Next Article in Journal
The Sin3A/MAD1 Complex, through Its PAH2 Domain, Acts as a Second Repressor of Retinoic Acid Receptor Beta Expression in Breast Cancer Cells
Previous Article in Journal
Mesenchymal Stromal Cell-Derived Extracellular Vesicles for Neonatal Lung Disease: Tiny Particles, Major Promise, Rigorous Requirements for Clinical Translation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regulation of Aging and Longevity by Ion Channels and Transporters

by
Kartik Venkatachalam
1,2
1
Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center (UTHealth-Houston), Houston, TX 77030, USA
2
Graduate Programs in Neuroscience and Biochemistry and Cell Biology, MD Anderson Cancer Center and UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
Cells 2022, 11(7), 1180; https://doi.org/10.3390/cells11071180
Submission received: 1 February 2022 / Revised: 22 March 2022 / Accepted: 29 March 2022 / Published: 31 March 2022
(This article belongs to the Section Intracellular and Plasma Membranes)

Abstract

:
Despite significant advances in our understanding of the mechanisms that underlie age-related physiological decline, our ability to translate these insights into actionable strategies to extend human healthspan has been limited. One of the major reasons for the existence of this barrier is that with a few important exceptions, many of the proteins that mediate aging have proven to be undruggable. The argument put forth here is that the amenability of ion channels and transporters to pharmacological manipulation could be leveraged to develop novel therapeutic strategies to combat aging. This review delves into the established roles for ion channels and transporters in the regulation of aging and longevity via their influence on membrane excitability, Ca2+ homeostasis, mitochondrial and endolysosomal function, and the transduction of sensory stimuli. The goal is to provide the reader with an understanding of emergent themes, and prompt further investigation into how the activities of ion channels and transporters sculpt the trajectories of cellular and organismal aging.

1. Introduction

All biological processes lose their integrity with the passage of time. Aging is the single greatest risk factor for diseases associated with diminished survivorship such as cancer, diabetes, cardiovascular diseases, and neurodegeneration [1,2]. By extension, strategies that ameliorate age-dependent diseases also extend lifespan. Over the past three decades, major advances in gerobiology have been made using model organisms such as C. elegans and Drosophila [3,4,5,6]. These pioneering studies have established that various biochemical pathways influence the rates of cellular and organismal aging [2]. Emerging insights have informed investigations into the genetics of aging, and prompted the quest to identify drugs that extend healthspan by ameliorating age-related diseases or by slowing the rates of biological aging.
The problem, however, is that barring a few exceptions (e.g., rapamycin and metformin [2]) not many drugs have emerged as suitable candidates to combat aging. Although this barrier has many explanations, the most obvious is the sheer complexity of aging. Age-related loss of physiological integrity stems from a host of defects, including genomic and epigenetic instability, telomere shortening, proteostatic imbalance, chronic inflammation, dysregulated nutrient sensing, mitochondrial defects, cellular senescence, and stem cell exhaustion [1,2]. Most proteins and signaling cascades involved in the regulation of these hallmarks are undruggable.
With perhaps the exception of G-protein coupled receptors, ion channels and transporters count among the most druggable targets in the proteome. This is because domains that mediate ion transport usually interface well with small molecules. Indeed, many naturally occurring biological toxins target ion channels and transporters with exquisite sensitivity, and these entities have been used to engineer novel pharmacological modifiers. Furthermore, since the activity of ion channels and transporters can be assessed in vitro using electrophysiology or optical imaging, it is possible to conduct high-throughput screens to discover suitable activators and inhibitors. Although it might not be possible to extend healthspan or longevity by targeting channels that function in just one pathway, a cogent strategy for the amelioration of age-dependent loss of function could involve the identification and targeting of ion channels and transporters that affect multiple pathways related to organismal aging. With this rationale in mind, this article provides an overview of how aging and longevity are determined by the activities of various ion channels and transporters. The overarching goal of this review is to highlight commonalities and encourage further investigation into the modulation of the activities of ion channels and transporters to influence the rates of cellular and organismal aging.

2. Regulation of Aging and Longevity by Ion Channels and Transporters Related to Ca2+ Homeostasis and Electrical Excitability

Ca2+ is a key second messenger that is involved in the regulation of a host of different pathways, including synaptic transmission, neuronal survival, cellular bioenergetics, and gene expression that range over timescales spanning from milliseconds to hours and days [7,8]. Critical to cellular Ca2+ signaling are ATP-consuming transporters that maintain cytosolic Ca2+ concentration ([Ca2+]) in the nanomolar range by pumping the cation against its steep concentration gradient (~10,000-fold across the plasma membrane and endoplasmic reticulum (ER) membrane) [7,8]. Movement of Ca2+ down its concentration gradient involves the opening of Ca2+-permeable channels that serve as conduits that permit transient elevations of cytosolic [Ca2+], and the attendant activation of relevant cellular pathways [7,9].
One of the most exciting recent findings in gerobiology is that expression of genes associated with the maintenance of neuronal Ca2+ homeostasis, excitability, and synaptic function is anticorrelated with lifespan in many organisms including humans [10]. Analysis of RNA-seq and microarray datasets generated from frontal cortices isolated immediately post-mortem revealed the downregulation of genes associated with neurotransmission in exceptionally long-lived humans [10] (Figure 1A). Advanced age is also associated with increased neuronal activity in C. elegans, and pharmacological attenuation of excitability and its consequences using the Cl channel agonist, ivermectin, or the L-type voltage-gated Ca2+ channel (VGCC) blocker, nimodipine, extended worm lifespan [10] (Figure 1B). Roles for excitability in the regulation of neuronal viability and organismal lifespan are also observed in Drosophila [11,12,13,14,15] (Figure 1B). Deletion of the fly K+ channel genes, ether a go-go (eag) and shaker (Sh)—expected to augment neuronal excitability—led to severe loss of motor coordination, sleep deficits, and shorter lifespan [11,15]. Diminished Na+/K+ ATPase activity, which leads to a loss of the resting membrane potential and severe neuronal hyperexcitability, also caused shorter lifespan in flies [11,12,16].
The relationship between neuronal excitability and lifespan is conserved in mammals [10]. As is the case in C. elegans, ivermectin extends lifespan in a mouse model of amyotrophic lateral sclerosis (ALS) [17]. Plasma membrane excitability is also an important determinant of oncogene-induced senescence (OIS) in non-excitable mammalian cells [18] (Figure 1B). A genetic screen revealed that loss of SCN9A, the gene encoding a voltage-gated Na+ channel, protected human fibroblasts from OIS [18]. Oncogenic signals or activation of the tumor suppressor, p53, led to the induction of SCN9A with NF-kB as an intermediary [18]. Elevations in SCN9A abundance, which is expected to augment plasma membrane excitability, led to senescence via the retinoblastoma (Rb) pathway [18]. Whether NF-kB signaling, p53, or Rb also contribute to the shorter lifespan observed in animals with chronic neuronal depolarization (see below) remains to be addressed.

2.1. Regulation of Aging by Ca2+ Channels in the Plasma Membrane

Converging lines of evidence indicate a central role for plasma membrane-resident Ca2+ channels in the regulation of longevity [19,20]. Aged mammalian nervous system is characterized by increased abundance of L-type VGCCs [21]. Concordantly, verapamil-mediated inhibition of L-type channels ameliorated neurodegeneration and extended lifespan in a mouse model of ALS [22]. Similarly, N-type VGCCs (Cav2.2) contribute to age-related neuroinflammation [23]. Pharmacological inhibition of Cav2.2 channels blunts neuroinflammation in aged mice [23]. In endothelial cells, VGCC activity contributes to replicative senescence, and the VGCC inhibitor, nifedipine, reduces the fraction of senescent cells in the endothelium [24]. These findings in mammalian models agree with those in aged C. elegans as pertaining to the extension of lifespan conferred by the administration of L-type VGCC inhibitors, nimodipine, and verapamil [10,19]. In further agreement, knockdown of the VGCC subunit, unc-36, extends C. elegans lifespan [20].
Members of the transient receptor potential (TRP) superfamily of cation channels are also important in age-dependent changes in various cell types. Loss of the gene encoding TRPC5 protects mouse endothelial cells from the induction of senescence markers such as p53 and p21, and from oxidative stress-induced senescence [25]. These findings point to TRPC5 as an actionable target in the regulation of vascular aging. Activity of members of the Melastatin subfamily of TRP channels (TRPM) also impacts the onset of cellular senescence. TRPM2 conductance is positively correlated with oxidative stress-induced neuronal senescence [26]. Indeed, the mitigation of oxidative stress by glutathione is associated with the inhibition of TRPM2, whereas depletion of glutathione is associated with augmented TRPM2 activity [26]. In contrast, TRPM7 and TRPM8 abundances are elevated in pancreatic cancer where they play roles in the amelioration of OIS and replicative senescence [27,28,29]. Knockdown of the genes encoding TRPM7 and TRPM8 induces senescence in pancreatic cancer cells [27,28,29].
Years of study have pointed to a critical role for NMDA receptors in Ca2+ excitotoxicity [30]. As such, hyperactive N-methyl D-aspartate (NMDA) receptors contribute to the shortening of lifespan observed in models of neurodegenerative disease. For instance, spinocerebellar ataxia type 1 (SCA1), which is caused by a CAG repeat expansion in the SCA1 gene, is associated with the pathologically elevated activity of extrasynaptic NMDA receptors [31]. Long-term administration of the NMDA receptor antagonist, memantine, attenuated cerebellar neurodegeneration and extended animal lifespan in a mouse model of SCA1 [31]. Similarly, in a Drosophila model of C9ORF72 ALS, inhibition of NMDA receptors delayed motor dysfunction and extended animal lifespan [32]. These studies demonstrate that excessive NMDA receptor activity in models of neurodegenerative diseases underlies the loss of neuronal viability and shortening of lifespan.
On the other hand, loss of cognitive function associated with advanced age may stem from an age-dependent decrease in NMDA function. NMDA receptor densities in parts of the brain such as the hippocampus decrease with age, and in models of Alzheimer’s disease [33,34,35]. These changes underlie aspects of age-dependent cognitive decline. In this context, NMDA receptor activity has been linked to a transcriptional antioxidant response aimed at preventing activity-dependent loss of synaptic transmission [36]. Interventions that promote NMDA receptor function in aged animals, therefore, ameliorate the onset of learning and memory deficits [34,35,37,38,39]. Together, these studies point to the importance of optimal NMDA receptor activity for the maintenance of brain resilience with the passage of time, such that either an increase or decrease in NMDA receptor activity can shorten lifespan.

2.2. Regulation of Aging and Longevity by Ca2+ Channels in the Endoplasmic Reticulum (ER)

The notion that deviation from the optimal range of channel activity can become toxic is also the case for Ca2+ channels that localize to the ER [8]. Inositol trisphosphate receptors (IP3Rs) are ER-resident Ca2+ channels that are activated by IP3 generated by phospholipase C isoforms (PLCβ) such as those coupled to G-protein coupled receptors (GPCRs) [40] (Figure 2). In case of the gene encoding IP3R type 1 (IP3R1, encoded by ITPR1), loss-of-function mutations result in spinocerebellar ataxia types 15 and 29 (SCA15 and SCA29, respectively), Gillespie syndrome, and sporadic infantile-onset cerebellar ataxia [41,42,43,44,45,46]. While most mice carrying Itpr1 deletion die as embryos, the escapers exhibit severe ataxia and tonic-clonic seizures that lead to death by the weaning period [47]. Furthermore, mutations that lowered the abundance of IP3R1 in the cerebellar Purkinje neurons induced ataxia, and sensitized neurons to cell death during ER stress [41,48]. On the other hand, elevated IP3R1 activity underlies the neuropathology in SCA2 and SCA3 [49,50]. Augmented IP3R1 activity also contributes to neuronal cell death in Huntington disease (HD) and Niemann-Pick C1 disease [51,52]. Therefore, either an increase or decrease in IP3R1 activity is toxic to neurons, and can thus shorten lifespan.
Neurons are also sensitive to the dosage of the IP3R2 isoform. Suggesting that IP3R2 protects against ALS, deletion of Itpr2 in the SOD1G93A mouse model of ALS promoted inflammation and diminished animal survival [53]. Therefore, higher ITPR2 expression in the spinal cord of humans with ALS [53], suggests the engagement of a protective response involving IP3R2. In the absence of the ALS-related mutations, however, IP3R2 contributes to the adverse consequences of advanced age. Itpr2 knockout mice exhibit decreased age-related pathology resulting in longer lifespan, resistance to metabolic stress, and diminished immunosenescence [54]. Even at the cellular level, deletion or repression of Itpr2 prevented OIS or ROS-induced senescence [54,55,56,57]. This effect on senescence is due to diminished interorganellar transfer of Ca2+ between the ER and mitochondria, which is otherwise mediated very effectively by IP3R2.
In Drosophila, itpr overexpression in glutamatergic neurons is sufficient to shorten animal lifespan [12]. Knockdown of itpr mitigates the effects of neurodegeneration-causing transgenes on fly lifespan and locomotion [12,58]. Together, these findings point to a necessary and sufficient role for neuronal IP3R activity in the regulation of fly lifespan. Several lines of evidence suggest that elevated IP3R activity was a consequence of chronic depolarization of fly neurons expressing neurodegeneration-causing transgenes [12]. First, expression of ALS and tauopathy-related transgenes in fly glutamatergic neurons led to concomitant loss of membrane potential and elevated IP3R-mediated ER Ca2+ release. Second, abbreviated lifespans in flies expressing these transgenes were ameliorated by the knockdown of itpr. Third, expression of dominant-negative variant of a subunit of the Na+/K+ ATPase to prevent neuronal repolarization shortened animal lifespan via IP3R activity. Fourth, Ca2+ imaging experiments demonstrated that depolarization was sufficient to induce greater IP3R-dependent ER Ca2+ release in response to invariant concentrations of agonists of PLCβ-coupled receptors.
Investigation of the mechanisms by which plasma membrane potential modulates IP3R activity revealed that depolarization induced greater association between PLCβ and its phosphoinositide substrate, phosphatidylinositol-(4,5)-bisphosphate (PIP2). Therefore, stimulation of PLCβ-coupled receptors in depolarized neurons enhanced PIP2 hydrolysis, which resulted in higher levels of IP3 and augmented IP3R activity [12] (Figure 3A). Either the deletion of the gene encoding a fly PLCβ isoform or the knockdown of the gene encoding a PLCβ-coupled GPCR, prevented depolarization from abbreviating fly lifespan [12]. Even in C. elegans, attenuation of PLCβ signaling extended lifespan by repressing the insulin–DAF16/FOXO axis [59,60,61].

2.3. Transcriptional Control of Excitability and Longevity

The relationship between longevity and the neuronal excitability is mediated by Repressor Element-1 (RE1) Silencing Transcription Factor (REST, also called NRSF), which represses genes carrying the RE1 motif [10,62,63]. REST appears to promote resilience to age by decreasing the expression of genes necessary for promoting neuronal excitability [10,64] (Figure 3B). REST-deficient mice exhibit cortical hyperexcitability, a propensity for epileptic seizures, and enhanced mortality [10]. In C. elegans, knockdown of REST orthologs augments neuronal excitability and shortens lifespan, whereas elevated REST activity represses neuronal excitability and lengthens animal lifespan [10]. In worms, REST and attendant neuronal excitability impact on longevity via the regulation of DAF16/FOXO signaling [10,59,60].
The findings that expression of neurodegeneration-causing transgenes in Drosophila glutamatergic neurons results in both hyperexcitability and shorter lifespan [12] is consistent with the notion of REST dysregulation in models of neurodegeneration [65]. Deletion of REST is sufficient to induce degeneration in mouse and C. elegans nervous systems [66]. REST is neuroprotective in aged human neurons, iPSC-derived neurons in patients with Alzheimer’s disease (AD), and in mouse models of AD [10,66,67]. Mechanistically, REST function is lost in AD patients and in individuals with age-dependent cognitive impairment due to increased sequestration into autophagosomes, and depletion from the nucleus [66,67] (Figure 3B). It is also possible that REST might get trapped in protein aggregates that are observed in many neurodegenerative diseases (Figure 3B), although this possibility has not been demonstrated directly. An exception to the model that REST is ubiquitously neuroprotective is the case in HD. In HD neurons, REST exhibits aberrantly high nuclear localization, increased RE1 occupancy, and expected gene repression [68,69]. Whether these outcomes contribute to HD pathology, and how this phenotype relates to that observed in AD remain incompletely understood.

3. Regulation of Aging and Longevity by Mitochondrial Ion Channels and Transporters

Mitochondria are critical for life at both cellular and organismal levels. Mitochondrial function declines in older animals, and the underlying mechanisms are major contributors to the adverse outcomes of advanced age [70]. This section aspires to describe the contributions of mitochondrial ion channels and transporters to the regulation of aging and longevity.

3.1. Mitochondrial Ca2+ Uniporter

Uptake of Ca2+ into the mitochondrial matrix is critical for many aspects of mitochondrial function, including ATP production (Figure 4). Ca2+ transport across the inner mitochondrial membrane is mediated by the mitochondrial Ca2+ uniporter (MCU) and its regulators, mitochondrial Ca2+ uptake 1-3 (MICU1-3), mitochondrial Ca2+ uniporter regulator 1 (MCUR1), and essential MCU regulator (EMRE) [71,72,73,74,75]. Close coupling between the ER and mitochondria permits the Ca2+ ions released via IP3Rs to be taken up into the mitochondrial matrix through the uniporter [71,76] (Figure 4). Coordinated activities of IP3Rs and MCU, therefore, permit Ca2+ transport from the ER to mitochondria, which in turn, stimulates mitochondrial bioenergetics (Figure 4). This mode of interorganellar Ca2+ transport is needed for C. elegans longevity [61]. In agreement, knockdown of MCU in Drosophila neurons further abbreviates the lifespan of animals experiencing precocious mortality due to diminished ER–mitochondria Ca2+ exchange [12]. In mice, MICU3 abundance and MCU-dependent mitochondrial Ca2+ uptake decrease in the aging skeletal muscle leading to sarcopenia [77]. Restoration of MICU3 abundance in mouse muscle increased myogenesis and delayed sarcopenia [77].
While uniporter activity within the physiological range is necessary for animal viability, transporter overactivity has been observed in many pathological conditions (Figure 4). MCU and MICU1 contribute mitochondrial Ca2+ overload that occurs in various neurodegenerative and pathological conditions that shorten lifespan. Untrammeled MCU activity contribute to neurodegeneration in SCA, and knockdown of Mcu in mouse neurons ameliorates mitochondrial Ca2+ overload and excitotoxicity stemming from NMDA receptor activation [78,79]. In mouse cardiomyocytes, deletion of Mcu protects from ischemia reperfusion injury [80,81]. Furthermore, knockdown of the gene encoding a regulatory partner of MCU, MCUR1, attenuates mitochondrial Ca2+ uptake and ATP production, and drives pro-survival pathways such as AMPK activation and autophagy [75].
MCU also senses and responds to mitochondrial oxidative stress and redox balance. Glutathione moieties can be added to MCU, which attenuates the Ca2+ transport activity of the protein, and thereby, limits mitochondrial metabolism and reactive oxygen species (ROS) production [82]. In Drosophila muscle, MCU is necessary for oxidative stress-induced cell death, and deletion of fly MCU confers robust resistance to oxidative stress-dependent lethality [83]. Even OIS requires MCU-dependent mitochondrial Ca2+ overload subsequent to ER Ca2+ release via IP3Rs [56]. In view of the model that ROS production contributes to aging [84,85], MCU could be considered as drivers of aging and senescence.

3.2. Other Mitochondrial Channels and Transporters

Opposing the effects of the uniporter are transporters that extrude matrix Ca2+. Removal of matrix Ca2+ under physiological conditions is largely dependent on the mitochondrial Na+/Ca2+/Li+ exchanger (NCLX), with the permeability transition pore playing a role during Ca2+ overload [86,87,88,89]. Although Ca2+/H+ exchange has also been proposed to serve a role in the removal of mitochondrial Ca2+, molecular identity of the exchanger remains controversial [89,90,91].
Deletion of NCLX in the adult mouse heart has led to sudden death due to heart failure [88]. The underlying mechanism, not surprisingly, involved mitochondrial Ca2+ overload, unremitting ROS production, and cell death [88]. Conversely, increased abundance of NCLX in the mouse heart protects against ischemic cell death and heart failure [88]. In dissociated hippocampal neurons challenged with excitotoxic stimuli, knockdown of the gene encoding NCLX further dysregulates mitochondrial Ca2+ homeostasis and promotes ROS production [92]. Diminished abundance of NCLX in neurons and glia, and attendant impairments in the removal of mitochondrial Ca2+ are sufficient to elicit neurodegeneration [92]. Indeed, deletion of NCLX accelerates amyloid plaque formation, tau neuropathology, and the rate of memory decline in a mouse model of AD [93]. Mitochondrial [Ca2+] is also constitutively elevated due to diminished NCLX activity in multiple mouse models of Parkinson’s disease [94,95,96].
Many studies have pointed to roles for K+ channels in mitochondrial function. Small conductance Ca2+-activated K+ channels (SK2 channels) localize to the inner mitochondrial membrane and mediate mitochondrial K+ currents [97]. Activation of mitochondrial SK2 channels protects cells against mitochondria-dependent cell death when challenged by excitotoxic insults [97]. In C. elegans, pharmacological activation of SK channels promotes longevity by conferring resistance to ferroptosis [98]. Big conductance Ca2+-activated K+ channels (BK channels) are also found in the inner mitochondrial membrane of cardiomyocytes, where they mediate Ca2+-activated K+ currents and protect the heart from ischemic injury [99]. In Drosophila, absence of mitochondrial BK channels led to increased ROS production and abbreviated fly lifespan, whereas overexpression of these channels led to long-lived animals [100].

