive Oxygen Species in Heart Failure. Curr. Heart Fail. Rep. 2017, 14, 338–349. [Google Scholar] [CrossRef] [PubMed]
  215. Tarnopolsky, M.A. The mitochondrial cocktail: Rationale for combined nutraceutical therapy in mitochondrial cytopathies. Adv. Drug Deliv. Rev. 2008, 60, 1561–1567. [Google Scholar] [CrossRef]
  216. Pagano, G.; Talamanca, A.A.; Castello, G.; Cordero, M.D.; d’Ischia, M.; Gadaleta, M.N.; Pallardó, F.V.; Petrović, S.; Tiano, L.; Zatterale, A. Current experience in testing mitochondrial nutrients in disorders featuring oxidative stress and mitochondrial dysfunction: Rational design of chemoprevention trials. Int. J. Mol. Sci. 2014, 15, 20169–20208. [Google Scholar] [CrossRef] [PubMed]
  217. Beal, M.F. Bioenergetic approaches for neuroprotection in Parkinson’s disease. Ann. Neurol. 2003, 53, S39–S48. [Google Scholar] [CrossRef]
  218. Pallardó, F.V.; Pagano, G.; Rodríguez, L.R.; Gonzalez-Cabo, P.; Lyakhovich, A.; Trifuoggi, M. Friedreich Ataxia: Current state-of-the-art, and future prospects for mitochondrial-focused therapies. Transl. Res. 2021, 229, 135–141. [Google Scholar] [CrossRef]
  219. Birk, A.V.; Liu, S.; Soong, Y.; Mills, W.; Singh, P.; Warren, J.D.; Seshan, S.V.; Pardee, J.D.; Szeto, H.H. The mitochondrial-targeted compound SS-31 re-energizes ischemic mitochondria by interacting with cardiolipin. J. Am. Soc. Nephrol. 2013, 24, 1250–1261. [Google Scholar] [CrossRef]
  220. Zhao, H.; Li, H.; Hao, S.; Chen, J.; Wu, J.; Song, C.; Zhang, M.; Qiao, T.; Li, K. Peptide SS-31 upregulates frataxin expression and improves the quality of mitochondria: Implications in the treatment of Friedreich ataxia. Sci. Rep. 2017, 7, 9840. [Google Scholar] [CrossRef] [Green Version]
  221. Zhao, W.; Xu, Z.; Cao, J.; Fu, Q.; Wu, Y.; Zhang, X.; Long, Y.; Zhang, X.; Yang, Y.; Li, Y.; et al. Elamipretide (SS-31) improves mitochondrial dysfunction, synaptic and memory impairment induced by lipopolysaccharide in mice. J. Neuroinflammation 2019, 16, 1–19. [Google Scholar] [CrossRef]
  222. Karaa, A.; Haas, R.; Goldstein, A.; Vockley, J.; Cohen, B.H. A randomized crossover trial of elamipretide in adults with primary mitochondrial myopathy. J. Cachexia. Sarcopenia Muscle 2020, 11, 909–918. [Google Scholar] [CrossRef] [Green Version]
  223. An Intermediate Size Expanded Access Protocol of Elamipretide-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04689360?term=elamipretide&draw=2&rank=8 (accessed on 26 October 2021).