4. Mitochondrial Uncoupling Proteins and Longevity

Uncoupling proteins (UCPs) are integral proteins of the inner mitochondrial membrane that mediate the transport of protons from the intermembrane space (IMS) to the matrix [101,102] (Figure 5). As such, mitochondrial UCPs dissipate the electrochemical gradient across the inner mitochondrial membrane, which otherwise drives ATP synthesis via ATP synthase [101] (Figure 5). Because dissipation of the protonmotive force leads to release of the energy derived from oxidized substrates in the form of heat, UCP1-mediated proton leak drives thermogenesis in brown adipose tissue [101,102,103,104,105]. UCPs 2-5, on the other hand, have limited roles in thermogenesis, and are needed for the attenuation of mitochondrial oxidative stress, regulation of cellular and organismal metabolism, and antimicrobial immunity [101,106,107,108,109,110,111,112].
The mechanisms of UCP-dependent proton transport have been studied extensively in the context of UCP1. These studies have revealed that UCPs are dimers of 6 transmembrane domain-containing proteins [102,113]. At the core of the dimer is a hydrophilic pore [102]. UCP-mediated proton transport requires free fatty acids, which are purported to function via one of two possible mechanisms [101,114]. The “proton buffering model” argues that fatty acids transfer protons to proton-buffering amino acids in the pore, which then shuttle the ions across the membrane [101]. Alternatively, the “fatty acid cycling model” contends that fatty acid anions accept protons in the intermembrane space and transport protons across the membrane by directly traversing the pore [101].

Role of UCP Proteins in the Response to Oxidative Stress

ROS—well-established byproducts of mitochondrial metabolism—can damage DNA, proteins, and lipids, and are therefore drivers of senescence. Because the mitochondrial protonmotive force is positively correlated with superoxide production [101,115], activation of UCP2 and UCP3 mitigates cellular oxidative stress, and thereby, counters the onset of senescence [101,102,107,108,109,110]. Conversely, loss of either UCP2 or UCP3 is associated with increased levels of mitochondrial ROS [109,110,116,117].
There are clearly some benefits associated with increased ROS in the absence of UCPs. Elevated ROS production in macrophages isolated from Ucp2−/− mice greatly restricts growth of Toxoplasma gondii, making the animals resistant to death from Toxoplasma infection [109]. More generally, however, ROS promotes senescence and aging [84,85,102,118,119,120,121,122]. Therefore, higher levels of proton leak via UCP2 or UCP3, which mitigate oxidative stress, are correlated with longer lifespans in mice [123,124]. Ectopic overexpression of human UCP2 or UCP3 in fly and/or mouse neurons is sufficient to prolong the animals’ lifespans, and confer greater resistance to ROS [120,125,126,127]. These beneficial effects depend on the induction of pro-longevity genes in hypothalamic neurons of mice, or in insulin-producing fly neurons, which are reminiscent of hypothalamic neurons [125,126,127]. Given the role for the hypothalamus in the regulation of feeding and energy homeostasis, these findings raise the intriguing possibility that UCP activity regulates lifespan by influencing the animals’ feeding behavior. Indeed, caloric restriction in mice—a reliable pro-longevity factor—augments the expression of Ucp2 and Ucp3, promotes mitochondrial proton leak, and lowers ROS production [128,129]. The convergence of multiple longevity-related pathways on UCP2 and UCP3 speaks to the pro-survival roles of uncoupling, and supports the “uncoupling to survive” model [123,130].
Although the predominant role for UCP1 is thermogenesis in brown adipose tissue, abundance of this protein does correlate with lower incidence of age-related disease [131]. In humans, polymorphisms in the regulatory regions of UCP1 that result in greater expression of the gene are associated with improved longevity [132]. Overexpression of Ucp1 in mice is sufficient to extend lifespan by lowering the incidence of cancer and atherosclerotic lesions, and by correcting preexisting metabolic dysfunction [131]. Conversely, Ucp1−/− mice exhibit greater susceptibility to obesity at advanced age when reared on a high-fat diet (HFD) [133]. Given the adverse effects of obesity on lifespan, these findings point to a role for UCP1 in mitigating the consequences of unhealthy diet on age-related decline of healthspan.

5. Regulation of Lifespan by Ion Channels Involved in Autophagy and Lysosomal Proteostasis

Autophagy and lysosomal protein degradation constitute major axes of proteostasis in metazoans. By coordinating the removal of toxic macromolecules and damaged organelles, these processes counteract the stochastic accumulation of cellular debris that otherwise promote aging [134,135,136]. Therefore, hypermethylation of autophagy and lysosomal genes tends to diminish protein degradation in aged organisms [137], whereas upregulation of autophagy and lysosomal function improves proteostasis and extends healthy lifespan [134]. Induction of autophagy is sufficient to confer resistance to oxidative stress and insulin sensitivity in mice reared on a high-fat diet, and ameliorate the toxic consequences of polyglutamine expansion in mouse and fly models [138,139]. Ubiquitous overexpression of Atg5, a gene that encodes a protein needed for autophagosome formation, extends mouse lifespan via the augmentation of leanness, insulin-sensitivity, and oxidative stress tolerance [140]. Even the extension of lifespan brought about by diminished insulin-signaling in Drosophila or dietary restriction in C. elegans are dependent on autophagy and lysosomal activity [141,142,143]. Genetic upregulation of chaperone-mediated autophagy (CMA), which involves LAMP-2A-dependent direct lysosomal targeting of select proteins, also enhances proteostasis, mitigates oxidative stress, and preserves organ function in aged mammals [144,145]. Conversely, the repression of autophagy by deletion of Atg5 or Atg7 in mice and flies promotes aging, neurodegeneration, and shorter lifespan via dysregulation of proteostasis and mitochondrial function [146,147,148,149,150,151].

5.1. Involvement of Vesicular Ion Channels in Endolysosomal Function and Lifespan

Autophagic protein degradation is comprised of a series of vesicular trafficking events that originate with the sequestration of cargo into autophagosomes. These double membrane-bound organelles then fuse with endolysosomal vesicles in order to permit hydrolytic enzymes to gain access to material destined for degradation [152]. In Drosophila, hybrid organelles formed upon the fusion of late-endosomes and autophagosomes (amphisomes) fuse with lysosomes in a Ca2+-dependent process requiring the endolysosomal cation channel, TRPML [153,154]. Loss of trpml, therefore, prevents the fusion of amphisomes with lysosomes, which leads to the accumulation of amphisomes and diminished endolysosomal degradation [153,154]. These phenotypes also characterize Mucolipidosis type IV (MLIV)—a lysosomal storage disease that stems from the loss of MCOLN1, the gene encoding the human ortholog [155,156,157,158].
In flies, diminished amphisome–lysosome fusion in the absence of functional TRPML leads to decreased production of amino acids that are generated from endolysosomal protein degradation [153,154]. Given the roles for endolysosomal amino acids in the activation of the mTORC1 kinase complex [159], loss of TRPML results in diminished mTORC1 activation—a phenotype that can be suppressed by dietary administration of a high-protein diet [153,154]. The role for TRPML1 in mTORC1 activation is also conserved in mammals [160,161,162] (Figure 6). Interestingly, the inverse relationship between mTORC1 activity and endolysosome/autophagosome biogenesis (via TFEB, see below) ensures that loss of TRPML results in upregulation of endolysosomal biogenesis, which may explain the accumulation of lysosomes in MLIV [153,163,164,165].
Based on the aforementioned insights, one can appreciate that the regulation of aging and lifespan by the TRPML family of endolysosomal ion channels is complex. On the one hand, loss of TRPMLs leads to the accumulation of undigested endosomal material, whereas their transcriptional upregulation promotes the exocytosis and clearance of toxic macromolecules [166]. In this regard, TRPMLs are necessary for proteostasis. Indeed, the loss of mouse TRPML1 or fly TRPML leads to proteostatic imbalance, severe loss of neuronal function, and abbreviated lifespans [167,168]. It is worth noting that besides inhibiting the mTORC1 complex, the FDA-approved, longevity-promoting drug, rapamycin [169] also activates TRPML1 independently of mTORC1 [170]. Consequently, rapamycin-induced autophagic flux is attenuated in TRPML1-deficient cells [170]. It is, therefore, possible that some of the pro-longevity effects of rapamycin stem from the activation of TRPML1 and the attendant improvement of cellular proteostasis.
On the other hand, since decreased mTORC1 activity is associated with the extension of lifespan in a variety of organisms [171,172,173,174,175], attenuation of mTORC1 activity in the absence of TRPMLs also have beneficial consequences. In agreement, abbreviation of lifespan upon the expression of neurodegeneration-causing transgenes in fly neurons is suppressed by the concomitant knockdown of trpml [12]. Additionally, TRPML1 contributes to the toxicity in a presenilin knockout mouse model of Alzheimer’s disease [176]. Delineating the relative roles of TRPMLs in the regulation of lifespan under physiologically normal or pathological conditions would likely require further investigation.
Two-pore channels (TPCs) are a class of endolysosomal cation channels that orchestrate vesicular trafficking events by releasing vesicular Ca2+ and Na+ in response to NAADP and PI(3,5)P2 [177,178,179,180,181,182,183,184]. TPC function is of relevance to autophagy, mTORC1 activity, protein homeostasis, and cholesterol homeostasis [185,186,187,188] (Figure 6). Not surprisingly, deletion of the gene encoding TPC2 in mice results in skeletal muscle atrophy due to defective lysosomal proteolysis, accumulation of undigested autophagic vacuoles, and heightened sensitivity to starvation [186]. Similar roles for TPCs in ensuring autophagic and endolysosomal flux has also been reported in cardiac muscle [185]. Elevated TPC2 activity is observed in cells expressing Parkinson’s disease associated mutations in LRRK2 [189], suggesting that attenuation of TPC2 activity (as is the case of TRPML1) might be beneficial in late-onset neurodegenerative diseases.
Cl represents the major endolysosomal counter ion. ClC-6 and ClC-7 and endolysosomal chloride transporters are required for normal lysosomal function [190,191,192]. Loss of these transporters result in a variety of neuropathological alterations, including lysosomal storage, progressive neuron loss, and microglial activation [193,194,195]. The progressive nature of these phenotypes suggests a role for these proteins in the maintenance of neuronal function and survival at advanced age.
Critical to the regulation of amino acid availability and mTORC1 activity are vesicular solute carriers belonging to the SLC family of transporters. Among the many amino acid transporters critical for mTORC1 activation is the lysosomal amino acid transporter, SLC38A9 [196,197,198]. Loss of SLC38A9 decouples mTORC1 from amino acids such as arginine, whereas overexpression of SLC38A9 results in the constitutive activation of mTORC1, even in the absence of amino acids [196,197,198]. Therefore, SLC38A9 is not only a vesicular transporter needed for the efflux of amino acids from the lysosome, but is also an amino acid sensor that synchronizes mTORC1 activity to amino acid abundance [196,197,198,199]. Besides serving as a sensor and transporter for amino acids, SLC38A9 also harbors cholesterol-binding motifs [200]. Association of cholesterol with SLC38A9 activates mTORC1 independently of the transporter’s role in endolysosomal amino acid efflux [200]. Other amino acid transporters linked to the regulation of mTORC1 include SLC38A5, SLC7A5 (also called LAT1) and the glutamine transporter SLC1A5 (also called ASCT2) [201,202,203,204,205]. Consistent with the roles for mTORC1 in aging [171,172,173,174,175], emerging evidence suggests that modulation of the activity of amino acid sensors, such as the ones described here, could be an effective strategy to influence rates of aging [206]. Indeed, polymorphisms in genes encoding amino acid transporters are associated with age-related changes in physical performance (e.g., grip strength and walking speed) [207]. Conversely, there is increased abundance of amino acid transporters such as SLC7A5, SLC1A5, and SLC38A5 in inflammation and cancer [204,205,206,208]. Together, these studies point to critical roles for amino acid transporters in the regulation of aging and longevity.

5.2. Transcriptional Regulation of Endolysosomal Function

A major advancement in the field of lysosomal biology was the identification of transcription factors (TFEB, TFE3, and MITF) that function as master regulators of autophagy and endolysosomal biogenesis [163,164,165,209,210,211,212,213,214]. Genetic or pharmacological activation of these transcription factors promotes the clearance of cellular debris and aggregate-prone toxic macromolecules [166,215,216,217]. As such, TFEB maintains the quiescent state of neural stems cells, and has been positively correlated with longevity in C. elegans and mice [218,219,220,221,222,223,224]. TFEB and TFE3 are needed for the maintenance of whole-body metabolism in response to changes in various environmental stimuli including diet [225]. Dietary restriction—an established mode of lifespan extension—promotes TFEB-dependent gene expression in mouse hepatocytes [222].
Many genes that encode endolysosomal ion channels, including TRPML1 and TPC2, are under the control of TFEB [211,212,226,227] (Figure 6). In turn, nuclear translocation of TFEB, which is dependent on the phosphorylation status of the protein, is regulated by TRPML1 and TPC2 (Figure 6). On the one hand, TRPML1- and TPC2-dependent activation of mTORC1 ensures TFEB phosphorylation and cytosolic retention [160,161,162,163,164] (Figure 6). These findings explain why the lack of TRPML1, results in nuclear translocation of TFEB and unremitting endolysosomal biogenesis. On the other hand, endolysosomal Ca2+ release can also dephosphorylate and promote nuclear translocation of TFEB via the protein phosphatase, calcineurin (CaN) [228] (Figure 6). This model portends a feed-forward cycle, whereby TRPML1 and TPC2 activation, which are under TFEB transcriptional control, promotes further TFEB-dependent endolysosomal biogenesis.
While TFEB-driven autophagy and endolysosomal biogenesis delays aging via the clearance of toxic macromolecules, unremitting activation of TFEB can paradoxically shorten lifespan [141]. Either the overexpression and/or constitutive nuclear localization of TFEB and TFE3 promote the growth of various types of cancer [226,227,229,230,231,232,233,234,235,236,237,238,239,240]. Additionally, amplification of the MITF locus and functional upregulation of MITF are potent drivers of melanoma [213,241,242,243,244,245,246]. These findings point to the importance of maintaining autophagy and endolysosomal function at an optimal level because deviation from this optimum in either direction has deleterious consequences to healthy lifespan. In cancers driven by TFEB/TFE3/MITF, expression of genes encoding endolysosomal cation channels, TRPML1, TRPML2 and TPC2, is elevated, and either the knockdown or pharmacological inhibition of these channels attenuates the growth and invasiveness of the tumors [160,226,227,247,248,249,250,251,252]. Taken together, these studies point to the highly context-dependent relationship between endolysosomal ion channels and healthspan.

6. Regulation of Ion Channel Activity by the Longevity Factor, Klotho

Mutations in the mouse klotho gene results in animals that develop normally, but prematurely exhibit several features of accelerated aging including neurodegeneration, atherosclerosis, osteoporosis, infertility, atrophy of the skin and other organs, and shorter lifespan [253,254]. Conversely, overexpression of klotho results in a 20–30% extension of mouse lifespan via the repression of insulin signaling and the amelioration of oxidative stress [255,256]. Overexpressed klotho also enhances cognition by the enhancement of synaptic plasticity [257]. These roles for Klotho in healthy aging are also observed in humans [258,259,260].
The product of the klotho gene is a type-I single pass transmembrane protein that localizes to the cell surface [256]. The extracellular domain of the protein exhibits homology to glycosidases/sialidases that can cleave the b-glycosidic linkage in sugars, glycoproteins and glycolipids, and remove sialic acid residues from membrane proteins [253,256,261,262,263]. Although expression of klotho is highest in the kidneys, the extracellular sialidase domain of the protein product is cleaved by ADAM10 and ADAM17 transmembrane proteases, and released into the bloodstream where it can function as an endocrine factor [256,261,262,264]. ADAM10/ADAM17-dependent cleavage of membrane-bound Klotho is under the control of insulin signaling, which when taken in consideration that soluble Klotho represses insulin signaling, points to the existence of a feedback loop to prevent unremitting insulin signaling [255,264]. While the membrane-bound form of Klotho serves as a coreceptor for fibroblast growth factor-23 (FGF23) and regulates phosphate and vitamin D metabolism, the secreted form neither binds to, nor serves as a coreceptor for FGF23 [256,265]. Rather, soluble Klotho serves as an enzyme capable of removing sugars and sialic acid residues from various membrane glycoproteins [253,256,261,262,263]. It is via the influence on membrane glycoproteins that soluble Klotho prolongs longevity.

Regulation of Ion Channel Activity by Klotho

By removing glycan moieties from the extracellular side of the plasma membrane, soluble Klotho promotes the surface retention and activities of TRPV5 and TRPV6 ion channels [263,266,267,268,269]. Removal of terminal sialic acid residues allows the remaining sugars to bind lectins leading to channel crosslinking on the cell surface and diminished internalization [263]. Given that TRPV5 participates in systemic Ca2+ and phosphate homeostasis by functioning in the kidney [270], it is not clear whether Klotho influences longevity via these channels. Nevertheless, it is possible that hyperphosphatemia and hypercalcemia resulting from diminished TRPV5/TRPV6 function in the absence of Klotho could contribute to age-dependent vascular calcification and osteopenia [271]. A putative link to longevity can be gleaned from the demonstration that Klotho promotes the surface retention and overall activity of the Na+/K+ ATPase [272]. Given the relationship between neuronal excitability and longevity, and demonstrated roles for the Na+/K+ ATPase in the regulation of these parameters [10,11,12,16], soluble Klotho could influence lifespan by stabilizing this ATPase at the cell surface.
Alternatively, Klotho could influence animal longevity by its influence on cardiac function. Klotho counteracts cardiac arrhythmia by ensuring the cell surface expression of KCNQ1/KCNE1 K+ channels that are involved in cardiac repolarization [273]. Loss of soluble Klotho promotes cardiac arrhythmia owing to decreased activity of the KCNQ1/KCNE1 channels in cardiomyocytes. Cardioprotective roles of Klotho also stem from downregulation of TRPC6 channel activity [274,275]. TRPC6 channels have vital roles in stress-induced pathological cardiac hypertrophy and remodeling [274]. By attenuating TRPC6 conductance, Klotho ameliorates cardiotoxicity [274,276].

7. Roles for Ion Channels in Regulation of Longevity by Temperature

All physiological systems are under the control of temperature. While an increase in temperature accelerates the rates of biological reactions, a decrease in temperature has the opposite effect [277]. The inverse relationship between rates of metabolism and longevity is why lower body temperatures are associated with longer lifespans, whereas higher body temperatures are more common in short-lived individuals [278,279,280]. In homeotherms, interventions that influence rates of aging and lifespan, for instance caloric restriction, also tend to alter body temperature in a manner that is consistent between the aforementioned relationship between temperature and longevity [278,281]. Cold temperatures in homeotherms lead to the generation of heat in brown adipose tissue via UCP1 [101,102,103,104,105]. Given the involvement of UCP proteins in regulation of longevity (as discussed above), cold-induced uncoupling could constitute another mechanism by which low temperatures promote longevity. In agreement, overexpression of Ucp2 in the hypocretin neurons of mouse hypothalamus extended the animals’ longevity by lowering their core body temperature [125].
In poikilotherms such as C. elegans and Drosophila, a decrease in ambient temperature promotes stress resistance and counteracts inflammatory signaling [282]. While it is tempting to speculate that the relationship between temperature and age-related pathology stems from the propensity of temperature to increase thermodynamic entropy [278,283], it is likely that additional levels of complexity bear upon this relationship. Indeed, flies cycled between hot and cold ambient temperatures live for as long as those that are reared at steady low ambient temperature [278,279,284]. These intriguing data suggested that the process of sensing a drop in ambient temperature is sufficient to impart the benefits of low temperature [278].

7.1. Thermosensitive Channels and Lifespan

In agreement with the notion that the act of sensing ambient temperature influences longevity, recent studies have shown that thermosensitive ion channels actuate the effects of temperature on lifespan. A pioneering study in C. elegans challenged the passive thermodynamic model of aging by demonstrating that a cold-sensitive TRP channel (TRPA-1) detects a drop in environmental temperature, and signals to the well-established modulator of longevity, DAF-16/FOXO [5,285,286] (Figure 7). trpa-1 deficient adult worms exhibited significantly shorter lifespans when reared at a cool 15 °C, whereas no such difference between wild-type and trpa-1 mutants was observed in animals raised at 25 °C [285,286]. These findings suggest that cold-induced extension of lifespan involves an ion channel that is directly activated by lower temperature rather than a general reduction in rates of metabolism (Figure 7).
Mice lacking the thermosensitive TRP channel, TRPV1, exhibit youthful metabolism and are relatively long-lived [287]. The regulation of lifespan via TRPV1 involves nuclear localization of the CREB-regulated transcriptional coactivator, CRTC1. In animals lacking TRPV1, CRTC1 is excluded from the nucleus, which results in diminished production of the neuropeptide CGRP and attendant improvement of metabolic health [287]. The relationship between CGRP and systemic metabolism is ensured by the repression of insulin release by CGRP [287]. In C. elegans, deletion of the TRPV channel genes, ocr-2 and osm-9, led to the extension of lifespan [287,288]. Whether the activities of TRPV1, OSM-9, and OCR-2 in these contexts involve temperature remain to be addressed.

7.2. Heat-shock Response and Lifespan

While low temperature is associated with the extension of lifespan in many species, transient heat shocks in poikilotherms induce the expression of molecular chaperones that enforce proteostasis and promote longevity [289,290,291]. Interestingly, longevity-promoting effects of the heat-shock response in C. elegans are not cell autonomous, but rather stem from thermosensitive neurons that express cyclic nucleotide-gated (CNG) TAX-2/TAX-4 channels [292]. Deletion of guanylyl cyclase responsible for the activation of these CNG channels in thermosensitive neurons prevents the induction of a heat-shock response in other tissues [292]. The underlying mechanism involves the secretion of FMRFamide neuropeptide from the thermosensitive neurons, which greatly influences peripheral insulin signaling [293]. Furthermore, induction of the C. elegans heat shock factor, HSF-1, and the attendant effects on proteostasis can occur independently of a bona fide heat shock stimulus. Cholinergic neurotransmission at the neuromuscular junction leads to a Ca2+-dependent induction of the hsf-1 in postsynaptic muscle cells via a process requiring the VGCCs and ryanodine receptors in the ER [294].

8. Regulation of Longevity by Channels Involved in Other Sensory Modalities

Besides temperature, many sensory modalities can modulate lifespan. In Drosophila, critical insights were obtained from the demonstration that deletion of the gene encoding a water-sensitive channel belonging to the amiloride-sensitive ENaC family of Na+ channels, pickpocket 28 (ppk28), altered the metabolic status of animals, and thereby, extended lifespan [295,296,297]. Loss of ppk28 improved systemic metabolism via the augmentation of neuronal signaling involving glucagon-related, adipokinetic hormone (AKH) [295]. Remarkably, the apparent lack of water—mimicked by the deletion of ppk28—triggers AKH-dependent alterations in metabolic pathways such as b-oxidation that produce molecular water, and in the process, extends lifespan [295].
In C. elegans, activity of sensory neurons, which is partially dependent on the ENaC channel MEC-4, is necessary for the protection of mitochondria from fragmentation in aged animals [298]. Furthermore, the TRPV channel, OSM-9, functions in sensory neurons to mediate the avoidance of hypertonic stress [299,300]. Deletion of osm-9 led to enhanced proteostasis and survival of the worms when placed in hypertonic stress [300]. Deletion of another TRPV channel gene in C. elegans, ocr-2, also extended adult lifespan [287,288]. In Drosophila, loss of a similar TRPV channel, Inactive (Iav), which has been proposed to play roles in mechanosensation, is associated with reduced lifespan [301,302].
Relationships between lifespan and reproduction and/or food availability have been observed across taxa. In flies, detection of female pheromones via gustatory sensory neurons leads to reduction in stored nutrient reserves, greater sensitivity to starvation, and shorter lifespan in males [303]. This deleterious effect of female pheromone sensation on male longevity was observed only if the males were not allowed to mate, and mating reversed the effects of pheromone sensation on longevity. These findings indicate that the relationship sensory and reward circuits in the brain modulates aging and lifespan [303]. Although the involvement of ion channels in this axis of longevity have not been described, indirect evidence suggests the involvement of ENaC channels. Deletion of ppk23 and ppk25 result in diminished intensities of male courtship, likely due to altered responses to female pheromones [304,305]. It would be worth evaluating whether the absences of ppk23 and ppk25 influence the lifespan of the animals in a pheromone- and courtship-dependent manner. As was the case with female pheromones regulating male lifespan in fruit flies, food-derived odors shorten fly lifespan, and even deter the longevity-promoting effects of dietary restriction [306]. Therefore, deletion of the gene encoding, Or83b, an ionotropic odorant receptor that functions as a non-selective cation channel needed for the detection of food-derived odors, augmented stress resistance and extended animal lifespan [306,307,308].

9. Closing Remarks and Future Directions

Despite the plethora of ion channels and transporters that influence aging and longevity, holistic evaluation of the studies described here also reveal commonalities. One general principle is that mechanisms of ion transport constitute major regulatory axes in pathways that determine the rates of cellular or organismal aging. Some channels even function at the intersection of one or more hallmarks of aging. A better understanding of how these channels and transporters impact age-related loss of biological integrity could aid in the development of effective anti-aging strategies. Another theme that emerges from these studies is that homeostatic regulation of ion channel and transporter activity is the key to human healthspan. Any deviation of ion transport from a healthy optimum leads to elevated rates of mortality. Consequently, the trajectory of age-dependent loss of biological function likely varies greatly from one individual to the next. What we need, therefore, is the development of strategies that would enable personalized approaches to combat aging. A suite of drugs that restores ionic homeostasis in various tissues by appropriately modifying channel and transporter activities could be the key to translate biological insights into actionable therapeutic strategies. Future studies into the regulation of aging and longevity by ion channels and transporters will undoubtedly bring us closer to realizing this vision.

Funding

Work in the Venkatachalam lab is supported by NIH grants RF1AG069076, RF1AG067414, and R21AG072176 (all to K.V.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The author wishes to thank Dr. Richa Gupta and Dr. Elham Rastegari for reading the manuscript and providing valuable feedback.

Conflicts of Interest

The author declares no competing interest.