  224. Tauskela, J.S. MitoQ--a mitochondria-targeted antioxidant. IDrugs 2007, 10, 399–412. [Google Scholar]
  225. Xiao, L.; Xu, X.; Zhang, F.; Wang, M.; Xu, Y.; Tang, D.; Wang, J.; Qin, Y.; Liu, Y.; Tang, C.; et al. The mitochondria-targeted antioxidant MitoQ ameliorated tubular injury mediated by mitophagy in diabetic kidney disease via Nrf2/PINK1. Redox Biol. 2017, 11, 297–311. [Google Scholar] [CrossRef]
  226. Zhang, S.; Zhou, Q.; Li, Y.; Zhang, Y.; Wu, Y. MitoQ Modulates Lipopolysaccharide-Induced Intestinal Barrier Dysfunction via Regulating Nrf2 Signaling. Mediators Inflamm. 2020, 2020, 3276148. [Google Scholar] [CrossRef] [Green Version]
  227. Escribano-Lopez, I.; Bañuls, C.; Diaz-Morales, N.; Iannantuoni, F.; Rovira-Llopis, S.; Gomis, R.; Rocha, M.; Hernandez-Mijares, A.; Murphy, M.P.; Victor, V.M. The mitochondria-targeted antioxidant MitoQ modulates mitochondrial function and endoplasmic reticulum stress in pancreatic β cells exposed to hyperglycaemia. Cell. Physiol. Biochem. 2019, 52, 186–197. [Google Scholar] [CrossRef]
  228. Ribeiro, R.F., Jr.; Dabkowski, E.R.; Shekar, K.C.; O’Connell, K.A.; Hecker, P.A.; Murphy, M.P. MitoQ improves mitochondrial dysfunction in heart failure induced by pressure overload. Free Radic. Biol. Med. 2018, 117, 18–29. [Google Scholar] [CrossRef]
  229. Kang, L.; Liu, S.; Li, J.; Tian, Y.; Xue, Y.; Liu, X. The mitochondria-targeted anti-oxidant MitoQ protects against intervertebral disc degeneration by ameliorating mitochondrial dysfunction and redox imbalance. Cell Prolif. 2020, 53, e12779. [Google Scholar] [CrossRef]
  230. Effects of Mitochondrial-targeted Antioxidant on Mild Cognitive Impairment (MCI) Patients-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03514875?term=mitoQ&draw=3&rank=14 (accessed on 27 October 2021).
  231. Vascular Function in Health and Disease-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT02966665?term=mitoQ&draw=3&rank=19 (accessed on 27 October 2021).
  232. MitoQ for Fatigue in Multiple Sclerosis (MS)-Full Text View-ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04267926?term=mitoQ&draw=2&rank=3 (accessed on 27 October 2021).
  233. Goodfellow, M.J.; Borcar, A.; Proctor, J.L.; Greco, T.; Rosenthal, R.E.; Fiskum, G. Transcriptional activation of antioxidant gene expression by Nrf2 protects against mitochondrial dysfunction and neuronal death associated with acute and chronic neurodegeneration. Exp. Neurol. 2020, 328, 113247. [Google Scholar] [CrossRef]
  234. Rodríguez, L.R.; Lapeña, T.; Calap-Quintana, P.; Moltó, M.D.; Gonzalez-Cabo, P.; Langa, J.A.N. Antioxidant therapies and oxidative stress in friedreich’s ataxia: The right path or just a diversion? Antioxidants 2020, 9, 664. [Google Scholar] [CrossRef]
  235. Huang, M.L.H.; Chiang, S.; Kalinowski, D.S.; Bae, D.H.; Sahni, S.; Richardson, D.R. The role of the antioxidant response in mitochondrial dysfunction in degenerative diseases: Cross-talk between antioxidant defense, autophagy, and apoptosis. Oxid. Med. Cell. Longev. 2019, 2019, 6392763. [Google Scholar] [CrossRef] [Green Version]
  236. Miller, E.D.; Dziedzic, A.; Saluk-Bijak, J.; Bijak, M. A review of various antioxidant compounds and their potential utility as complementary therapy in multiple sclerosis. Nutrients 2019, 11, 1528. [Google Scholar] [CrossRef] [Green Version]
  237. Brandes, M.S.; Gray, N.E. NRF2 as a Therapeutic Target in Neurodegenerative Diseases. ASN Neuro 2020, 12, 12. [Google Scholar] [CrossRef]
  238. Silvestro, S.; Sindona, C.; Bramanti, P.; Mazzon, E. A state of the art of antioxidant properties of curcuminoids in neurodegenerative diseases. Int. J. Mol. Sci. 2021, 22, 3168. [Google Scholar] [CrossRef] [PubMed]
  239. Larrea, D.; Pera, M.; Gonnelli, A.; Quintana-Cabrera, R.; Akman, H.O.; Guardia-Laguarta, C.; Velasco, K.R.; Area-Gomez, E.; Dal Bello, F.; de Stefani, D.; et al. MFN2 mutations in charcot-marie-Tooth disease alter mitochondria-Associated er membrane function but do not impair bioenergetics. Hum. Mol. Genet. 2019, 28, 1782–1800. [Google Scholar] [CrossRef] [Green Version]