References

  1. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Campisi, J.; Kapahi, P.; Lithgow, G.J.; Melov, S.; Newman, J.C.; Verdin, E. From Discoveries in Ageing Research to Therapeutics for Healthy Ageing. Nature 2019, 571, 183–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kenyon, C.; Chang, J.; Gensch, E.; Rudner, A.; Tabtiang, R. A C. elegans Mutant That Lives Twice as Long as Wild Type. Nature 1993, 366, 461–464. [Google Scholar] [CrossRef]
  4. Friedman, D.B.; Johnson, T.E. A Mutation in the Age-1 Gene in Caenorhabditis Elegans Lengthens Life and Reduces Hermaphrodite Fertility. Genetics 1988, 118, 75–86. [Google Scholar] [CrossRef] [PubMed]
  5. Ogg, S.; Paradis, S.; Gottlieb, S.; Patterson, G.I.; Lee, L.; Tissenbaum, H.A.; Ruvkun, G. The Fork Head Transcription Factor DAF-16 Transduces Insulin-like Metabolic and Longevity Signals in C. elegans. Nature 1997, 389, 994–999. [Google Scholar] [CrossRef]
  6. Tatar, M.; Kopelman, A.; Epstein, D.; Tu, M.P.; Yin, C.M.; Garofalo, R.S. A Mutant Drosophila Insulin Receptor Homolog That Extends Life-Span and Impairs Neuroendocrine Function. Science 2001, 292, 107–110. [Google Scholar] [CrossRef] [Green Version]
  7. Berridge, M.J. Calcium Signalling Remodelling and Disease. Biochem. Soc. Trans. 2012, 40, 297–309. [Google Scholar] [CrossRef] [Green Version]
  8. Karagas, N.E.; Venkatachalam, K. Roles for the Endoplasmic Reticulum in Regulation of Neuronal Calcium Homeostasis. Cells 2019, 8, 1232. [Google Scholar] [CrossRef] [Green Version]
  9. Berridge, M.J. Inositol Trisphosphate and Calcium Signalling Mechanisms. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2009, 1793, 933–940. [Google Scholar] [CrossRef] [Green Version]
  10. Zullo, J.M.; Drake, D.; Aron, L.; O’Hern, P.; Dhamne, S.C.; Davidsohn, N.; Mao, C.-A.; Klein, W.H.; Rotenberg, A.; Bennett, D.A.; et al. Regulation of Lifespan by Neural Excitation and REST. Nature 2019, 574, 359–364. [Google Scholar] [CrossRef]
  11. Fergestad, T.; Ganetzky, B.; Palladino, M.J. Neuropathology in Drosophila Membrane Excitability Mutants. Genetics 2006, 172, 1031. [Google Scholar] [CrossRef] [PubMed]
  12. Wong, C.-O.; Karagas, N.E.; Jung, J.; Wang, Q.; Rousseau, M.A.; Chao, Y.; Insolera, R.; Soppina, P.; Collins, C.A.; Zhou, Y.; et al. Regulation of Longevity by Depolarization-Induced Activation of PLC-β-IP3R Signaling in Neurons. Proc. Natl. Acad. Sci. USA 2021, 118, e2004253118. [Google Scholar] [CrossRef]
  13. Reynolds, E.R. Shortened Lifespan and Other Age-Related Defects in Bang Sensitive Mutants of Drosophila Melanogaster. G3 Genes Genomes Genet. 2018, 8, 3953–3960. [Google Scholar] [CrossRef] [Green Version]
  14. Garber, G.; Smith, L.A.; Reenan, R.A.; Rogina, B. Effect of Sodium Channel Abundance on Drosophila Development, Reproductive Capacity and Aging. Fly 2012, 6, 57–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cirelli, C.; Bushey, D.; Hill, S.; Huber, R.; Kreber, R.; Ganetzky, B.; Tononi, G. Reduced Sleep in Drosophila Shaker Mutants. Nature 2005, 434, 1087–1092. [Google Scholar] [CrossRef]
  16. Palladino, M.J.; Bower, J.E.; Kreber, R.; Ganetzky, B. Neural Dysfunction and Neurodegeneration in Drosophila Na+/K+ ATPase Alpha Subunit Mutants. J. Neurosci. Off. J. Soc. Neurosci. 2003, 23, 1276–1286. [Google Scholar] [CrossRef] [Green Version]
  17. Andries, M.; van Damme, P.; Robberecht, W.; van den Bosch, L. Ivermectin Inhibits AMPA Receptor-Mediated Excitotoxicity in Cultured Motor Neurons and Extends the Life Span of a Transgenic Mouse Model of Amyotrophic Lateral Sclerosis. Neurobiol. Dis. 2007, 25, 8–16. [Google Scholar] [CrossRef] [PubMed]
  18. Warnier, M.; Flaman, J.-M.; Chouabe, C.; Wiel, C.; Gras, B.; Griveau, A.; Blanc, E.; Foy, J.-P.; Mathot, P.; Saintigny, P.; et al. The SCN9A Channel and Plasma Membrane Depolarization Promote Cellular Senescence through Rb Pathway. Aging Cell 2018, 17, e12736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Liu, W.; Lin, H.; Mao, Z.; Zhang, L.; Bao, K.; Jiang, B.; Xia, C.; Li, W.; Hu, Z.; Li, J. Verapamil Extends Lifespan in Caenorhabditis Elegans by Inhibiting Calcineurin Activity and Promoting Autophagy. Aging 2020, 12, 5300–5317. [Google Scholar] [CrossRef]
  20. Sutphin, G.L.; Backer, G.; Sheehan, S.; Bean, S.; Corban, C.; Liu, T.; Peters, M.J.; van Meurs, J.B.J.; Murabito, J.M.; Johnson, A.D.; et al. Caenorhabditis Elegans Orthologs of Human Genes Differentially Expressed with Age Are Enriched for Determinants of Longevity. Aging Cell 2017, 16, 672–682. [Google Scholar] [CrossRef] [Green Version]
  21. Moore, S.J.; Murphy, G.G. The Role of L-Type Calcium Channels in Neuronal Excitability and Aging. Neurobiol. Learn. Mem. 2020, 173, 107230. [Google Scholar] [CrossRef] [PubMed]
  22. Zhang, X.; Chen, S.; Lu, K.; Wang, F.; Deng, J.; Xu, Z.; Wang, X.; Zhou, Q.; Le, W.; Zhao, Y. Verapamil Ameliorates Motor Neuron Degeneration and Improves Lifespan in the SOD1 G93A Mouse Model of ALS by Enhancing Autophagic Flux. Aging Dis. 2019, 10, 1159–1173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Huntula, S.; Saegusa, H.; Wang, X.; Zong, S.; Tanabe, T. Involvement of N-Type Ca2+ Channel in Microglial Activation and Its Implications to Aging-Induced Exaggerated Cytokine Response. Cell Calcium 2019, 82, 102059. [Google Scholar] [CrossRef] [PubMed]
  24. Hayashi, T.; Yamaguchi, T.; Sakakibara, Y.; Taguchi, K.; Maeda, M.; Kuzuya, M.; Hattori, Y. ENOS-Dependent Antisenscence Effect of a Calcium Channel Blocker in Human Endothelial Cells. PLoS ONE 2014, 9, e88391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Li, Z.; Guo, G.; Wang, H.; Si, X.; Zhou, G.; Xiong, Y.; Li, S.; Dai, R.; Yang, C. TRPC5 Channel Modulates Endothelial Cells Senescence. Eur. J. Pharmacol. 2017, 802, 27–35. [Google Scholar] [CrossRef] [PubMed]
  26. Belrose, J.C.; Xie, Y.-F.; Gierszewski, L.J.; MacDonald, J.F.; Jackson, M.F. Loss of Glutathione Homeostasis Associated with Neuronal Senescence Facilitates TRPM2 Channel Activation in Cultured Hippocampal Pyramidal Neurons. Mol. Brain 2012, 5, 11. [Google Scholar] [CrossRef] [Green Version]
  27. Yee, N.S.; Brown, R.D.; Lee, M.S.; Zhou, W.; Jensen, C.; Gerke, H.; Yee, R.K. TRPM8 Ion Channel Is Aberrantly Expressed and Required for Preventing Replicative Senescence in Pancreatic Adenocarcinoma: Potential Role of TRPM8 as a Biomarker and Target. Cancer Biol. Ther. 2012, 13, 592–599. [Google Scholar] [CrossRef] [Green Version]
  28. Yee, N.S.; Zhou, W.; Lee, M. Transient Receptor Potential Channel TRPM8 Is Over-Expressed and Required for Cellular Proliferation in Pancreatic Adenocarcinoma. Cancer Lett. 2010, 297, 49–55. [Google Scholar] [CrossRef] [Green Version]
  29. Yee, N.S.; Zhou, W.; Lee, M.; Yee, R.K. Targeted Silencing of TRPM7 Ion Channel Induces Replicative Senescence and Produces Enhanced Cytotoxicity with Gemcitabine in Pancreatic Adenocarcinoma. Cancer Lett. 2012, 318, 99–105. [Google Scholar] [CrossRef] [Green Version]
  30. Szydlowska, K.; Tymianski, M. Calcium, Ischemia and Excitotoxicity. Cell Calcium 2010, 47, 122–129. [Google Scholar] [CrossRef]
  31. Iizuka, A.; Nakamura, K.; Hirai, H. Long-Term Oral Administration of the NMDA Receptor Antagonist Memantine Extends Life Span in Spinocerebellar Ataxia Type 1 Knock-in Mice. Neurosci. Lett. 2015, 592, 37–41. [Google Scholar] [CrossRef] [PubMed]
  32. Xu, W.; Xu, J. C9orf72 Dipeptide Repeats Cause Selective Neurodegeneration and Cell-Autonomous Excitotoxicity in Drosophila Glutamatergic Neurons. J. Neurosci. Off. J. Soc. Neurosci. 2018, 38, 7741–7752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Wenk, G.L.; Barnes, C.A. Regional Changes in the Hippocampal Density of AMPA and NMDA Receptors across the Lifespan of the Rat. Brain Res. 2000, 885, 1–5. [Google Scholar] [CrossRef]
  34. Adams, M.M.; Shi, L.; Linville, M.C.; Forbes, M.E.; Long, A.B.; Bennett, C.; Newton, I.G.; Carter, C.S.; Sonntag, W.E.; Riddle, D.R.; et al. Caloric Restriction and Age Affect Synaptic Proteins in Hippocampal CA3 and Spatial Learning Ability. Exp. Neurol. 2008, 211, 141–149. [Google Scholar] [CrossRef] [Green Version]
  35. Dubal, D.B.; Zhu, L.; Sanchez, P.E.; Worden, K.; Broestl, L.; Johnson, E.; Ho, K.; Yu, G.Q.; Kim, D.; Betourne, A.; et al. Life Extension Factor Klotho Prevents Mortality and Enhances Cognition in HAPP Transgenic Mice. J. Neurosci. Off. J. Soc. Neurosci. 2015, 35, 2358–2371. [Google Scholar] [CrossRef]
  36. Papadia, S.; Soriano, F.X.; Léveillé, F.; Martel, M.A.; Dakin, K.A.; Hansen, H.H.; Kaindl, A.; Sifringer, M.; Fowler, J.; Stefovska, V.; et al. Synaptic NMDA Receptor Activity Boosts Intrinsic Antioxidant Defenses. Nat. Neurosci. 2008, 11, 476–487. [Google Scholar] [CrossRef] [PubMed]
  37. Majumder, S.; Caccamo, A.; Medina, D.X.; Benavides, A.D.; Javors, M.A.; Kraig, E.; Strong, R.; Richardson, A.; Oddo, S. Lifelong Rapamycin Administration Ameliorates Age-Dependent Cognitive Deficits by Reducing IL-1β and Enhancing NMDA Signaling. Aging Cell 2012, 11, 326–335. [Google Scholar] [CrossRef] [Green Version]
  38. Shi, L.; Adams, M.M.; Linville, M.C.; Newton, I.G.; Forbes, M.E.; Long, A.B.; Riddle, D.R.; Brunso-Bechtold, J.K. Caloric Restriction Eliminates the Aging-Related Decline in NMDA and AMPA Receptor Subunits in the Rat Hippocampus and Induces Homeostasis. Exp. Neurol. 2007, 206, 70–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Yilmaz, N.; Vural, H.; Yilmaz, M.; Sutcu, R.; Sirmali, R.; Hicyilmaz, H.; Delibas, N. Calorie Restriction Modulates Hippocampal NMDA Receptors in Diet-Induced Obese Rats. J. Recept. Signal Transduct. Res. 2011, 31, 214–219. [Google Scholar] [CrossRef]
  40. Woll, K.A.; van Petegem, F. Calcium-Release Channels: Structure and Function of IP 3 Receptors and Ryanodine Receptors. Physiol. Rev. 2022, 102, 209–268. [Google Scholar] [CrossRef]
  41. Van de Leemput, J.; Chandran, J.; Knight, M.A.; Holtzclaw, L.A.; Scholz, S.; Cookson, M.R.; Houlden, H.; Gwinn-Hardy, K.; Fung, H.-C.; Lin, X.; et al. Deletion at ITPR1 Underlies Ataxia in Mice and Spinocerebellar Ataxia 15 in Humans. PLoS Genet. 2007, 3, e108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Klar, J.; Ali, Z.; Farooq, M.; Khan, K.; Wikström, J.; Iqbal, M.; Zulfiqar, S.; Faryal, S.; Baig, S.M.; Dahl, N. A Missense Variant in ITPR1 Provides Evidence for Autosomal Recessive SCA29 with Asymptomatic Cerebellar Hypoplasia in Carriers. Eur. J. Hum. Genet. 2017, 25, 848–853. [Google Scholar] [CrossRef] [Green Version]
  43. Sasaki, M.; Ohba, C.; Iai, M.; Hirabayashi, S.; Osaka, H.; Hiraide, T.; Saitsu, H.; Matsumoto, N. Sporadic Infantile-Onset Spinocerebellar Ataxia Caused by Missense Mutations of the Inositol 1,4,5-Triphosphate Receptor Type 1 Gene. J. Neurol. 2015, 262, 1278–1284. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, L.; Chardon, J.W.; Carter, M.T.; Friend, K.L.; Dudding, T.E.; Schwartzentruber, J.; Zou, R.; Schofield, P.W.; Douglas, S.; Bulman, D.E.; et al. Missense Mutations in ITPR1 Cause Autosomal Dominant Congenital Nonprogressive Spinocerebellar Ataxia. Orphanet J. Rare Dis. 2012, 7, 67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Gerber, S.; Alzayady, K.J.; Burglen, L.; Brémond-Gignac, D.; Marchesin, V.; Roche, O.; Rio, M.; Funalot, B.; Calmon, R.; Durr, A.; et al. Recessive and Dominant De Novo ITPR1 Mutations Cause Gillespie Syndrome. Am. J. Hum. Genet. 2016, 98, 971–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. McEntagart, M.; Williamson, K.A.; Rainger, J.K.; Wheeler, A.; Seawright, A.; De Baere, E.; Verdin, H.; Bergendahl, L.T.; Quigley, A.; Rainger, J.; et al. A Restricted Repertoire of De Novo Mutations in ITPR1 Cause Gillespie Syndrome with Evidence for Dominant-Negative Effect. Am. J. Hum. Genet. 2016, 98, 981–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Matsumoto, M.; Nakagawa, T.; Inoue, T.; Nagata, E.; Tanaka, K.; Takano, H.; Kuno, J.; Sakakibara, S.; Yamada, M.; Yoneshima, H.; et al. Ataxia and Epileptic Seizures in Mice Lacking Type 1 Inositol 1,4,5-Trisphosphate Receptor. Nature 1996, 379, 168–171. [Google Scholar] [CrossRef]
  48. Higo, T.; Hamada, K.; Hisatsune, C.; Nukina, N.; Hashikawa, T.; Hattori, M.; Nakamura, T.; Mikoshiba, K. Mechanism of ER Stress-Induced Brain Damage by IP(3) Receptor. Neuron 2010, 68, 865–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Kasumu, A.W.; Liang, X.; Egorova, P.; Vorontsova, D.; Bezprozvanny, I. Chronic Suppression of Inositol 1,4,5-Triphosphate Receptor-Mediated Calcium Signaling in Cerebellar Purkinje Cells Alleviates Pathological Phenotype in Spinocerebellar Ataxia 2 Mice. J. Neurosci. Off. J. Soc. Neurosci. 2012, 32, 12786–12796. [Google Scholar] [CrossRef] [Green Version]
  50. 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. Off. J. Soc. Neurosci. 2008, 28, 12713–12724. [Google Scholar] [CrossRef] [Green Version]
  51. Tang, T.-S.; Tu, H.; Chan, E.Y.W.; Maximov, A.; Wang, Z.; Wellington, C.L.; Hayden, M.R.; Bezprozvanny, I. Huntingtin and Huntingtin-Associated Protein 1 Influence Neuronal Calcium Signaling Mediated by Inositol-(1,4,5) Triphosphate Receptor Type 1. Neuron 2003, 39, 227–239. [Google Scholar] [CrossRef] [Green Version]
  52. Tiscione, S.A.; Casas, M.; Horvath, J.D.; Lam, V.; Hino, K.; Ory, D.S.; Santana, L.F.; Simó, S.; Dixon, R.E.; Dickson, E.J. IP3R-Driven Increases in Mitochondrial Ca2+ Promote Neuronal Death in NPC Disease. Proc. Natl. Acad. Sci. USA 2021, 118, e2110629118. [Google Scholar] [CrossRef] [PubMed]
  53. Staats, K.A.; Humblet-Baron, S.; Bento-Abreu, A.; Scheveneels, W.; Nikolaou, A.; Deckers, K.; Lemmens, R.; Goris, A.; van Ginderachter, J.A.; van Damme, P.; et al. Genetic Ablation of IP3 Receptor 2 Increases Cytokines and Decreases Survival of SOD1G93A Mice. Hum. Mol. Genet. 2016, 25, 3491–3499. [Google Scholar] [CrossRef] [Green Version]
  54. Ziegler, D.V.; Vindrieux, D.; Goehrig, D.; Jaber, S.; Collin, G.; Griveau, A.; Wiel, C.; Bendridi, N.; Djebali, S.; Farfariello, V.; et al. Calcium Channel ITPR2 and Mitochondria-ER Contacts Promote Cellular Senescence and Aging. Nat. Commun. 2021, 12, 720. [Google Scholar] [CrossRef]
  55. Bartok, A.; Weaver, D.; Golenár, T.; Nichtova, Z.; Katona, M.; Bánsághi, S.; Alzayady, K.J.; Thomas, V.K.; Ando, H.; Mikoshiba, K.; et al. IP3 Receptor Isoforms Differently Regulate ER-Mitochondrial Contacts and Local Calcium Transfer. Nat. Commun. 2019, 10, 3726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Wiel, C.; Lallet-Daher, H.; Gitenay, D.; Gras, B.; le Calvé, B.; Augert, A.; Ferrand, M.; Prevarskaya, N.; Simonnet, H.; Vindrieux, D.; et al. Endoplasmic Reticulum Calcium Release through ITPR2 Channels Leads to Mitochondrial Calcium Accumulation and Senescence. Nat. Commun. 2014, 5, 3792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Ma, X.; Warnier, M.; Raynard, C.; Ferrand, M.; Kirsh, O.; Defossez, P.-A.; Martin, N.; Bernard, D. The Nuclear Receptor RXRA Controls Cellular Senescence by Regulating Calcium Signaling. Aging Cell 2018, 17, e12831. [Google Scholar] [CrossRef]
  58. Kim, S.H.; Zhan, L.; Hanson, K.A.; Tibbetts, R.S. High-Content RNAi Screening Identifies the Type 1 Inositol Triphosphate Receptor as a Modifier of TDP-43 Localization and Neurotoxicity. Hum. Mol. Genet. 2012, 21, 4845–4856. [Google Scholar] [CrossRef] [Green Version]
  59. Chun, L.; Gong, J.; Yuan, F.; Zhang, B.; Liu, H.; Zheng, T.; Yu, T.; Xu, X.Z.S.; Liu, J. Metabotropic GABA Signalling Modulates Longevity in C. elegans. Nat. Commun. 2015, 6, 8828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Kawli, T.; Wu, C.; Tan, M.-W. Systemic and Cell Intrinsic Roles of Gqalpha Signaling in the Regulation of Innate Immunity, Oxidative Stress, and Longevity in Caenorhabditis Elegans. Proc. Natl. Acad. Sci. USA 2010, 107, 13788–13793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Burkewitz, K.; Feng, G.; Dutta, S.; Kelley, C.A.; Steinbaugh, M.; Cram, E.J.; Mair, W.B. Atf-6 Regulates Lifespan through ER-Mitochondrial Calcium Homeostasis. Cell Rep. 2020, 32, 108125. [Google Scholar] [CrossRef] [PubMed]
  62. Schoenherr, C.J.; Anderson, D.J. The Neuron-Restrictive Silencer Factor (NRSF): A Coordinate Repressor of Multiple Neuron-Specific Genes. Science 1995, 267, 1360–1363. [Google Scholar] [CrossRef] [PubMed]
  63. Ballas, N.; Grunseich, C.; Lu, D.D.; Speh, J.C.; Mandel, G. REST and Its Corepressors Mediate Plasticity of Neuronal Gene Chromatin throughout Neurogenesis. Cell 2005, 121, 645–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Chong, J.A.; Tapia-Ramirez, J.; Kim, S.; Toledo-Aral, J.J.; Zheng, Y.; Boutros, M.C.; Altshuller, Y.M.; Frohman, M.A.; Kraner, S.D.; Mandel, G. REST: A Mammalian Silencer Protein That Restricts Sodium Channel Gene Expression to Neurons. Cell 1995, 80, 949–957. [Google Scholar] [CrossRef] [Green Version]
  65. Hwang, J.-Y.; Zukin, R.S. REST, a Master Transcriptional Regulator in Neurodegenerative Disease. Curr. Opin. Neurobiol. 2018, 48, 193–200. [Google Scholar] [CrossRef] [PubMed]
  66. Lu, T.; Aron, L.; Zullo, J.; Pan, Y.; Kim, H.; Chen, Y.; Yang, T.-H.; Kim, H.-M.; Drake, D.; Liu, X.S.; et al. REST and Stress Resistance in Ageing and Alzheimer’s Disease. Nature 2014, 507, 448–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Meyer, K.; Feldman, H.M.; Lu, T.; Drake, D.; Lim, E.T.; Ling, K.-H.; Bishop, N.A.; Pan, Y.; Seo, J.; Lin, Y.-T.; et al. REST and Neural Gene Network Dysregulation in IPSC Models of Alzheimer’s Disease. Cell Rep. 2019, 26, 1112–1127.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Zuccato, C.; Tartari, M.; Crotti, A.; Goffredo, D.; Valenza, M.; Conti, L.; Cataudella, T.; Leavitt, B.R.; Hayden, M.R.; Timmusk, T.; et al. Huntingtin Interacts with REST/NRSF to Modulate the Transcription of NRSE-Controlled Neuronal Genes. Nat. Genet. 2003, 35, 76–83. [Google Scholar] [CrossRef]
  69. Zuccato, C.; Belyaev, N.; Conforti, P.; Ooi, L.; Tartari, M.; Papadimou, E.; MacDonald, M.; Fossale, E.; Zeitlin, S.; Buckley, N.; et al. Widespread Disruption of Repressor Element-1 Silencing Transcription Factor/Neuron-Restrictive Silencer Factor Occupancy at Its Target Genes in Huntington’s Disease. J. Neurosci. Off. J. Soc. Neurosci. 2007, 27, 6972–6983. [Google Scholar] [CrossRef]
  70. Sun, N.; Youle, R.J.; Finkel, T. The Mitochondrial Basis of Aging. Mol. Cell 2016, 61, 654–666. [Google Scholar] [CrossRef] [Green Version]
  71. 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] [PubMed]
  72. Perocchi, F.; Gohil, V.M.; Girgis, H.S.; Bao, X.R.; McCombs, J.E.; Palmer, A.E.; Mootha, V.K. MICU1 Encodes a Mitochondrial EF Hand Protein Required for Ca2+ Uptake. Nature 2010, 467, 291–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. 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] [PubMed] [Green Version]
  74. Plovanich, M.; Bogorad, R.L.; Sancak, Y.; Kamer, K.J.; Strittmatter, L.; Li, A.A.; Girgis, H.S.; Kuchimanchi, S.; de Groot, J.; Speciner, L.; et al. MICU2, a Paralog of MICU1, Resides within the Mitochondrial Uniporter Complex to Regulate Calcium Handling. PLoS ONE 2013, 8, e55785. [Google Scholar] [CrossRef] [Green Version]
  75. 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]
  76. 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] [PubMed]
  77. Yang, Y.-F.; Yang, W.; Liao, Z.-Y.; Wu, Y.-X.; Fan, Z.; Guo, A.; Yu, J.; Chen, Q.-N.; Wu, J.-H.; Zhou, J.; et al. MICU3 Regulates Mitochondrial Ca2+-Dependent Antioxidant Response in Skeletal Muscle Aging. Cell Death Dis. 2021, 12, 1115. [Google Scholar] [CrossRef]
  78. Qiu, J.; Tan, Y.-W.; Hagenston, A.M.; Martel, M.-A.; Kneisel, N.; Skehel, P.A.; Wyllie, D.J.A.; Bading, H.; Hardingham, G.E. Mitochondrial Calcium Uniporter Mcu Controls Excitotoxicity and Is Transcriptionally Repressed by Neuroprotective Nuclear Calcium Signals. Nat. Commun. 2013, 4, 2034. [Google Scholar] [CrossRef] [Green Version]
  79. König, T.; Tröder, S.E.; Bakka, K.; Korwitz, A.; Richter-Dennerlein, R.; Lampe, P.A.; Patron, M.; Mühlmeister, M.; Guerrero-Castillo, S.; Brandt, U.; et al. The M-AAA Protease Associated with Neurodegeneration Limits MCU Activity in Mitochondria. Mol. Cell 2016, 64, 148–162. [Google Scholar] [CrossRef] [Green Version]
  80. Kwong, J.Q.; Lu, X.; Correll, R.N.; Schwanekamp, J.A.; Vagnozzi, R.J.; Sargent, M.A.; York, A.J.; Zhang, J.; Bers, D.M.; Molkentin, J.D. The Mitochondrial Calcium Uniporter Selectively Matches Metabolic Output to Acute Contractile Stress in the Heart. Cell Rep. 2015, 12, 15–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Holmström, K.M.; Pan, X.; Liu, J.C.; Menazza, S.; Liu, J.; Nguyen, T.T.; Pan, H.; Parks, R.J.; Anderson, S.; Noguchi, A.; et al. Assessment of Cardiac Function in Mice Lacking the Mitochondrial Calcium Uniporter. J. Mol. Cell. Cardiol. 2015, 85, 178–182. [Google Scholar] [CrossRef] [Green Version]
  82. Dong, Z.; Shanmughapriya, S.; Tomar, D.; Siddiqui, N.; Lynch, S.; Nemani, N.; Breves, S.L.; Zhang, X.; Tripathi, A.; Palaniappan, P.; et al. Mitochondrial Ca2+ Uniporter Is a Mitochondrial Luminal Redox Sensor That Augments MCU Channel Activity. Mol. Cell 2017, 65, 1014–1028.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Choi, S.; Quan, X.; Bang, S.; Yoo, H.; Kim, J.; Park, J.; Park, K.-S.; Chung, J. Mitochondrial Calcium Uniporter in Drosophila Transfers Calcium between the Endoplasmic Reticulum and Mitochondria in Oxidative Stress-Induced Cell Death. J. Biol. Chem. 2017, 292, 14473–14485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Ishii, N.; Fujii, M.; Hartman, P.S.; Tsuda, M.; Yasuda, K.; Senoo-Matsuda, N.; Yanase, S.; Ayusawa, D.; Suzuki, K. A Mutation in Succinate Dehydrogenase Cytochrome b Causes Oxidative Stress and Ageing in Nematodes. Nature 1998, 394, 694–697. [Google Scholar] [CrossRef] [PubMed]
  85. Senoo-Matsuda, N.; Yasuda, K.; Tsuda, M.; Ohkubo, T.; Yoshimura, S.; Nakazawa, H.; Hartman, P.S.; Ishii, N. A Defect in the Cytochrome b Large Subunit in Complex II Causes Both Superoxide Anion Overproduction and Abnormal Energy Metabolism in Caenorhabditis Elegans. J. Biol. Chem. 2001, 276, 41553–41558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Jung, H.; Kim, S.Y.; Canbakis Cecen, F.S.; Cho, Y.; Kwon, S.-K. Dysfunction of Mitochondrial Ca2+ Regulatory Machineries in Brain Aging and Neurodegenerative Diseases. Front. Cell Dev. Biol. 2020, 8, 599792. [Google Scholar] [CrossRef] [PubMed]
  87. Palty, R.; Silverman, W.F.; Hershfinkel, M.; Caporale, T.; Sensi, S.L.; Parnis, J.; Nolte, C.; Fishman, D.; Shoshan-Barmatz, V.; Herrmann, S.; et al. NCLX Is an Essential Component of Mitochondrial Na+/Ca2+ Exchange. Proc. Natl. Acad. Sci. USA 2010, 107, 436–441. [Google Scholar] [CrossRef] [Green Version]
  88. Luongo, T.S.; Lambert, J.P.; Gross, P.; Nwokedi, M.; Lombardi, A.A.; Shanmughapriya, S.; Carpenter, A.C.; Kolmetzky, D.; Gao, E.; van Berlo, J.H.; et al. The Mitochondrial Na+/Ca2+ Exchanger Is Essential for Ca2+ Homeostasis and Viability. Nature 2017, 545, 93–97. [Google Scholar] [CrossRef] [Green Version]
  89. De Marchi, U.; Santo-Domingo, J.; Castelbou, C.; Sekler, I.; Wiederkehr, A.; Demaurex, N. NCLX Protein, but Not LETM1, Mediates Mitochondrial Ca2+ Extrusion, Thereby Limiting Ca2+-Induced NAD(P)H Production and Modulating Matrix Redox State. J. Biol. Chem. 2014, 289, 20377–20385. [Google Scholar] [CrossRef] [Green Version]
  90. Natarajan, G.K.; Glait, L.; Mishra, J.; Stowe, D.F.; Camara, A.K.S.; Kwok, W.-M. Total Matrix Ca2+ Modulates Ca2+ Efflux via the Ca2+/H+ Exchanger in Cardiac Mitochondria. Front. Physiol. 2020, 11, 510600. [Google Scholar] [CrossRef]
  91. Jiang, D.; Zhao, L.; Clapham, D.E. Genome-Wide RNAi Screen Identifies Letm1 as a Mitochondrial Ca2+/H+ Antiporter. Science 2009, 326, 144–147. [Google Scholar] [CrossRef] [Green Version]
  92. Hagenston, A.M.; Yan, J.; Bas-Orth, C.; Tan, Y.; Sekler, I.; Bading, H. Disrupted Expression of Mitochondrial NCLX Sensitizes Neuroglial Networks to Excitotoxic Stimuli and Renders Synaptic Activity Toxic. J. Biol. Chem. 2021, 298, 101508. [Google Scholar] [CrossRef] [PubMed]
  93. Jadiya, P.; Kolmetzky, D.W.; Tomar, D.; di Meco, A.; Lombardi, A.A.; Lambert, J.P.; Luongo, T.S.; Ludtmann, M.H.; Praticò, D.; Elrod, J.W. Impaired Mitochondrial Calcium Efflux Contributes to Disease Progression in Models of Alzheimer’s Disease. Nat. Commun. 2019, 10, 3885. [Google Scholar] [CrossRef] [PubMed]
  94. Ludtmann, M.H.R.; Kostic, M.; Horne, A.; Gandhi, S.; Sekler, I.; Abramov, A.Y. LRRK2 Deficiency Induced Mitochondrial Ca2+ Efflux Inhibition Can Be Rescued by Na+/Ca2+/Li+ Exchanger Upregulation. Cell Death Dis. 2019, 10, 265. [Google Scholar] [CrossRef] [PubMed]
  95. Verma, M.; Callio, J.; Otero, P.A.; Sekler, I.; Wills, Z.P.; Chu, C.T. Mitochondrial Calcium Dysregulation Contributes to Dendrite Degeneration Mediated by PD/LBD-Associated LRRK2 Mutants. J. Neurosci. Off. J. Soc. Neurosci. 2017, 37, 11151–11165. [Google Scholar] [CrossRef] [PubMed]
  96. Kostic, M.; Ludtmann, M.H.R.; Bading, H.; Hershfinkel, M.; Steer, E.; Chu, C.T.; Abramov, A.Y.; Sekler, I. PKA Phosphorylation of NCLX Reverses Mitochondrial Calcium Overload and Depolarization, Promoting Survival of PINK1-Deficient Dopaminergic Neurons. Cell Rep. 2015, 13, 376–386. [Google Scholar] [CrossRef] [Green Version]
  97. Dolga, A.M.; Netter, M.F.; Perocchi, F.; Doti, N.; Meissner, L.; Tobaben, S.; Grohm, J.; Zischka, H.; Plesnila, N.; Decher, N.; et al. Mitochondrial Small Conductance SK2 Channels Prevent Glutamate-Induced Oxytosis and Mitochondrial Dysfunction. J. Biol. Chem. 2013, 288, 10792–10804. [Google Scholar] [CrossRef] [Green Version]
  98. Krabbendam, I.E.; Honrath, B.; Dilberger, B.; Iannetti, E.F.; Branicky, R.S.; Meyer, T.; Evers, B.; Dekker, F.J.; Koopman, W.J.H.; Beyrath, J.; et al. SK Channel-Mediated Metabolic Escape to Glycolysis Inhibits Ferroptosis and Supports Stress Resistance in C. elegans. Cell Death Dis. 2020, 11, 263. [Google Scholar] [CrossRef]
  99. Xu, W.; Liu, Y.; Wang, S.; McDonald, T.; van Eyk, J.E.; Sidor, A.; O’Rourke, B. Cytoprotective Role of Ca2+-Activated K+ Channels in the Cardiac Inner Mitochondrial Membrane. Science 2002, 298, 1029–1033. [Google Scholar] [CrossRef] [PubMed]
  100. Gururaja Rao, S.; Bednarczyk, P.; Towheed, A.; Shah, K.; Karekar, P.; Ponnalagu, D.; Jensen, H.N.; Addya, S.; Reyes, B.A.S.; van Bockstaele, E.J.; et al. BKCa (Slo) Channel Regulates Mitochondrial Function and Lifespan in Drosophila Melanogaster. Cells 2019, 8, 945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Krauss, S.; Zhang, C.Y.; Lowell, B.B. The Mitochondrial Uncoupling-Protein Homologues. Nat. Rev. Mol. Cell Biol. 2005, 6, 248–261. [Google Scholar] [CrossRef] [PubMed]
  102. Dietrich, M.O.; Horvath, T.L. The Role of Mitochondrial Uncoupling Proteins in Lifespan. Pflug. Arch. Eur. J. Physiol. 2010, 459, 269–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Enerbäck, S.; Jacobsson, A.; Simpson, E.M.; Guerra, C.; Yamashita, H.; Harper, M.E.; Kozak, L.P. Mice Lacking Mitochondrial Uncoupling Protein Are Cold-Sensitive but Not Obese. Nature 1997, 387, 90–94. [Google Scholar] [CrossRef]
  104. Van Marken Lichtenbelt, W.D.; Vanhommerig, J.W.; Smulders, N.M.; Drossaerts, J.M.A.F.L.; Kemerink, G.J.; Bouvy, N.D.; Schrauwen, P.; Teule, G.J.J. Cold-Activated Brown Adipose Tissue in Healthy Men. N. Engl. J. Med. 2009, 360, 1500–1508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Cypess, A.M.; Lehman, S.; Williams, G.; Tal, I.; Rodman, D.; Goldfine, A.B.; Kuo, F.C.; Palmer, E.L.; Tseng, Y.-H.; Doria, A.; et al. Identification and Importance of Brown Adipose Tissue in Adult Humans. N. Engl. J. Med. 2009, 360, 1509–1517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Fleury, C.; Neverova, M.; Collins, S.; Raimbault, S.; Champigny, O.; Levi-Meyrueis, C.; Bouillaud, F.; Seldin, M.F.; Surwit, R.S.; Ricquier, D.; et al. Uncoupling Protein-2: A Novel Gene Linked to Obesity and Hyperinsulinemia. Nat. Genet. 1997, 15, 269–272. [Google Scholar] [CrossRef]
  107. Boss, O.; Samec, S.; Paoloni-Giacobino, A.; Rossier, C.; Dulloo, A.; Seydoux, J.; Muzzin, P.; Giacobino, J.P. Uncoupling Protein-3: A New Member of the Mitochondrial Carrier Family with Tissue-Specific Expression. FEBS Lett. 1997, 408, 39–42. [Google Scholar] [CrossRef] [Green Version]
  108. Echtay, K.S.; Roussel, D.; St-Plerre, J.; Jekabsons, M.B.; Cadenas, S.; Stuart, J.A.; Harper, J.A.; Roebuck, S.J.; Morrison, A.; Pickering, S.; et al. Superoxide Activates Mitochondrial Uncoupling Proteins. Nature 2002, 415, 96–99. [Google Scholar] [CrossRef]
  109. Arsenijevic, D.; Onuma, H.; Pecqueur, C.; Raimbault, S.; Manning, B.S.; Miroux, B.; Couplan, E.; Alves-Guerra, M.C.; Goubern, M.; Surwit, R.; et al. Disruption of the Uncoupling Protein-2 Gene in Mice Reveals a Role in Immunity and Reactive Oxygen Species Production. Nat. Genet. 2000, 26, 435–439. [Google Scholar] [CrossRef] [PubMed]
  110. Vidal-Puig, A.J.; Grujic, D.; Zhang, C.Y.; Hagen, T.; Boss, O.; Ido, Y.; Szczepanik, A.; Wade, J.; Mootha, V.; Cortright, R.; et al. Energy Metabolism in Uncoupling Protein 3 Gene Knockout Mice. J. Biol. Chem. 2000, 275, 16258–16266. [Google Scholar] [CrossRef] [Green Version]
  111. Esterbauer, H.; Schneitler, C.; Oberkofler, H.; Ebenbichler, C.; Paulweber, B.; Sandhofer, F.; Ladurner, G.; Hell, E.; Strosberg, A.D.; Patsch, J.R.; et al. A Common Polymorphism in the Promoter of UCP2 Is Associated with Decreased Risk of Obesity in Middle-Aged Humans. Nat. Genet. 2001, 28, 178–183. [Google Scholar] [CrossRef]
  112. Wolkow, C.A.; Iser, W.B. Uncoupling Protein Homologs May Provide a Link between Mitochondria, Metabolism and Lifespan. Ageing Res. Rev. 2006, 5, 196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Klingenberg, M.; Appel, M. The Uncoupling Protein Dimer Can Form a Disulfide Cross-Link between the Mobile C-Terminal SH Groups. Eur. J. Biochem. 1989, 180, 123–131. [Google Scholar] [CrossRef] [PubMed]
  114. Skulachev, V.P. Fatty Acid Circuit as a Physiological Mechanism of Uncoupling of Oxidative Phosphorylation. FEBS Lett. 1991, 294, 158–162. [Google Scholar] [CrossRef] [Green Version]
  115. Korshunov, S.S.; Skulachev, V.P.; Starkov, A.A. High Protonic Potential Actuates a Mechanism of Production of Reactive Oxygen Species in Mitochondria. FEBS Lett. 1997, 416, 15–18. [Google Scholar] [CrossRef] [Green Version]
  116. Duval, C.; Nègre-Salvayre, A.; Doglio, A.; Salvayre, R.; Pénicaud, L.; Casteilla, L. Increased Reactive Oxygen Species Production with Antisense Oligonucleotides Directed against Uncoupling Protein 2 in Murine Endothelial Cells. Biochem. Cell Biol. 2002, 80, 757–764. [Google Scholar] [CrossRef] [PubMed]
  117. Nègre-Salvayre, A.; Hirtz, C.; Carrera, G.; Cazenave, R.; Troly, M.; Salvayre, R.; Pénicaud, L.; Casteilla, L. A Role for Uncoupling Protein-2 as a Regulator of Mitochondrial Hydrogen Peroxide Generation. FASEB J. 1997, 11, 809–815. [Google Scholar] [CrossRef] [PubMed]
  118. Feng, J.; Bussière, F.; Hekimi, S. Mitochondrial Electron Transport Is a Key Determinant of Life Span in Caenorhabditis Elegans. Dev. Cell 2001, 1, 633–644. [Google Scholar] [CrossRef] [Green Version]
  119. Holzenberger, M.; Dupont, J.; Ducos, B.; Leneuve, P.; Géloën, A.; Even, P.C.; Cervera, P.; le Bouc, Y. IGF-1 Receptor Regulates Lifespan and Resistance to Oxidative Stress in Mice. Nature 2003, 421, 182–187. [Google Scholar] [CrossRef]
  120. Fridell, Y.W.C.; Sánchez-Blanco, A.; Silvia, B.A.; Helfand, S.L. Targeted Expression of the Human Uncoupling Protein 2 (HUCP2) to Adult Neurons Extends Life Span in the Fly. Cell Metab. 2005, 1, 145–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Lee, S.S.; Lee, R.Y.N.; Fraser, A.G.; Kamath, R.S.; Ahringer, J.; Ruvkun, G. A Systematic RNAi Screen Identifies a Critical Role for Mitochondria in C. elegans Longevity. Nat. Genet. 2003, 33, 40–48. [Google Scholar] [CrossRef] [PubMed]
  122. Dillin, A.; Hsu, A.L.; Arantes-Oliveira, N.; Lehrer-Graiwer, J.; Hsin, H.; Fraser, A.G.; Kamath, R.S.; Ahringer, J.; Kenyon, C. Rates of Behavior and Aging Specified by Mitochondrial Function during Development. Science 2002, 298, 2398–2401. [Google Scholar] [CrossRef] [PubMed]
  123. Speakman, J.R.; Talbot, D.A.; Selman, C.; Snart, S.; McLaren, J.S.; Redman, P.; Krol, E.; Jackson, D.M.; Johnson, M.S.; Brand, M.D. Uncoupled and Surviving: Individual Mice with High Metabolism Have Greater Mitochondrial Uncoupling and Live Longer. Aging Cell 2004, 3, 87–95. [Google Scholar] [CrossRef]
  124. Andrews, Z.B.; Horvath, T.L. Uncoupling Protein-2 Regulates Lifespan in Mice. Am. J. Physiol. Endocrinol. Metab. 2009, 296, E621–E627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Conti, B.; Sanchez-Alavez, M.; Winsky-Sommerer, R.; Morale, M.C.; Lucero, J.; Brownell, S.; Fabre, V.; Huitron-Resendiz, S.; Henriksen, S.; Zorrilla, E.P.; et al. Transgenic Mice with a Reduced Core Body Temperature Have an Increased Life Span. Science 2006, 314, 825–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Fridell, Y.W.C.; Hoh, M.; Kréneisz, O.; Hosier, S.; Chang, C.; Scantling, D.; Mulkey, D.K.; Helfand, S.L. Increased Uncoupling Protein (UCP) Activity in Drosophila Insulin-Producing Neurons Attenuates Insulin Signaling and Extends Lifespan. Aging 2009, 1, 699–713. [Google Scholar] [CrossRef] [Green Version]
  127. Humphrey, D.M.; Toivonen, J.M.; Giannakou, M.; Partridge, L.; Brand, M.D. Expression of Human Uncoupling Protein-3 in Drosophila Insulin-Producing Cells Increases Insulin-like Peptide (DILP) Levels and Shortens Lifespan. Exp. Gerontol. 2009, 44, 316–327. [Google Scholar] [CrossRef]
  128. Bevilacqua, L.; Ramsey, J.J.; Hagopian, K.; Weindruch, R.; Harper, M.E. Effects of Short- and Medium-Term Calorie Restriction on Muscle Mitochondrial Proton Leak and Reactive Oxygen Species Production. Am. J. Physiol. Endocrinol. Metab. 2004, 286, E852–E861. [Google Scholar] [CrossRef] [Green Version]
  129. McDonald, R.B.; Walker, K.M.; Warman, D.B.; Griffey, S.M.; Warden, C.H.; Ramsey, J.J.; Horwitz, B.A. Characterization of Survival and Phenotype throughout the Life Span in UCP2/UCP3 Genetically Altered Mice. Exp. Gerontol. 2008, 43, 1061–1068. [Google Scholar] [CrossRef]
  130. Brand, M.D. Uncoupling to Survive? The Role of Mitochondrial Inefficiency in Ageing. Exp. Gerontol. 2000, 35, 811–820. [Google Scholar] [CrossRef]
  131. Gates, A.C.; Bernal-Mizrachi, C.; Chinault, S.L.; Feng, C.; Schneider, J.G.; Coleman, T.; Malone, J.P.; Townsend, R.R.; Chakravarthy, M.V.; Semenkovich, C.F. Respiratory Uncoupling in Skeletal Muscle Delays Death and Diminishes Age-Related Disease. Cell Metab. 2007, 6, 497–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Rose, G.; Crocco, P.; D’Aquila, P.; Montesanto, A.; Bellizzi, D.; Passarino, G. Two Variants Located in the Upstream Enhancer Region of Human UCP1 Gene Affect Gene Expression and Are Correlated with Human Longevity. Exp. Gerontol. 2011, 46, 897–904. [Google Scholar] [CrossRef] [PubMed]
  133. Kontani, Y.; Wang, Y.; Kimura, K.; Inokuma, K.I.; Saito, M.; Suzuki-Miura, T.; Wang, Z.; Sato, Y.; Mori, N.; Yamashita, H. UCP1 Deficiency Increases Susceptibility to Diet-Induced Obesity with Age. Aging Cell 2005, 4, 147–155. [Google Scholar] [CrossRef] [PubMed]
  134. Barbosa, M.C.; Grosso, R.A.; Fader, C.M. Hallmarks of Aging: An Autophagic Perspective. Front. Endocrinol. 2019, 10, 790. [Google Scholar] [CrossRef]
  135. Vellai, T.; Takács-Vellai, K.; Sass, M.; Klionsky, D.J. The Regulation of Aging: Does Autophagy Underlie Longevity? Trends Cell Biol. 2009, 19, 487–494. [Google Scholar] [CrossRef] [Green Version]
  136. Nakamura, S.; Yoshimori, T. Autophagy and Longevity. Mol. Cells 2018, 41, 65–72. [Google Scholar] [CrossRef] [PubMed]
  137. Khalil, H.; Tazi, M.; Caution, K.; Ahmed, A.; Kanneganti, A.; Assani, K.; Kopp, B.; Marsh, C.; Dakhlallah, D.; Amer, A.O. Aging Is Associated with Hypermethylation of Autophagy Genes in Macrophages. Epigenetics 2016, 11, 381–388. [Google Scholar] [CrossRef] [Green Version]
  138. Liu, Y.; Palanivel, R.; Rai, E.; Park, M.; Gabor, T.V.; Scheid, M.P.; Xu, A.; Sweeney, G. Adiponectin Stimulates Autophagy and Reduces Oxidative Stress to Enhance Insulin Sensitivity during High-Fat Diet Feeding in Mice. Diabetes 2015, 64, 36–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Ravikumar, B.; Vacher, C.; Berger, Z.; Davies, J.E.; Luo, S.; Oroz, L.G.; Scaravilli, F.; Easton, D.F.; Duden, R.; O’Kane, C.J.; et al. Inhibition of MTOR Induces Autophagy and Reduces Toxicity of Polyglutamine Expansions in Fly and Mouse Models of Huntington Disease. Nat. Genet. 2004, 36, 585–595. [Google Scholar] [CrossRef] [Green Version]
  140. Pyo, J.O.; Yoo, S.M.; Ahn, H.H.; Nah, J.; Hong, S.H.; Kam, T.I.; Jung, S.; Jung, Y.K. Overexpression of Atg5 in Mice Activates Autophagy and Extends Lifespan. Nat. Commun. 2013, 4, 2300. [Google Scholar] [CrossRef] [Green Version]
  141. Bjedov, I.; Cochemé, H.M.; Foley, A.; Wieser, D.; Woodling, N.S.; Castillo-Quan, J.I.; Norvaisas, P.; Lujan, C.; Regan, J.; Toivonen, J.M.; et al. Fine-Tuning Autophagy Maximises Lifespan and Is Associated with Changes in Mitochondrial Gene Expression in Drosophila. PLoS Genet. 2020, 16, e1009083. [Google Scholar] [CrossRef] [PubMed]
  142. Hansen, M.; Chandra, A.; Mitic, L.L.; Onken, B.; Driscoll, M.; Kenyon, C. A Role for Autophagy in the Extension of Lifespan by Dietary Restriction in C. elegans. PLoS Genet. 2008, 4, e24. [Google Scholar] [CrossRef] [Green Version]
  143. Sun, Y.; Li, M.; Zhao, D.; Li, X.; Yang, C.; Wang, X. Lysosome Activity Is Modulated by Multiple Longevity Pathways and Is Important for Lifespan Extension in C. elegans. eLife 2020, 9, e55745. [Google Scholar] [CrossRef] [PubMed]