  240. Gdynia, H.J.; Kurt, A.; Endruhn, S.; Ludolph, A.C.; Sperfeld, A.D. Cardiomyopathy in motor neuron diseases. J. Neurol. Neurosurg. Psychiatry 2006, 77, 671–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Liang, J.J.; Goodsell, K.; Grogan, M.; Ackerman, M.J. LMNA-Mediated Arrhythmogenic Right Ventricular Cardiomyopathy and Charcot-Marie-Tooth Type 2B1: A Patient-Discovered Unifying Diagnosis. J. Cardiovasc. Electrophysiol. 2016, 27, 868–871. [Google Scholar] [CrossRef] [PubMed]
  242. Weidemann, F.; Störk, S.; Liu, D.; Hu, K.; Herrmann, S.; Ertl, G.; Niemann, M. Cardiomyopathy of Friedreich ataxia. J. Neurochem. 2013, 126, 88–93. [Google Scholar] [CrossRef] [PubMed]
  243. Espinós, C.; Galindo, M.I.; García-Gimeno, M.A.; Ibáñez-Cabellos, J.S.; Martínez-Rubio, D.; Millán, J.M.; Rodrigo, R.; Sanz, P.; Seco-Cervera, M.; Sevilla, T.; et al. Oxidative stress, a crossroad between rare diseases and neurodegeneration. Antioxidants 2020, 9, 313. [Google Scholar] [CrossRef] [Green Version]
  244. Jiang, T.; Sun, Q.; Chen, S. Oxidative stress: A major pathogenesis and potential therapeutic target of antioxidative agents in Parkinson’s disease and Alzheimer’s disease. Prog. Neurobiol. 2016, 147, 1–19. [Google Scholar] [CrossRef]
  245. Lu, X.; Yang, H.; Chen, Y.; Li, Q.; He, S.; Jiang, X.; Feng, F.; Qu, W.; Sun, H. The Development of Pharmacophore Modeling: Generation and Recent Applications in Drug Discovery. Curr. Pharm. Des. 2018, 24, 3424–3439. [Google Scholar] [CrossRef]
  246. Pinzi, L.; Rastelli, G. Molecular docking: Shifting paradigms in drug discovery. Int. J. Mol. Sci. 2019, 20, 4331. [Google Scholar] [CrossRef] [Green Version]
  247. Kutlushina, A.; Khakimova, A.; Madzhidov, T.; Polishchuk, P. Ligand-Based Pharmacophore Modeling Using Novel 3D Pharmacophore Signatures. Molecules 2018, 23, 3094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic representation showing the effects of mitochondrial Ca2+ uptake promoters. The ER is the main Ca2+ storage in the cell and Ca2+ exchange between the ER and the mitochondria requires the formation of tethers composed by proteins of both compartments. Sig-1R resides in the ER membrane, in a dormant, Ca2+-dependent state. Upon activation by agonists, Sig-1R dissociates from BiP/GRP78 and reallocates within the ER membrane, interacting with IP3R and chaperoning the protein complex that transfers Ca2+ to the mitochondria. This complex formed by IP3R-GRP75-VDAC ensures a rapid Ca2+ flux to the mitochondrial intermembrane space, which triggers MCU opening and Ca2+ to cross the mitochondrial inner membrane. Several models of neurological disorders such as ALS, CMT and FRDA have exhibited alterations in mitochondrial Ca2+ buffering by defective appositions between the two organelles. Both Sig-1R agonists and MCU enhancers promote Ca2+ exchange between the ER and the mitochondria, exerting beneficial effects in different models of neurological diseases. On the one hand, Sig-1R agonists (pridopidine, SA4503, Blacarmesine, PRE-084, and fluvoxamine) have been demonstrated to exert neuroprotective effects, improving mitochondrial dysfunction, preventing cells from apoptosis, activating the antioxidant response, ameliorating ER stress, and improving axonal defects. On the other hand, the MCU enhancer, Kaempferol, has helped to improve mitochondrial dysfunction, activate the oxidative stress response, modulate autophagy, regulate ER stress, and prevent cells from apoptosis. This figure has been created using Creative Commons resources from Servier Medical Art [99]. ALS: amyotrophic lateral sclerosis; Bip/GRP78: binding immunoglobulin protein/glucose-regulated protein 78; CMT: Charcot–Marie–Tooth; ER: endoplasmic reticulum; FRDA: Friedreich’s ataxia; GRP75: glucose-regulated protein 75; IP3R: inositol 1,4,5-trisphosphate receptor; MCU: mitochondrial calcium uniporter; Sig-1R: sigma non-opioid intracellular receptor 1; VDAC: voltage-dependent anion channel.