  144. Zhang, C.; Cuervo, A.M. Restoration of Chaperone-Mediated Autophagy in Aging Liver Improves Cellular Maintenance and Hepatic Function. Nat. Med. 2008, 14, 959–965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Kiffin, R.; Christian, C.; Knecht, E.; Cuervo, A.M. Activation of Chaperone-Mediated Autophagy during Oxidative Stress. Mol. Biol. Cell 2004, 15, 4829–4840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Bouderlique, T.; Vuppalapati, K.K.; Newton, P.T.; Li, L.; Barenius, B.; Chagin, A.S. Targeted Deletion of Atg5 in Chondrocytes Promotes Age-Related Osteoarthritis. Ann. Rheum. Dis. 2016, 75, 627–631. [Google Scholar] [CrossRef] [Green Version]
  147. Wu, J.J.; Quijano, C.; Chen, E.; Liu, H.; Cao, L.; Fergusson, M.M.; Rovira, I.I.; Gutkind, S.; Daniels, M.P.; Komatsu, M.; et al. Mitochondrial Dysfunction and Oxidative Stress Mediate the Physiological Impairment Induced by the Disruption of Autophagy. Aging 2009, 1, 425–437. [Google Scholar] [CrossRef] [Green Version]
  148. Juhász, G.; Érdi, B.; Sass, M.; Neufeld, T.P. Atg7-Dependent Autophagy Promotes Neuronal Health, Stress Tolerance, and Longevity but Is Dispensable for Metamorphosis in Drosophila. Genes Dev. 2007, 21, 3061–3066. [Google Scholar] [CrossRef] [Green Version]
  149. Inoue, K.; Rispoli, J.; Kaphzan, H.; Klann, E.; Chen, E.I.; Kim, J.; Komatsu, M.; Abeliovich, A. Macroautophagy Deficiency Mediates Age-Dependent Neurodegeneration through a Phospho-Tau Pathway. Mol. Neurodegener. 2012, 7, 48. [Google Scholar] [CrossRef] [Green Version]
  150. Komatsu, M.; Waguri, S.; Chiba, T.; Murata, S.; Iwata, J.I.; Tanida, I.; Ueno, T.; Koike, M.; Uchiyama, Y.; Kominami, E.; et al. Loss of Autophagy in the Central Nervous System Causes Neurodegeneration in Mice. Nature 2006, 441, 880–884. [Google Scholar] [CrossRef] [PubMed]
  151. Hara, T.; Nakamura, K.; Matsui, M.; Yamamoto, A.; Nakahara, Y.; Suzuki-Migishima, R.; Yokoyama, M.; Mishima, K.; Saito, I.; Okano, H.; et al. Suppression of Basal Autophagy in Neural Cells Causes Neurodegenerative Disease in Mice. Nature 2006, 441, 885–889. [Google Scholar] [CrossRef] [PubMed]
  152. Venkatachalam, K.; Wong, C.-O.; Zhu, M.X. The Role of TRPMLs in Endolysosomal Trafficking and Function. Cell Calcium 2015, 58, 48–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Wong, C.-O.; Li, R.; Montell, C.; Venkatachalam, K. Drosophila TRPML Is Required for TORC1 Activation. Curr. Biol. 2012, 22, 1616–1621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Venkatachalam, K.; Wong, C.-O.; Montell, C. Feast or Famine. Autophagy 2013, 9, 98–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Bargal, R.; Avidan, N.; Ben-Asher, E.; Olender, Z.; Zeigler, M.; Frumkin, A.; Raas-Rothschild, A.; Glusman, G.; Lancet, D.; Bach, G. Identification of the Gene Causing Mucolipidosis Type IV. Nat. Genet. 2000, 26, 118–121. [Google Scholar] [CrossRef]
  156. Bassi, M.T.; Manzoni, M.; Monti, E.; Pizzo, M.T.; Ballabio, A.; Borsani, G. Cloning of the Gene Encoding a Novel Integral Membrane Protein, Mucolipidin-and Identification of the Two Major Founder Mutations Causing Mucolipidosis Type IV. Am. J. Hum. Genet. 2000, 67, 1110–1120. [Google Scholar] [CrossRef]
  157. Sun, M.; Goldin, E.; Stahl, S.; Falardeau, J.L.; Kennedy, J.C.; Acierno, J.S.; Bove, C.; Kaneski, C.R.; Nagle, J.; Bromley, M.C.; et al. Mucolipidosis Type IV Is Caused by Mutations in a Gene Encoding a Novel Transient Receptor Potential Channel. Hum. Mol. Genet. 2000, 9, 2471–2478. [Google Scholar] [CrossRef] [Green Version]
  158. Vergarajauregui, S.; Connelly, P.S.; Daniels, M.P.; Puertollano, R. Autophagic Dysfunction in Mucolipidosis Type IV Patients. Hum. Mol. Genet. 2008, 17, 2723–2737. [Google Scholar] [CrossRef]
  159. Sancak, Y.; Bar-Peled, L.; Zoncu, R.; Markhard, A.L.; Nada, S.; Sabatini, D.M. Ragulator-Rag Complex Targets MTORC1 to the Lysosomal Surface and Is Necessary for Its Activation by Amino Acids. Cell 2010, 141, 290–303. [Google Scholar] [CrossRef] [Green Version]
  160. Xu, M.; Almasi, S.; Yang, Y.; Yan, C.; Sterea, A.M.; Rizvi Syeda, A.K.; Shen, B.; Richard Derek, C.; Huang, P.; Gujar, S.; et al. The Lysosomal TRPML1 Channel Regulates Triple Negative Breast Cancer Development by Promoting MTORC1 and Purinergic Signaling Pathways. Cell Calcium 2019, 79, 80–88. [Google Scholar] [CrossRef]
  161. Yang, Y.; Xu, M.; Zhu, X.; Yao, J.; Shen, B.; Dong, X.P. Lysosomal Ca2+ Release Channel TRPML1 Regulates Lysosome Size by Promoting MTORC1 Activity. Eur. J. Cell Biol. 2019, 98, 116–123. [Google Scholar] [CrossRef] [PubMed]
  162. Li, R.-J.; Xu, J.; Fu, C.; Zhang, J.; Zheng, Y.G.; Jia, H.; Liu, J.O. Regulation of MTORC1 by Lysosomal Calcium and Calmodulin. eLife 2016, 5, e19360. [Google Scholar] [CrossRef] [PubMed]
  163. Roczniak-Ferguson, A.; Petit, C.S.; Froehlich, F.; Qian, S.; Ky, J.; Angarola, B.; Walther, T.C.; Ferguson, S.M. The Transcription Factor TFEB Links MTORC1 Signaling to Transcriptional Control of Lysosome Homeostasis. Sci. Signal. 2012, 5, ra42. [Google Scholar] [CrossRef] [Green Version]
  164. Martina, J.A.; Chen, Y.; Gucek, M.; Puertollano, R. MTORC1 Functions as a Transcriptional Regulator of Autophagy by Preventing Nuclear Transport of TFEB. Autophagy 2012, 8, 903–914. [Google Scholar] [CrossRef] [Green Version]
  165. Martina, J.A.; Diab, H.I.; Lishu, L.; Jeong-A, L.; Patange, S.; Raben, N.; Puertollano, R. The Nutrient-Responsive Transcription Factor TFE3 Promotes Autophagy, Lysosomal Biogenesis, and Clearance of Cellular Debris. Sci. Signal. 2014, 7, ra9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Medina, D.L.; Fraldi, A.; Bouche, V.; Annunziata, F.; Mansueto, G.; Spampanato, C.; Puri, C.; Pignata, A.; Martina, J.A.; Sardiello, M.; et al. Transcriptional Activation of Lysosomal Exocytosis Promotes Cellular Clearance. Dev. Cell 2011, 21, 421–430. [Google Scholar] [CrossRef] [PubMed]
  167. Venkatachalam, K.; Long, A.A.; Elsaesser, R.; Nikolaeva, D.; Broadie, K.; Montell, C. Motor Deficit in a Drosophila Model of Mucolipidosis Type IV Due to Defective Clearance of Apoptotic Cells. Cell 2008, 135, 838–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Venugopal, B.; Browning, M.F.; Curcio-Morelli, C.; Varro, A.; Michaud, N.; Nanthakumar, N.; Walkley, S.U.; Pickel, J.; Slaugenhaupt, S.A. Neurologic, Gastric, and Opthalmologic Pathologies in a Murine Model of Mucolipidosis Type IV. Am. J. Hum. Genet. 2007, 81, 1070–1083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Ehninger, D.; Neff, F.; Xie, K. Longevity, Aging and Rapamycin. Cell. Mol. Life Sci. CMLS 2014, 71, 4325–4346. [Google Scholar] [CrossRef] [Green Version]
  170. Zhang, X.; Chen, W.; Gao, Q.; Yang, J.; Yan, X.; Zhao, H.; Su, L.; Yang, M.; Gao, C.; Yao, Y.; et al. Rapamycin Directly Activates Lysosomal Mucolipin TRP Channels Independent of MTOR. PLoS Biol. 2019, 17, e3000252. [Google Scholar] [CrossRef]
  171. Selman, C.; Tullet, J.M.A.; Wieser, D.; Irvine, E.; Lingard, S.J.; Choudhury, A.I.; Claret, M.; Al-Qassab, H.; Carmignac, D.; Ramadani, F.; et al. Ribosomal Protein S6 Kinase 1 Signaling Regulates Mammalian Life Span. Science 2009, 326, 140–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Wu, J.J.; Liu, J.; Chen, E.B.; Wang, J.J.; Cao, L.; Narayan, N.; Fergusson, M.M.; Rovira, I.I.; Allen, M.; Springer, D.A.; et al. Increased Mammalian Lifespan and a Segmental and Tissue-Specific Slowing of Aging after Genetic Reduction of MTOR Expression. Cell Rep. 2013, 4, 913–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Templeman, N.M.; Murphy, C.T. Regulation of Reproduction and Longevity by Nutrient-Sensing Pathways. J. Cell Biol. 2018, 217, 93–106. [Google Scholar] [CrossRef]
  174. Castillo-Quan, J.I.; Tain, L.S.; Kinghorn, K.J.; Li, L.; Grönke, S.; Hinze, Y.; Blackwell, T.K.; Bjedov, I.; Partridge, L. A Triple Drug Combination Targeting Components of the Nutrient-Sensing Network Maximizes Longevity. Proc. Natl. Acad. Sci. USA 2019, 116, 20817–20819. [Google Scholar] [CrossRef] [Green Version]
  175. Lu, Y.X.; Regan, J.C.; Eßer, J.; Drews, L.F.; Weinseis, T.; Stinn, J.; Hahn, O.; Miller, R.A.; Grönke, S.; Partridge, L. A TORC1-Histone Axis Regulates Chromatin Organisation and Non-Canonical Induction of Autophagy to Ameliorate Ageing. eLife 2021, 10, e62233. [Google Scholar] [CrossRef] [PubMed]
  176. Lee, J.-H.; McBrayer, M.K.; Wolfe, D.M.; Haslett, L.J.; Kumar, A.; Sato, Y.; Lie, P.P.Y.; Mohan, P.; Coffey, E.E.; Kompella, U.; et al. Presenilin 1 Maintains Lysosomal Ca2+ Homeostasis via TRPML1 by Regulating VATPase-Mediated Lysosome Acidification. Cell Rep. 2015, 12, 1430–1444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Ruas, M.; Rietdorf, K.; Arredouani, A.; Davis, L.C.; Lloyd-Evans, E.; Koegel, H.; Funnell, T.M.; Morgan, A.J.; Ward, J.A.; Watanabe, K.; et al. Purified TPC Isoforms Form NAADP Receptors with Distinct Roles for Ca(2+) Signaling and Endolysosomal Trafficking. Curr. Biol. 2010, 20, 703–709. [Google Scholar] [CrossRef] [Green Version]
  178. Ruas, M.; Davis, L.C.; Chen, C.; Morgan, A.J.; Chuang, K.; Walseth, T.F.; Grimm, C.; Garnham, C.; Powell, T.; Platt, N.; et al. Expression of Ca2+-Permeable Two-Pore Channels Rescues NAADP Signalling in TPC-Deficient Cells. EMBO J. 2015, 34, 1743–1758. [Google Scholar] [CrossRef]
  179. Gerndt, S.; Chen, C.C.; Chao, Y.K.; Yuan, Y.; Burgstaller, S.; Rosato, A.S.; Krogsaeter, E.; Urban, N.; Jacob, K.; Nguyen, O.N.P.; et al. Agonist-Mediated Switching of Ion Selectivity in TPC2 Differentially Promotes Lysosomal Function. eLife 2020, 9, e54712. [Google Scholar] [CrossRef]
  180. Wang, X.; Zhang, X.; Dong, X.P.; Samie, M.; Li, X.; Cheng, X.; Goschka, A.; Shen, D.; Zhou, Y.; Harlow, J.; et al. TPC Proteins Are Phosphoinositide- Activated Sodium-Selective Ion Channels in Endosomes and Lysosomes. Cell 2012, 151, 372–383. [Google Scholar] [CrossRef] [Green Version]
  181. Pitt, S.J.; Funnell, T.M.; Sitsapesan, M.; Venturi, E.; Rietdorf, K.; Ruas, M.; Ganesan, A.; Gosain, R.; Churchill, G.C.; Zhu, M.X.; et al. TPC2 Is a Novel NAADP-Sensitive Ca2+ Release Channel, Operating as a Dual Sensor of Luminal PH and Ca2+. J. Biol. Chem. 2010, 285, 35039–35046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Ogunbayo, O.A.; Zhu, Y.; Rossi, D.; Sorrentino, V.; Ma, J.; Zhu, M.X.; Evans, A.M. Cyclic Adenosine Diphosphate Ribose Activates Ryanodine Receptors, Whereas NAADP Activates Two-Pore Domain Channels. J. Biol. Chem. 2011, 286, 9136–9140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Calcraft, P.J.; Ruas, M.; Pan, Z.; Cheng, X.; Arredouani, A.; Hao, X.; Tang, J.; Rietdorf, K.; Teboul, L.; Chuang, K.T.; et al. NAADP Mobilizes Calcium from Acidic Organelles through Two-Pore Channels. Nature 2009, 459, 596–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Müller, T.; Grossmann, S.; Mallmann, R.T.; Rommel, C.; Hein, L.; Klugbauer, N. Two-Pore Channels Affect EGF Receptor Signaling by Receptor Trafficking and Expression. iScience 2021, 24, 102099. [Google Scholar] [CrossRef] [PubMed]
  185. García-Rúa, V.; Feijóo-Bandín, S.; Rodríguez-Penas, D.; Mosquera-Leal, A.; Abu-Assi, E.; Beiras, A.; María Seoane, L.; Lear, P.; Parrington, J.; Portolés, M.; et al. Endolysosomal Two-Pore Channels Regulate Autophagy in Cardiomyocytes. J. Physiol. 2016, 594, 3061–3077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Lin, P.H.; Duann, P.; Komazaki, S.; Park, K.H.; Li, H.; Sun, M.; Sermersheim, M.; Gumpper, K.; Parrington, J.; Galione, A.; et al. Lysosomal Two-Pore Channel Subtype 2 (TPC2) Regulates Skeletal Muscle Autophagic Signaling. J. Biol. Chem. 2015, 290, 3377–3389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Pereira, G.J.S.; Antonioli, M.; Hirata, H.; Ureshino, R.P.; Nascimento, A.R.; Bincoletto, C.; Vescovo, T.; Piacentini, M.; Fimia, G.M.; Smaili, S.S. Glutamate Induces Autophagy via the Two-Pore Channels in Neural Cells. Oncotarget 2017, 8, 12730–12740. [Google Scholar] [CrossRef] [Green Version]
  188. Grimm, C.; Holdt, L.M.; Chen, C.C.; Hassan, S.; Müller, C.; Jörs, S.; Cuny, H.; Kissing, S.; Schröder, B.; Butz, E.; et al. High Susceptibility to Fatty Liver Disease in Two-Pore Channel 2-Deficient Mice. Nat. Commun. 2014, 5, 4699. [Google Scholar] [CrossRef]
  189. Hockey, L.N.; Kilpatrick, B.S.; Eden, E.R.; Lin-Moshier, Y.; Cristina Brailoiu, G.; Brailoiu, E.; Futter, C.E.; Schapira, A.H.; Marchant, J.S.; Patel, S. Dysregulation of Lysosomal Morphology by Pathogenic LRRK2 Is Corrected by TPC2 Inhibition. J. Cell Sci. 2015, 128, 232–238. [Google Scholar] [CrossRef] [Green Version]
  190. Lange, P.F.; Wartosch, L.; Jentsch, T.J.; Fuhrmann, J.C. ClC-7 Requires Ostm1 as a β-Subunit to Support Bone Resorption and Lysosomal Function. Nature 2006, 440, 220–223. [Google Scholar] [CrossRef] [Green Version]
  191. Kornak, U.; Kasper, D.; Bösl, M.R.; Kaiser, E.; Schweizer, M.; Schulz, A.; Friedrich, W.; Delling, G.; Jentsch, T.J. Loss of the ClC-7 Chloride Channel Leads to Osteopetrosis in Mice and Man. Cell 2001, 104, 205–215. [Google Scholar] [CrossRef] [Green Version]
  192. Jentsch, T.J. Chloride and the Endosomal-Lysosomal Pathway: Emerging Roles of CLC Chloride Transporters. J. Physiol. 2007, 578, 633–640. [Google Scholar] [CrossRef] [PubMed]
  193. Bose, S.; He, H.; Stauber, T. Neurodegeneration Upon Dysfunction of Endosomal/Lysosomal CLC Chloride Transporters. Front. Cell Dev. Biol. 2021, 9, 323. [Google Scholar] [CrossRef] [PubMed]
  194. Kasper, D.; Planells-Cases, R.; Fuhrmann, J.C.; Scheel, O.; Zeitz, O.; Ruether, K.; Schmitt, A.; Poët, M.; Steinfeld, R.; Schweizer, M.; et al. Loss of the Chloride Channel ClC-7 Leads to Lysosomal Storage Disease and Neurodegeneration. EMBO J. 2005, 24, 1079–1091. [Google Scholar] [CrossRef] [PubMed]
  195. Pressey, S.N.R.; O’Donnell, K.J.; Stauber, T.; Fuhrmann, J.C.; Tyynelä, J.; Jentsch, T.J.; Cooper, J.D. Distinct Neuropathologic Phenotypes after Disrupting the Chloride Transport Proteins ClC-6 or ClC-7/Ostm1. J. Neuropathol. Exp. Neurol. 2010, 69, 1228–1246. [Google Scholar] [CrossRef] [Green Version]
  196. Wang, S.; Tsun, Z.Y.; Wolfson, R.L.; Shen, K.; Wyant, G.A.; Plovanich, M.E.; Yuan, E.D.; Jones, T.D.; Chantranupong, L.; Comb, W.; et al. Metabolism. Lysosomal Amino Acid Transporter SLC38A9 Signals Arginine Sufficiency to MTORC1. Science 2015, 347, 188–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Rebsamen, M.; Pochini, L.; Stasyk, T.; de Araújo, M.E.G.; Galluccio, M.; Kandasamy, R.K.; Snijder, B.; Fauster, A.; Rudashevskaya, E.L.; Bruckner, M.; et al. SLC38A9 Is a Component of the Lysosomal Amino Acid Sensing Machinery That Controls MTORC1. Nature 2015, 519, 477–481. [Google Scholar] [CrossRef]
  198. Jung, J.; Genau, H.M.; Behrends, C. Amino Acid-Dependent MTORC1 Regulation by the Lysosomal Membrane Protein SLC38A9. Mol. Cell. Biol. 2015, 35, 2479–2494. [Google Scholar] [CrossRef] [Green Version]
  199. Wyant, G.A.; Abu-Remaileh, M.; Wolfson, R.L.; Chen, W.W.; Freinkman, E.; Danai, L.V.; vander Heiden, M.G.; Sabatini, D.M. MTORC1 Activator SLC38A9 Is Required to Efflux Essential Amino Acids from Lysosomes and Use Protein as a Nutrient. Cell 2017, 171, 642–654.e12. [Google Scholar] [CrossRef]
  200. Castellano, B.M.; Thelen, A.M.; Moldavski, O.; Feltes, M.; van der Welle, R.E.N.; Mydock-McGrane, L.; Jiang, X.; van Eijkeren, R.J.; Davis, O.B.; Louie, S.M.; et al. Lysosomal Cholesterol Activates MTORC1 via an SLC38A9-Niemann-Pick C1 Signaling Complex. Science 2017, 355, 1306–1311. [Google Scholar] [CrossRef] [Green Version]
  201. Cormerais, Y.; Massard, P.A.; Vucetic, M.; Giuliano, S.; Tambutté, E.; Durivault, J.; Vial, V.; Endou, H.; Wempe, M.F.; Parks, S.K.; et al. The Glutamine Transporter ASCT2 (SLC1A5) Promotes Tumor Growth Independently of the Amino Acid Transporter LAT1 (SLC7A5). J. Biol. Chem. 2018, 293, 2877–2887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Elorza, A.; Soro-Arnáiz, I.; Meléndez-Rodríguez, F.; Rodríguez-Vaello, V.; Marsboom, G.; de Cárcer, G.; Acosta-Iborra, B.; Albacete-Albacete, L.; Ordóñez, A.; Serrano-Oviedo, L.; et al. HIF2α Acts as an MTORC1 Activator through the Amino Acid Carrier SLC7A5. Mol. Cell 2012, 48, 681–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Nicklin, P.; Bergman, P.; Zhang, B.; Triantafellow, E.; Wang, H.; Nyfeler, B.; Yang, H.; Hild, M.; Kung, C.; Wilson, C.; et al. Bidirectional Transport of Amino Acids Regulates MTOR and Autophagy. Cell 2009, 136, 521–534. [Google Scholar] [CrossRef] [Green Version]
  204. Nakaya, M.; Xiao, Y.; Zhou, X.; Chang, J.H.; Chang, M.; Cheng, X.; Blonska, M.; Lin, X.; Sun, S.C. Inflammatory T Cell Responses Rely on Amino Acid Transporter ASCT2 Facilitation of Glutamine Uptake and MTORC1 Kinase Activation. Immunity 2014, 40, 692–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Kim, J.; Okamoto, H.; Huang, Z.J.; Anguiano, G.; Chen, S.; Liu, Q.; Cavino, K.; Xin, Y.; Na, E.; Hamid, R.; et al. Amino Acid Transporter Slc38a5 Controls Glucagon Receptor Inhibition-Induced Pancreatic α Cell Hyperplasia in Mice. Cell Metab. 2017, 25, 1348–1361.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Dato, S.; Hoxha, E.; Crocco, P.; Iannone, F.; Passarino, G.; Rose, G. Amino Acids and Amino Acid Sensing: Implication for Aging and Diseases. Biogerontology 2019, 20, 17–31. [Google Scholar] [CrossRef] [PubMed]
  207. Crocco, P.; Hoxha, E.; Dato, S.; de Rango, F.; Montesanto, A.; Rose, G.; Passarino, G. Physical Decline and Survival in the Elderly Are Affected by the Genetic Variability of Amino Acid Transporter Genes. Aging 2018, 10, 658–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Najumudeen, A.K.; Ceteci, F.; Fey, S.K.; Hamm, G.; Steven, R.T.; Hall, H.; Nikula, C.J.; Dexter, A.; Murta, T.; Race, A.M.; et al. The Amino Acid Transporter SLC7A5 Is Required for Efficient Growth of KRAS-Mutant Colorectal Cancer. Nat. Genet. 2021, 53, 16–26. [Google Scholar] [CrossRef] [PubMed]
  209. Settembre, C.; di Malta, C.; Polito, V.A.; Arencibia, M.G.; Vetrini, F.; Erdin, S.; Erdin, S.U.; Huynh, T.; Medina, D.; Colella, P.; et al. TFEB Links Autophagy to Lysosomal Biogenesis. Science 2011, 332, 1429–1433. [Google Scholar] [CrossRef] [Green Version]
  210. Settembre, C.; Zoncu, R.; Medina, D.L.; Vetrini, F.; Erdin, S.; Erdin, S.; Huynh, T.; Ferron, M.; Karsenty, G.; Vellard, M.C.; et al. A Lysosome-to-Nucleus Signalling Mechanism Senses and Regulates the Lysosome via MTOR and TFEB. EMBO J. 2012, 31, 1095–1108. [Google Scholar] [CrossRef] [Green Version]
  211. Palmieri, M.; Impey, S.; Kang, H.; di Ronza, A.; Pelz, C.; Sardiello, M.; Ballabio, A. Characterization of the CLEAR Network Reveals an Integrated Control of Cellular Clearance Pathways. Hum. Mol. Genet. 2011, 20, 3852–3866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Sardiello, M.; Palmieri, M.; di Ronza, A.; Medina, D.L.; Valenza, M.; Gennarino, V.A.; di Malta, C.; Donaudy, F.; Embrione, V.; Polishchuk, R.S.; et al. A Gene Network Regulating Lysosomal Biogenesis and Function. Science 2009, 325, 473–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Ploper, D.; Taelman, V.F.; Robert, L.; Perez, B.S.; Titz, B.; Chen, H.-W.; Graeber, T.G.; von Euw, E.; Ribas, A.; de Robertis, E.M. MITF Drives Endolysosomal Biogenesis and Potentiates Wnt Signaling in Melanoma Cells. Proc. Natl. Acad. Sci. USA 2015, 112, E420–E429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Bouché, V.; Espinosa, A.P.; Leone, L.; Sardiello, M.; Ballabio, A.; Botas, J. Drosophila Mitf Regulates the V-ATPase and the Lysosomal-Autophagic Pathway. Autophagy 2016, 12, 484–498. [Google Scholar] [CrossRef] [Green Version]
  215. Song, W.; Wang, F.; Lotfi, P.; Sardiello, M.; Segatori, L. 2-Hydroxypropyl-β-Cyclodextrin Promotes Transcription Factor EB-Mediated Activation of Autophagy. J. Biol. Chem. 2014, 289, 10211–10222. [Google Scholar] [CrossRef] [Green Version]
  216. Polito, V.A.; Li, H.; Martini-Stoica, H.; Wang, B.; Yang, L.; Xu, Y.; Swartzlander, D.B.; Palmieri, M.; di Ronza, A.; Lee, V.M.-Y.; et al. Selective Clearance of Aberrant Tau Proteins and Rescue of Neurotoxicity by Transcription Factor EB. EMBO Mol. Med. 2014, 6, 1142–1160. [Google Scholar] [CrossRef]
  217. Palmieri, M.; Pal, R.; Nelvagal, H.R.; Lotfi, P.; Stinnett, G.R.; Seymour, M.L.; Chaudhury, A.; Bajaj, L.; Bondar, V.V.; Bremner, L.; et al. MTORC1-Independent TFEB Activation via Akt Inhibition Promotes Cellular Clearance in Neurodegenerative Storage Diseases. Nat. Commun. 2017, 8, 14338. [Google Scholar] [CrossRef]
  218. Liu, Y.J.; McIntyre, R.L.; Janssens, G.E.; Williams, E.G.; Lan, J.; van Weeghel, M.; Schomakers, B.; van der Veen, H.; van der Wel, N.N.; Yao, P.; et al. Mitochondrial Translation and Dynamics Synergistically Extend Lifespan in C. elegans through HLH-30. J. Cell Biol. 2020, 219, e201907067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Lin, X.X.; Sen, I.; Janssens, G.E.; Zhou, X.; Fonslow, B.R.; Edgar, D.; Stroustrup, N.; Swoboda, P.; Yates, J.R.; Ruvkun, G.; et al. DAF-16/FOXO and HLH-30/TFEB Function as Combinatorial Transcription Factors to Promote Stress Resistance and Longevity. Nat. Commun. 2018, 9, 4400. [Google Scholar] [CrossRef] [Green Version]
  220. Wang, C.; Niederstrasser, H.; Douglas, P.M.; Lin, R.; Jaramillo, J.; Li, Y.; Olswald, N.W.; Zhou, A.; McMillan, E.A.; Mendiratta, S.; et al. Small-Molecule TFEB Pathway Agonists That Ameliorate Metabolic Syndrome in Mice and Extend C. elegans Lifespan. Nat. Commun. 2017, 8, 2270. [Google Scholar] [CrossRef]
  221. Nakamura, S.; Karalay, Ö.; Jäger, P.S.; Horikawa, M.; Klein, C.; Nakamura, K.; Latza, C.; Templer, S.E.; Dieterich, C.; Antebi, A. Mondo Complexes Regulate TFEB via TOR Inhibition to Promote Longevity in Response to Gonadal Signals. Nat. Commun. 2016, 7, 10944. [Google Scholar] [CrossRef] [PubMed]
  222. Lapierre, L.R.; de Magalhaes Filho, C.D.; McQuary, P.R.; Chu, C.C.; Visvikis, O.; Chang, J.T.; Gelino, S.; Ong, B.; Davis, A.E.; Irazoqui, J.E.; et al. The TFEB Orthologue HLH-30 Regulates Autophagy and Modulates Longevity in Caenorhabditis Elegans. Nat. Commun. 2013, 4, 2267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. O’Rourke, E.J.; Ruvkun, G. MXL-3 and HLH-30 Transcriptionally Link Lipolysis and Autophagy to Nutrient Availability. Nat. Cell Biol. 2013, 15, 668. [Google Scholar] [CrossRef] [PubMed]
  224. Kobayashi, T.; Piao, W.; Takamura, T.; Kori, H.; Miyachi, H.; Kitano, S.; Iwamoto, Y.; Yamada, M.; Imayoshi, I.; Shioda, S.; et al. Enhanced Lysosomal Degradation Maintains the Quiescent State of Neural Stem Cells. Nat. Commun. 2019, 10, 5446. [Google Scholar] [CrossRef] [PubMed]
  225. Pastore, N.; Vainshtein, A.; Klisch, T.J.; Armani, A.; Huynh, T.; Herz, N.J.; Polishchuk, E.V.; Sandri, M.; Ballabio, A. TFE3 Regulates Whole-Body Energy Metabolism in Cooperation with TFEB. EMBO Mol. Med. 2017, 9, 605–621. [Google Scholar] [CrossRef]
  226. Jung, J.; Liao, H.; Coker, S.A.; Liang, H.; Hancock, J.F.; Denicourt, C.; Venkatachalam, K. P53 Mitigates the Effects of Oncogenic HRAS in Urothelial Cells via the Repression of MCOLN1. iScience 2021, 24, 102701. [Google Scholar] [CrossRef]
  227. Jung, J.; Cho, K.-J.; Naji, A.K.; Clemons, K.N.; Wong, C.O.; Villanueva, M.; Gregory, S.; Karagas, N.E.; Tan, L.; Liang, H.; et al. HRAS-Driven Cancer Cells Are Vulnerable to TRPML1 Inhibition. EMBO Rep. 2019, 20, e46685. [Google Scholar] [CrossRef]
  228. Medina, D.L.; di Paola, S.; Peluso, I.; Armani, A.; de Stefani, D.; Venditti, R.; Montefusco, S.; Scotto-Rosato, A.; Prezioso, C.; Forrester, A.; et al. Lysosomal Calcium Signalling Regulates Autophagy through Calcineurin and TFEB. Nat. Cell Biol. 2015, 17, 288–299. [Google Scholar] [CrossRef] [Green Version]
  229. Kundu, S.T.; Grzeskowiak, C.L.; Fradette, J.J.; Gibson, L.A.; Rodriguez, L.B.; Creighton, C.J.; Scott, K.L.; Gibbons, D.L. TMEM106B Drives Lung Cancer Metastasis by Inducing TFEB-Dependent Lysosome Synthesis and Secretion of Cathepsins. Nat. Commun. 2018, 9, 2731. [Google Scholar] [CrossRef] [Green Version]
  230. Kauffman, E.C.; Ricketts, C.J.; Rais-Bahrami, S.; Yang, Y.; Merino, M.J.; Bottaro, D.P.; Srinivasan, R.; Linehan, W.M. Molecular Genetics and Cellular Features of TFE3 and TFEB Fusion Kidney Cancers. Nat. Rev. Urol. 2014, 11, 465–475. [Google Scholar] [CrossRef] [Green Version]
  231. Urbanelli, L.; Magini, A.; Ercolani, L.; Sagini, K.; Polchi, A.; Tancini, B.; Brozzi, A.; Armeni, T.; Principato, G.; Emiliani, C. Oncogenic H-Ras Up-Regulates Acid β-Hexosaminidase by a Mechanism Dependent on the Autophagy Regulator TFEB. PLoS ONE 2014, 9, e89485. [Google Scholar] [CrossRef] [PubMed]
  232. Jung, J.; Venkatachalam, K. TRPML1 and RAS-Driven Cancers–Exploring a Link with Great Therapeutic Potential. Channels 2019, 13, 374–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Perera, R.M.; Stoykova, S.; Nicolay, B.N.; Ross, K.N.; Fitamant, J.; Boukhali, M.; Lengrand, J.; Deshpande, V.; Selig, M.K.; Ferrone, C.R.; et al. Transcriptional Control of Autophagy-Lysosome Function Drives Pancreatic Cancer Metabolism. Nature 2015, 524, 361–365. [Google Scholar] [CrossRef] [PubMed]
  234. Calcagnì, A.; Kors, L.; Verschuren, E.; de Cegli, R.; Zampelli, N.; Nusco, E.; Confalonieri, S.; Bertalot, G.; Pece, S.; Settembre, C.; et al. Modelling TFE Renal Cell Carcinoma in Mice Reveals a Critical Role of WNT Signaling. eLife 2016, 5, e17047. [Google Scholar] [CrossRef] [Green Version]
  235. Blessing, A.M.; Rajapakshe, K.; Reddy Bollu, L.; Shi, Y.; White, M.A.; Pham, A.H.; Lin, C.; Jonsson, P.; Cortes, C.J.; Cheung, E.; et al. Transcriptional Regulation of Core Autophagy and Lysosomal Genes by the Androgen Receptor Promotes Prostate Cancer Progression. Autophagy 2017, 13, 506–521. [Google Scholar] [CrossRef] [PubMed]
  236. Wei, H.; Wang, C.; Croce, C.M.; Guan, J.L. P62/SQSTM1 Synergizes with Autophagy for Tumor Growth in Vivo. Genes Dev. 2014, 28, 1204–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Giatromanolaki, A.; Kalamida, D.; Sivridis, E.; Karagounis, I.V.; Gatter, K.C.; Harris, A.L.; Koukourakis, M.I. Increased Expression of Transcription Factor EB (TFEB) Is Associated with Autophagy, Migratory Phenotype and Poor Prognosis in Non-Small Cell Lung Cancer. Lung Cancer 2015, 90, 98–105. [Google Scholar] [CrossRef]
  238. Zhao, B.; Dierichs, L.; Gu, J.N.; Trajkovic-Arsic, M.; Axel Hilger, R.; Savvatakis, K.; Vega-Rubin-de-Celis, S.; Liffers, S.T.; Peña-Llopis, S.; Behrens, D.; et al. TFEB-Mediated Lysosomal Biogenesis and Lysosomal Drug Sequestration Confer Resistance to MEK Inhibition in Pancreatic Cancer. Cell Death Discov. 2020, 6, 12. [Google Scholar] [CrossRef] [Green Version]
  239. Kim, J.H.; Lee, J.; Cho, Y.R.; Lee, S.Y.; Sung, G.J.; Shin, D.M.; Choi, K.C.; Son, J. TFEB Supports Pancreatic Cancer Growth through the Transcriptional Regulation of Glutaminase. Cancers 2021, 13, 483. [Google Scholar] [CrossRef]
  240. Li, S.; Song, Y.; Quach, C.; Guo, H.; Jang, G.B.; Maazi, H.; Zhao, S.; Sands, N.A.; Liu, Q.; In, G.K.; et al. Transcriptional Regulation of Autophagy-Lysosomal Function in BRAF-Driven Melanoma Progression and Chemoresistance. Nat. Commun. 2019, 10, 1693. [Google Scholar] [CrossRef] [Green Version]
  241. Carreira, S.; Goodall, J.; Denat, L.; Rodriguez, M.; Nuciforo, P.; Hoek, K.S.; Testori, A.; Larue, L.; Goding, C.R. Mitf Regulation of Dia1 Controls Melanoma Proliferation and Invasiveness. Genes Dev. 2006, 20, 3426–3439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Cheli, Y.; Giuliano, S.; Fenouille, N.; Allegra, M.; Hofman, V.; Hofman, P.; Bahadoran, P.; Lacour, J.-P.; Tartare-Deckert, S.; Bertolotto, C.; et al. Hypoxia and MITF Control Metastatic Behaviour in Mouse and Human Melanoma Cells. Oncogene 2012, 31, 2461–2470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Aida, S.; Sonobe, Y.; Tanimura, H.; Oikawa, N.; Yuhki, M.; Sakamoto, H.; Mizuno, T. MITF Suppression Improves the Sensitivity of Melanoma Cells to a BRAF Inhibitor. Cancer Lett. 2017, 409, 116–124. [Google Scholar] [CrossRef] [PubMed]
  244. Garraway, L.A.; Widlund, H.R.; Rubin, M.A.; Getz, G.; Berger, A.J.; Ramaswamy, S.; Beroukhim, R.; Milner, D.A.; Granter, S.R.; Du, J.; et al. Integrative Genomic Analyses Identify MITF as a Lineage Survival Oncogene Amplified in Malignant Melanoma. Nature 2005, 436, 117–122. [Google Scholar] [CrossRef] [PubMed]
  245. Cheli, Y.; Guiliano, S.; Botton, T.; Rocchi, S.; Hofman, V.; Hofman, P.; Bahadoran, P.; Bertolotto, C.; Ballotti, R. Mitf Is the Key Molecular Switch between Mouse or Human Melanoma Initiating Cells and Their Differentiated Progeny. Oncogene 2011, 30, 2307–2318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Bourseguin, J.; Bonet, C.; Renaud, E.; Pandiani, C.; Boncompagni, M.; Giuliano, S.; Pawlikowska, P.; Karmous-Benailly, H.; Ballotti, R.; Rosselli, F.; et al. FANCD2 Functions as a Critical Factor Downstream of MiTF to Maintain the Proliferation and Survival of Melanoma Cells. Sci. Rep. 2016, 6, 36539. [Google Scholar] [CrossRef] [Green Version]
  247. Kasitinon, S.Y.; Eskiocak, U.; Martin, M.; Bezwada, D.; Khivansara, V.; Tasdogan, A.; Zhao, Z.; Mathews, T.; Aurora, A.B.; Morrison, S.J. TRPML1 Promotes Protein Homeostasis in Melanoma Cells by Negatively Regulating MAPK and MTORC1 Signaling. Cell Rep. 2019, 28, 2293–2305. [Google Scholar] [CrossRef] [Green Version]
  248. Xing, Y.; Wei, X.; Liu, Y.; Wang, M.-M.; Sui, Z.; Wang, X.; Zhu, W.; Wu, M.; Lu, C.; Fei, Y.-H.; et al. Autophagy Inhibition Mediated by MCOLN1/TRPML1 Suppresses Cancer Metastasis via Regulating a ROS-Driven TP53/P53 Pathway. Autophagy 2021, 1–23. [Google Scholar] [CrossRef]
  249. Nguyen, O.N.P.; Grimm, C.; Schneider, L.S.; Chao, Y.K.; Atzberger, C.; Bartel, K.; Watermann, A.; Ulrich, M.; Mayr, D.; Wahl-Schott, C.; et al. Two-Pore Channel Function Is Crucial for the Migration of Invasive Cancer Cells. Cancer Res. 2017, 77, 1427–1438. [Google Scholar] [CrossRef] [Green Version]
  250. Faviaa, A.; Desiderib, M.; Gambaraa, G.; D’Alessioa, A.; Ruas, M.; Esposito, B.; del Bufalo, D.; Parrington, J.; Ziparoa, E.; Palombia, F.; et al. VEGF-Induced Neoangiogenesis Is Mediated by NAADP and Two-Pore Channel-2-Dependent Ca2+ Signaling. Proc. Natl. Acad. Sci. USA 2014, 111, 4706–4715. [Google Scholar] [CrossRef] [Green Version]
  251. Favia, A.; Pafumi, I.; Desideri, M.; Padula, F.; Montesano, C.; Passeri, D.; Nicoletti, C.; Orlandi, A.; del Bufalo, D.; Sergi, M.; et al. NAADP-Dependent Ca(2+) Signaling Controls Melanoma Progression, Metastatic Dissemination and Neoangiogenesis. Sci. Rep. 2016, 6, 18925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Yu, H.; Xie, M.; Meng, Z.; Lo, C.-Y.; Chan, F.L.; Jiang, L.; Meng, X.; Yao, X. Endolysosomal Ion Channel MCOLN2 (Mucolipin-2) Promotes Prostate Cancer Progression via IL-1β/NF-ΚB Pathway. Br. J. Cancer 2021, 125, 1420–1431. [Google Scholar] [CrossRef] [PubMed]
  253. Kuro-o, M.; Matsumura, Y.; Aizawa, H.; Kawaguchi, H.; Suga, T.; Utsugi, T.; Ohyama, Y.; Kurabayashi, M.; Kaname, T.; Kume, E.; et al. Mutation of the Mouse Klotho Gene Leads to a Syndrome Resembling Ageing. Nature 1997, 390, 45–51. [Google Scholar] [CrossRef] [PubMed]
  254. Nagai, T.; Yamada, K.; Kim, H.C.; Kim, Y.S.; Noda, Y.; Imura, A.; Nabeshima, Y.-I.; Nabeshima, T. Cognition Impairment in the Genetic Model of Aging Klotho Gene Mutant Mice: A Role of Oxidative Stress. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2003, 17, 50–52. [Google Scholar] [CrossRef]
  255. Kurosu, H.; Yamamoto, M.; Clark, J.D.; Pastor, J.V.; Nandi, A.; Gurnani, P.; McGuinness, O.P.; Chikuda, H.; Yamaguchi, M.; Kawaguchi, H.; et al. Physiology: Suppression of Aging in Mice by the Hormone Klotho. Science 2005, 309, 1829–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Kuro-o, M. Klotho and Aging. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2009, 1790, 1049–1058. [Google Scholar] [CrossRef]
  257. Dubal, D.B.; Yokoyama, J.S.; Zhu, L.; Broestl, L.; Worden, K.; Wang, D.; Sturm, V.E.; Kim, D.; Klein, E.; Yu, G.Q.; et al. Life Extension Factor Klotho Enhances Cognition. Cell Rep. 2014, 7, 1065–1076. [Google Scholar] [CrossRef] [Green Version]
  258. Arking, D.E.; Krebsova, A.; Macek, M.; Macek, M.; Arking, A.; Mian, I.S.; Fried, L.; Hamosh, A.; Dey, S.; McIntosh, I.; et al. Association of Human Aging with a Functional Variant of Klotho. Proc. Natl. Acad. Sci. USA 2002, 99, 856–861. [Google Scholar] [CrossRef] [Green Version]
  259. Arking, D.E.; Becker, D.M.; Yanek, L.R.; Fallin, D.; Judge, D.P.; Moy, T.F.; Becker, L.C.; Dietz, H.C. KLOTHO Allele Status and the Risk of Early-Onset Occult Coronary Artery Disease. Am. J. Hum. Genet. 2003, 72, 1154–1161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  260. Arking, D.E.; Atzmon, G.; Arking, A.; Barzilai, N.; Dietz, H.C. Association between a Functional Variant of the KLOTHO Gene and High-Density Lipoprotein Cholesterol, Blood Pressure, Stroke, and Longevity. Circ. Res. 2005, 96, 412–418. [Google Scholar] [CrossRef] [Green Version]
  261. Matsumura, Y.; Aizawa, H.; Shiraki-Iida, T.; Nagai, R.; Kuro-O, M.; Nabeshima, Y.I. Identification of the Human Klotho Gene and Its Two Transcripts Encoding Membrane and Secreted Klotho Protein. Biochem. Biophys. Res. Commun. 1998, 242, 626–630. [Google Scholar] [CrossRef] [PubMed]
  262. Shiraki-Iida, T.; Aizawa, H.; Matsumura, Y.; Sekine, S.; Iida, A.; Anazawa, H.; Nagai, R.; Kuro-O, M.; Nabeshima, Y.I. Structure of the Mouse Klotho Gene and Its Two Transcripts Encoding Membrane and Secreted Protein. FEBS Lett. 1998, 424, 6–10. [Google Scholar] [CrossRef] [Green Version]
  263. Cha, S.K.; Ortega, B.; Kurosu, H.; Rosenblatt, K.P.; Kuro-o, M.; Huang, C.L. Removal of Sialic Acid Involving Klotho Causes Cell-Surface Retention of TRPV5 Channel via Binding to Galectin-1. Proc. Natl. Acad. Sci. USA 2008, 105, 9805–9810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Chen, C.D.; Podvin, S.; Gillespie, E.; Leeman, S.E.; Abraham, C.R. Insulin Stimulates the Cleavage and Release of the Extracellular Domain of Klotho by ADAM10 and ADAM17. Proc. Natl. Acad. Sci. USA 2007, 104, 19796–19801. [Google Scholar] [CrossRef] [Green Version]
  265. Kurosu, H.; Ogawa, Y.; Miyoshi, M.; Yamamoto, M.; Nandi, A.; Rosenblatt, K.P.; Baum, M.G.; Schiavi, S.; Hu, M.C.; Moe, O.W.; et al. Regulation of Fibroblast Growth Factor-23 Signaling by Klotho. J. Biol. Chem. 2006, 281, 6120–6123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Chang, Q.; Hoefs, S.; van der Kemp, A.W.; Topala, C.N.; Bindels, R.J.; Hoenderop, J.G. Cell Signalling: The β-Glucuronidase Klotho Hydrolyzes and Activates the TRPV5 Channel. Science 2005, 310, 490–493. [Google Scholar] [CrossRef]
  267. Lu, P.; Boros, S.; Chang, Q.; Bindels, R.J.; Hoenderop, J.G. The Beta-Glucuronidase Klotho Exclusively Activates the Epithelial Ca2+ Channels TRPV5 and TRPV6. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc.-Eur. Ren. Assoc. 2008, 23, 3397–3402. [Google Scholar] [CrossRef]
  268. Wolf, M.T.F.; An, S.W.; Nie, M.; Bal, M.S.; Huang, C.L. Klotho Up-Regulates Renal Calcium Channel Transient Receptor Potential Vanilloid 5 (TRPV5) by Intra- and Extracellular N-Glycosylation-Dependent Mechanisms. J. Biol. Chem. 2014, 289, 35849–35857. [Google Scholar] [CrossRef] [Green Version]
  269. Takumida, M.; Ishibashi, T.; Hamamoto, T.; Hirakawa, K.; Anniko, M. Age-Dependent Changes in the Expression of Klotho Protein, TRPV5 and TRPV6 in Mouse Inner Ear. Acta Oto-Laryngol. 2009, 129, 1340–1350. [Google Scholar] [CrossRef] [PubMed]
  270. Hoenderop, J.G.J.; van Leeuwen, J.P.T.M.; van der Eerden, B.C.J.; Kersten, F.F.J.; van derKemp, A.W.C.M.; Mérillat, A.-M.; Waarsing, J.H.; Rossier, B.C.; Vallon, V.; Hummler, E.; et al. Renal Ca2+ Wasting, Hyperabsorption, and Reduced Bone Thickness in Mice Lacking TRPV5. J. Clin. Investig. 2003, 112, 1906–1914. [Google Scholar] [CrossRef]
  271. Huang, C.L. Regulation of Ion Channels by Secreted Klotho: Mechanisms and Implications. Kidney Int. 2010, 77, 855–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  272. Imura, A.; Tsuji, Y.; Murata, M.; Maeda, R.; Kubota, K.; Iwano, A.; Obuse, C.; Togashi, K.; Tominaga, M.; Kita, N.; et al. α-Klotho as a Regulator of Calcium Homeostasis. Science 2007, 316, 1615–1618. [Google Scholar] [CrossRef] [PubMed]
  273. Almilaji, A.; Pakladok, T.; Munõz, C.; Elvira, B.; Sopjani, M.; Lang, F. Upregulation of KCNQ1/KCNE1 K+ Channels by Klotho. Channels 2014, 8, 222–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Xie, J.; Cha, S.K.; An, S.W.; Kuro-O, M.; Birnbaumer, L.; Huang, C.L. Cardioprotection by Klotho through Downregulation of TRPC6 Channels in the Mouse Heart. Nat. Commun. 2012, 3, 1238. [Google Scholar] [CrossRef] [Green Version]
  275. Wu, Y.L.; Xie, J.; An, S.W.; Oliver, N.; Barrezueta, N.X.; Lin, M.H.; Birnbaumer, L.; Huang, C.L. Inhibition of TRPC6 Channels Ameliorates Renal Fibrosis and Contributes to Renal Protection by Soluble Klotho. Kidney Int. 2017, 91, 830–841. [Google Scholar] [CrossRef] [Green Version]
  276. Xie, J.; An, S.W.; Jin, X.; Gui, Y.; Huang, C.L. Munc13 Mediates Klotho-Inhibitable Diacylglycerol-Stimulated Exocytotic Insertion of Pre-Docked TRPC6 Vesicles. PLoS ONE 2020, 15, e0229799. [Google Scholar] [CrossRef] [PubMed]
  277. Gillooly, J.F.; Brown, J.H.; West, G.B.; Savage, V.M.; Charnov, E.L. Effects of Size and Temperature on Metabolic Rate. Science 2001, 293, 2248–2251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Keil, G.; Cummings, E.; de Magalhães, J.P. Being Cool: How Body Temperature Influences Ageing and Longevity. Biogerontology 2015, 16, 383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  279. Liu, R.K.; Walford, R.L. The Effect of Lowered Body Temperature on Lifespan and Immune and Non-Immune Processes. Gerontologia 1972, 18, 363–388. [Google Scholar] [CrossRef]