Figure 1. Schematic representation showing the effects of mitochondrial Ca2+ uptake promoters. The ER is the main Ca2+ storage in the cell and Ca2+ exchange between the ER and the mitochondria requires the formation of tethers composed by proteins of both compartments. Sig-1R resides in the ER membrane, in a dormant, Ca2+-dependent state. Upon activation by agonists, Sig-1R dissociates from BiP/GRP78 and reallocates within the ER membrane, interacting with IP3R and chaperoning the protein complex that transfers Ca2+ to the mitochondria. This complex formed by IP3R-GRP75-VDAC ensures a rapid Ca2+ flux to the mitochondrial intermembrane space, which triggers MCU opening and Ca2+ to cross the mitochondrial inner membrane. Several models of neurological disorders such as ALS, CMT and FRDA have exhibited alterations in mitochondrial Ca2+ buffering by defective appositions between the two organelles. Both Sig-1R agonists and MCU enhancers promote Ca2+ exchange between the ER and the mitochondria, exerting beneficial effects in different models of neurological diseases. On the one hand, Sig-1R agonists (pridopidine, SA4503, Blacarmesine, PRE-084, and fluvoxamine) have been demonstrated to exert neuroprotective effects, improving mitochondrial dysfunction, preventing cells from apoptosis, activating the antioxidant response, ameliorating ER stress, and improving axonal defects. On the other hand, the MCU enhancer, Kaempferol, has helped to improve mitochondrial dysfunction, activate the oxidative stress response, modulate autophagy, regulate ER stress, and prevent cells from apoptosis. This figure has been created using Creative Commons resources from Servier Medical Art [99]. ALS: amyotrophic lateral sclerosis; Bip/GRP78: binding immunoglobulin protein/glucose-regulated protein 78; CMT: Charcot–Marie–Tooth; ER: endoplasmic reticulum; FRDA: Friedreich’s ataxia; GRP75: glucose-regulated protein 75; IP3R: inositol 1,4,5-trisphosphate receptor; MCU: mitochondrial calcium uniporter; Sig-1R: sigma non-opioid intracellular receptor 1; VDAC: voltage-dependent anion channel.
Antioxidants 11 00165 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rodríguez, L.R.; Lapeña-Luzón, T.; Benetó, N.; Beltran-Beltran, V.; Pallardó, F.V.; Gonzalez-Cabo, P.; Navarro, J.A. Therapeutic Strategies Targeting Mitochondrial Calcium Signaling: A New Hope for Neurological Diseases? Antioxidants 2022, 11, 165. https://doi.org/10.3390/antiox11010165

AMA Style

Rodríguez LR, Lapeña-Luzón T, Benetó N, Beltran-Beltran V, Pallardó FV, Gonzalez-Cabo P, Navarro JA. Therapeutic Strategies Targeting Mitochondrial Calcium Signaling: A New Hope for Neurological Diseases? Antioxidants. 2022; 11(1):165. https://doi.org/10.3390/antiox11010165

Chicago/Turabian Style

Rodríguez, Laura R., Tamara Lapeña-Luzón, Noelia Benetó, Vicent Beltran-Beltran, Federico V. Pallardó, Pilar Gonzalez-Cabo, and Juan Antonio Navarro. 2022. "Therapeutic Strategies Targeting Mitochondrial Calcium Signaling: A New Hope for Neurological Diseases?" Antioxidants 11, no. 1: 165. https://doi.org/10.3390/antiox11010165

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