  280. Loeb, J.; Northrop, J.H. Is There a Temperature Coefficient for the Duration of Life? Proc. Natl. Acad. Sci. USA 1916, 2, 456–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  281. Duffy, P.H.; Feuers, R.J.; Leakey, J.A.; Nakamura, K.D.; Turturro, A.; Hart, R.W. Effect of Chronic Caloric Restriction on Physiological Variables Related to Energy Metabolism in the Male Fischer 344 Rat. Mech. Ageing Dev. 1989, 48, 117–133. [Google Scholar] [CrossRef]
  282. Kim, K.; Lin, Y.R.; Park, Y. Enhancement of Stress Resistances and Downregulation of Imd Pathway by Lower Developmental Temperature in Drosophila Melanogaster. Exp. Gerontol. 2010, 45, 984–987. [Google Scholar] [CrossRef] [PubMed]
  283. Hayflick, L. Biological Aging Is No Longer an Unsolved Problem. Ann. N. Y. Acad. Sci. 2007, 1100, 1–13. [Google Scholar] [CrossRef] [PubMed]
  284. Rikke, B.A.; Johnson, T.E. Lower Body Temperature as a Potential Mechanism of Life Extension in Homeotherms. Exp. Gerontol. 2004, 39, 927–930. [Google Scholar] [CrossRef] [PubMed]
  285. Xiao, R.; Zhang, B.; Dong, Y.; Gong, J.; Xu, T.; Liu, J.; Xu, X.Z.S. A Genetic Program Promotes C. elegans Longevity at Cold Temperatures via a Thermosensitive TRP Channel. Cell 2013, 152, 806–817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Zhang, B.; Xiao, R.; Ronan, E.A.; He, Y.; Hsu, A.L.; Liu, J.; Xu, X.Z.S. Environmental Temperature Differentially Modulates C. elegans Longevity through a Thermosensitive TRP Channel. Cell Rep. 2015, 11, 1414–1424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Riera, C.E.; Huising, M.O.; Follett, P.; Leblanc, M.; Halloran, J.; van Andel, R.; de Magalhaes Filho, C.D.; Merkwirth, C.; Dillin, A. TRPV1 Pain Receptors Regulate Longevity and Metabolism by Neuropeptide Signaling. Cell 2014, 157, 1023–1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Lee, B.H.; Ashrafi, K. A TRPV Channel Modulates C. elegans Neurosecretion, Larval Starvation Survival, and Adult Lifespan. PLoS Genet. 2008, 4, e1000213. [Google Scholar] [CrossRef] [PubMed]
  289. Calderwood, S.K.; Murshid, A.; Prince, T. The Shock of Aging: Molecular Chaperones and the Heat Shock Response in Longevity and Aging—A Mini-Review. Gerontology 2009, 55, 550–558. [Google Scholar] [CrossRef] [Green Version]
  290. Volovik, Y.; Maman, M.; Dubnikov, T.; Bejerano-Sagie, M.; Joyce, D.; Kapernick, E.A.; Cohen, E.; Dillin, A. Temporal Requirements of Heat Shock Factor-1 for Longevity Assurance. Aging Cell 2012, 11, 491–499. [Google Scholar] [CrossRef] [Green Version]
  291. Seo, K.; Choi, E.; Lee, D.; Jeong, D.E.; Jang, S.K.; Lee, S.J. Heat Shock Factor 1 Mediates the Longevity Conferred by Inhibition of TOR and Insulin/IGF-1 Signaling Pathways in C. elegans. Aging Cell 2013, 12, 1073–1081. [Google Scholar] [CrossRef] [PubMed]
  292. Prahlad, V.; Cornelius, T.; Morimoto, R.I. Regulation of the Cellular Heat Shock Response in Caenorhabditis Elegans by Thermosensory Neurons. Science 2008, 320, 811. [Google Scholar] [CrossRef] [Green Version]
  293. Chen, Y.C.; Chen, H.J.; Tseng, W.C.; Hsu, J.M.; Huang, T.T.; Chen, C.H.; Pan, C.L. A C. elegans Thermosensory Circuit Regulates Longevity through Crh-1/CREB-Dependent Flp-6 Neuropeptide Signaling. Dev. Cell 2016, 39, 209–223. [Google Scholar] [CrossRef] [Green Version]
  294. Silva, M.C.; Amaral, M.D.; Morimoto, R.I. Neuronal Reprograming of Protein Homeostasis by Calcium-Dependent Regulation of the Heat Shock Response. PLoS Genet. 2013, 9, e1003711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Waterson, M.J.; Chung, B.Y.; Harvanek, Z.M.; Ostojic, I.; Alcedo, J.; Pletcher, S.D. Water Sensor Ppk28 Modulates Drosophila Lifespan and Physiology through AKH Signaling. Proc. Natl. Acad. Sci. USA 2014, 111, 8137–8142. [Google Scholar] [CrossRef] [Green Version]
  296. Zelle, K.M.; Lu, B.; Pyfrom, S.C.; Ben-Shahar, Y. The Genetic Architecture of Degenerin/Epithelial Sodium Channels in Drosophila. G3 Genes Genomes Genet. 2013, 3, 441–450. [Google Scholar] [CrossRef]
  297. Chen, Z.; Wang, Q.; Wang, Z. The Amiloride-Sensitive Epithelial Na+ Channel PPK28 Is Essential for Drosophila Gustatory Water Reception. J. Neurosci. Off. J. Soc. Neurosci. 2010, 30, 6247–6252. [Google Scholar] [CrossRef] [PubMed]
  298. Jiang, H.C.; Hsu, J.M.; Yen, C.P.; Chao, C.C.; Chen, R.H.; Pan, C.L. Neural Activity and CaMKII Protect Mitochondria from Fragmentation in Aging Caenorhabditis Elegans Neurons. Proc. Natl. Acad. Sci. USA 2015, 112, 8768–8773. [Google Scholar] [CrossRef] [Green Version]
  299. Colbert, H.A.; Smith, T.L.; Bargmann, C.I. OSM-9, a Novel Protein with Structural Similarity to Channels, Is Required for Olfaction, Mechanosensation, and Olfactory Adaptation in Caenorhabditis Elegans. J. Neurosci. Off. J. Soc. Neurosci. 1997, 17, 8259–8269. [Google Scholar] [CrossRef] [Green Version]
  300. Lee, E.C.; Kim, H.; Ditano, J.; Manion, D.; King, B.L.; Strange, K. Abnormal Osmotic Avoidance Behavior in C. elegans Is Associated with Increased Hypertonic Stress Resistance and Improved Proteostasis. PLoS ONE 2016, 11, e0154156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  301. Odell, K.M.C. The Effect of the Inactive Mutation on Longevity, Sex, Rhythm and Resistance to p-Cresol in Drosophila Melanogaster. Heredity 1993, 70, 393–399. [Google Scholar] [CrossRef] [Green Version]
  302. Gong, Z. Two Interdependent TRPV Channel Subunits, Inactive and Nanchung, Mediate Hearing in Drosophila. J. Neurosci. 2004, 24, 9059–9066. [Google Scholar] [CrossRef] [Green Version]
  303. Gendron, C.M.; Kuo, T.H.; Harvanek, Z.M.; Chung, B.Y.; Yew, J.Y.; Dierick, H.A.; Pletcher, S.D. Drosophila Life Span and Physiology Are Modulated by Sexual Perception and Reward. Science 2014, 343, 544–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Lu, B.; LaMora, A.; Sun, Y.; Welsh, M.J.; Ben-Shahar, Y. Ppk23-Dependent Chemosensory Functions Contribute to Courtship Behavior in Drosophila Melanogaster. PLoS Genet. 2012, 8, e1002587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Vijayan, V.; Thistle, R.; Liu, T.; Starostina, E.; Pikielny, C.W. Drosophila Pheromone-Sensing Neurons Expressing the Ppk25 Ion Channel Subunit Stimulate Male Courtship and Female Receptivity. PLoS Genet. 2014, 10, e1004238. [Google Scholar] [CrossRef] [PubMed]
  306. Libert, S.; Zwiener, J.; Chu, X.; VanVoorhies, W.; Roman, G.; Pletcher, S.D. Regulation of Drosophila Life Span by Olfaction and Food-Derived Odors. Science 2007, 315, 1133–1137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Wicher, D.; Schäfer, R.; Bauernfeind, R.; Stensmyr, M.C.; Heller, R.; Heinemann, S.H.; Hansson, B.S. Drosophila Odorant Receptors Are Both Ligand-Gated and Cyclic-Nucleotide-Activated Cation Channels. Nature 2008, 452, 1007–1011. [Google Scholar] [CrossRef]
  308. Sato, K.; Pellegrino, M.; Nakagawa, T.; Nakagawa, T.; Vosshall, L.B.; Touhara, K. Insect Olfactory Receptors Are Heteromeric Ligand-Gated Ion Channels. Nature 2008, 452, 1002–1006. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Regulation of cellular and organismal aging by neuronal excitability. (A) Sequencing of RNA extracted from the frontal cortices of deceased individuals revealed that expression of genes related to neuronal excitability and excitatory synaptic transmission was relatively lower in long-lived humans (i.e., those who died between the ages of 85–101 years) compared to that in people who died between the ages of 70–80 years. (B) In both C. elegans and Drosophila, neuronal excitability is inversely correlated with longevity, and pharmacological repression of neuronal activity extends lifespan. In non-excitable cells such as fibroblasts, increased electrical excitability is associated with precocious onset of senescence. Images created with BioRender.com (accessed on 20 January 2022).
Figure 1. Regulation of cellular and organismal aging by neuronal excitability. (A) Sequencing of RNA extracted from the frontal cortices of deceased individuals revealed that expression of genes related to neuronal excitability and excitatory synaptic transmission was relatively lower in long-lived humans (i.e., those who died between the ages of 85–101 years) compared to that in people who died between the ages of 70–80 years. (B) In both C. elegans and Drosophila, neuronal excitability is inversely correlated with longevity, and pharmacological repression of neuronal activity extends lifespan. In non-excitable cells such as fibroblasts, increased electrical excitability is associated with precocious onset of senescence. Images created with BioRender.com (accessed on 20 January 2022).
Cells 11 01180 g001
Figure 2. The PLCβ–IP3R signaling axis. Stimulation of Gαq-coupled GPCRs leads to the activation of PLCβ. PLCβ hydrolyzes the membrane phosphoinositide, PIP2, to generate the second messengers, diacylglycerol (DAG) and IP3. While DAG is retained in the plasma membrane, IP3 diffuses through the cytosol and activates IP3Rs, which are localized to the ER membrane. Activation of IP3Rs results in the release of ER Ca2+. Image created with BioRender.com (accessed on 20 January 2022).
Figure 2. The PLCβ–IP3R signaling axis. Stimulation of Gαq-coupled GPCRs leads to the activation of PLCβ. PLCβ hydrolyzes the membrane phosphoinositide, PIP2, to generate the second messengers, diacylglycerol (DAG) and IP3. While DAG is retained in the plasma membrane, IP3 diffuses through the cytosol and activates IP3Rs, which are localized to the ER membrane. Activation of IP3Rs results in the release of ER Ca2+. Image created with BioRender.com (accessed on 20 January 2022).
Cells 11 01180 g002
Figure 3. Consequences and regulators of neuronal excitability. (A) Left, at resting membrane potentials, PLCβ hydrolyzes the PIP2 moieties associated with the enzyme. Right, plasma membrane depolarization results in enhanced PIP2–PLCβ clustering. Subsequent, stimulation of PLCβ-coupled receptors in depolarized neurons results in the augmentation of PIP2 hydrolysis, IP3 production, and IP3R-dependent ER Ca2+ release. (B) Left, REST is a master regulator of membrane excitability in healthy neurons. Nuclear translocation of REST allows for the repression of RE1-containing gene such as those encoding channels and pumps. Repression of these genes is necessary for the proper maintenance of membrane potential and excitability. Right, in neurodegenerative disease, REST is sequestered away from the nucleus resulting in derepression of target genes and the augmentation of excitability and genes related to synaptic transmission. Images created with BioRender.com (accessed 25 January 2022).
Figure 3. Consequences and regulators of neuronal excitability. (A) Left, at resting membrane potentials, PLCβ hydrolyzes the PIP2 moieties associated with the enzyme. Right, plasma membrane depolarization results in enhanced PIP2–PLCβ clustering. Subsequent, stimulation of PLCβ-coupled receptors in depolarized neurons results in the augmentation of PIP2 hydrolysis, IP3 production, and IP3R-dependent ER Ca2+ release. (B) Left, REST is a master regulator of membrane excitability in healthy neurons. Nuclear translocation of REST allows for the repression of RE1-containing gene such as those encoding channels and pumps. Repression of these genes is necessary for the proper maintenance of membrane potential and excitability. Right, in neurodegenerative disease, REST is sequestered away from the nucleus resulting in derepression of target genes and the augmentation of excitability and genes related to synaptic transmission. Images created with BioRender.com (accessed 25 January 2022).
Cells 11 01180 g003
Figure 4. Interorganellar Ca2+ transfer between ER and mitochondria. ER and mitochondrial outer membrane are physically tethered at sites called mitochondria associated membranes (MAMs). MAMs constitute the regions of metabolite exchange between the two organelles. A fraction of IP3Rs localize to MAMs, where they mediate the transfer of Ca2+ from the ER lumen to the perimitochondrial region. The mitochondrial Ca2+ uniporter resides in the inner mitochondrial membrane, and is responsible for the transfer of Ca2+ into the mitochondrial matrix. Physiological Ca2+ elevations in the matrix are necessary for mitochondrial bioenergetics and the production of ATP via TCA and ETC. Ca2+ overload, however, results in pathology and eventual cell death. Image created with BioRender.com (accessed 24 January 2022).
Figure 4. Interorganellar Ca2+ transfer between ER and mitochondria. ER and mitochondrial outer membrane are physically tethered at sites called mitochondria associated membranes (MAMs). MAMs constitute the regions of metabolite exchange between the two organelles. A fraction of IP3Rs localize to MAMs, where they mediate the transfer of Ca2+ from the ER lumen to the perimitochondrial region. The mitochondrial Ca2+ uniporter resides in the inner mitochondrial membrane, and is responsible for the transfer of Ca2+ into the mitochondrial matrix. Physiological Ca2+ elevations in the matrix are necessary for mitochondrial bioenergetics and the production of ATP via TCA and ETC. Ca2+ overload, however, results in pathology and eventual cell death. Image created with BioRender.com (accessed 24 January 2022).
Cells 11 01180 g004
Figure 5. Uncoupling protein (UCP) mediated dissipation of the mitochondrial proton (H+) gradient. ETC involves the transfers of electrons (e) from reducing equivalents (NADH and FADH2) to molecular oxygen (O2). e transport is coupled with the movement of H+ from the mitochondrial matrix to the intermembrane space (IMS). Subsequent dissipation of the electrochemical gradient through ATP synthase drives the production of ATP. Transfer of e to O2 also leads to the generation of superoxide ions and ROS. By mediating the transport of H+ down their electrochemical gradient, UCPs dissipate the protonmotive force. Loss of protonmotive force results in the production of heat and prevents ATP production. Upon UCP-mediated H+ leak, diminished transfer of e to O2 mitigates ROS production. Image created with BioRender.com (accessed 24 January 2022).
Figure 5. Uncoupling protein (UCP) mediated dissipation of the mitochondrial proton (H+) gradient. ETC involves the transfers of electrons (e) from reducing equivalents (NADH and FADH2) to molecular oxygen (O2). e transport is coupled with the movement of H+ from the mitochondrial matrix to the intermembrane space (IMS). Subsequent dissipation of the electrochemical gradient through ATP synthase drives the production of ATP. Transfer of e to O2 also leads to the generation of superoxide ions and ROS. By mediating the transport of H+ down their electrochemical gradient, UCPs dissipate the protonmotive force. Loss of protonmotive force results in the production of heat and prevents ATP production. Upon UCP-mediated H+ leak, diminished transfer of e to O2 mitigates ROS production. Image created with BioRender.com (accessed 24 January 2022).
Cells 11 01180 g005
Figure 6. Transcriptional regulation of genes encoding endolysosomal ion channels. Within the cell nucleus, TFEB, and related transcription factors such as TFE3 and MITF, bind to the promoters of genes that encode autophagy and endolysosomal proteins. Figure shows dephosphorylated TFEB bound to the promoters of MCOLN1 and TPCN2, which encode TRPML1 and TPC2, respectively. Phosphorylated TFEB is retained in the cytosol leading to diminished expression of endolysosomal genes. TRPML1 and TPC2 are endolysosomal cation channels that regulate nucleocytoplasmic translocation of TFEB activity in different ways. By driving autophagic and endolysosomal flux, TRPML1 and TPC2 are needed for mTORC1 activation, which in turn, phosphorylates TFEB leading to retention of the transcription factor in the cytosol. On the other hand, release of endolysosomal Ca2+ via TRPML1 and TPC2 activates calcineurin (CaN), which dephosphorylates TFEB, and thereby, promotes the translocation of the transcription factor into the nucleus. Image created with BioRender.com (accessed 24 January 2022).
Figure 6. Transcriptional regulation of genes encoding endolysosomal ion channels. Within the cell nucleus, TFEB, and related transcription factors such as TFE3 and MITF, bind to the promoters of genes that encode autophagy and endolysosomal proteins. Figure shows dephosphorylated TFEB bound to the promoters of MCOLN1 and TPCN2, which encode TRPML1 and TPC2, respectively. Phosphorylated TFEB is retained in the cytosol leading to diminished expression of endolysosomal genes. TRPML1 and TPC2 are endolysosomal cation channels that regulate nucleocytoplasmic translocation of TFEB activity in different ways. By driving autophagic and endolysosomal flux, TRPML1 and TPC2 are needed for mTORC1 activation, which in turn, phosphorylates TFEB leading to retention of the transcription factor in the cytosol. On the other hand, release of endolysosomal Ca2+ via TRPML1 and TPC2 activates calcineurin (CaN), which dephosphorylates TFEB, and thereby, promotes the translocation of the transcription factor into the nucleus. Image created with BioRender.com (accessed 24 January 2022).
Cells 11 01180 g006
Figure 7. Effect of low-temperature on the lifespan of C. elegans is mediated by the cold-sensitive ion channel, TRPA-1. TRPA-1 is a cold sensitive ion channel that resides in the plasma membrane of C. elegans neurons and intestinal cells. Activation of the channels at 15 °C, results in cytosolic Ca2+ elevation and the activation of a signaling cascade that culminates in DAF-16/FOXO-mediated gene transcription, which in turn, promotes worm longevity. Image created with BioRender.com (accessed 27 January 2022).
Figure 7. Effect of low-temperature on the lifespan of C. elegans is mediated by the cold-sensitive ion channel, TRPA-1. TRPA-1 is a cold sensitive ion channel that resides in the plasma membrane of C. elegans neurons and intestinal cells. Activation of the channels at 15 °C, results in cytosolic Ca2+ elevation and the activation of a signaling cascade that culminates in DAF-16/FOXO-mediated gene transcription, which in turn, promotes worm longevity. Image created with BioRender.com (accessed 27 January 2022).
Cells 11 01180 g007
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Venkatachalam, K. Regulation of Aging and Longevity by Ion Channels and Transporters. Cells 2022, 11, 1180. https://doi.org/10.3390/cells11071180

AMA Style

Venkatachalam K. Regulation of Aging and Longevity by Ion Channels and Transporters. Cells. 2022; 11(7):1180. https://doi.org/10.3390/cells11071180

Chicago/Turabian Style

Venkatachalam, Kartik. 2022. "Regulation of Aging and Longevity by Ion Channels and Transporters" Cells 11, no. 7: 1180. https://doi.org/10.3390/cells11071180

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