Abstract
Acidity, generated in hypoxia or hypermetabolic states, perturbs homeostasis and is a feature of solid tumors. That acid peripherally disperses lysosomes is a three-decade-old observation, yet one little understood or appreciated. However, recent work has recognized the inhibitory impact this spatial redistribution has on the key regulator of metabolism mTORC1. This mechanism argues for a paradigm shift in localization of mTORC1 activator RHEB, a conclusion multiple others have now independently reached. Thus, mTORC1, known to sense amino acids, mitogens, and energy to restrict biosynthesis to times of adequate resources, also senses pH and, via dampened mTOR-governed synthesis of clock proteins, regulates the circadian clock to achieve concerted responses to metabolic stress. While this may allow cancer to endure metabolic deprivation, immune cell mTOR signaling likewise exhibits pH sensitivity, suggesting suppression of antitumor immune function by solid tumor acidity may additionally fuel cancers, an obstacle potentially reversible through therapeutic pH manipulation.
Keywords: acidity, cancer immunity, circadian clock, lysosome trafficking, mTOR, pH, RHEB
Graphical Abstract
Multiple studies now indicate mTORC1 senses intracellular pH. Acid drives lysosome redistribution separating bound mTORC1 from its activator RHEB. This suggests RHEB resides on non-lysosomal endomembranes in contrast to prevailing models. We review emerging reports regarding RHEB localization and discuss ways to translate mTOR’s pH sensing into cancer immunotherapy.

1. Introduction:
Acidity of cells and tissues manifests as yellowed cell culture media in the laboratory, decreased hyperpolarized carbon-13 bicarbonate magnetic resonance signal within animal tumors,[1] and the altered mentation of patients in the clinic. This acidity reflects the generation of protons by metabolism. The level of acidity is quantified as pH, the negative log10 of the proton concentration. The major extracellular buffering system in the body involves interchange between carbon dioxide (CO2), carbonic acid (H2CO3), and bicarbonate (CO3−). The relationship of pH to this bicarbonate buffering system is expressed by the Henderson-Hasselbach equation: pH = pKa + log10([HCO3−]/[H2CO3]), where pKa is the dissociation constant for carbonic acid and [H2CO3] can be readily calculated from the partial pressure of CO2 using Henry’s law. As taught in basic chemistry, a pH of 7 is defined as neutral; greater than this is alkaline and less is acidic. Human blood—and, through compartment exchange, interstitial fluid—is normally maintained in tight range centered at pH 7.4.
2. Solid tumors are acidic
The generation of acid relevant to cancer biology is attributable largely to glycolysis. Respiration’s production of carbon dioxide, which in water hydrates to carbonic acid that dissociates to release a proton, accounts for a smaller acid load (Figure 1).[2] Solid tumors tend to outstrip their vasculature rendering regions hypoxic.[3–5] Lack of oxygen—the final electron acceptor in oxidative phosphorylation—precludes respiration. Therefore, in hypoxia, cells upregulate glycolysis, metabolizing glucose to lactic acid as an oxygen-independent means to generate ATP. Indeed, Louis Pasteur noted this ability to use oxygen levels to toggle metabolism between respiration and anaerobic glycolysis when he studied ethanol fermentation in yeast, a pathway analogous to lactic acid production in mammalian cells.[6] Notably, glycolysis is considerably more acidifying than oxidative phosphorylation to generate the same number of ATP.[2] In hypoxic mammalian cells, the upregulation of glycolysis is orchestrated by hypoxia-inducible factor (HIF) transcription factors that are stabilized under low oxygen tensions. HIFs transcriptionally activate rate-limiting glycolytic enzymes as well as pyruvate dehydrogenase kinase which deflects pyruvate away from the tricarboxylic acid cycle toward lactic acid (Figure 1).[7–9]
Figure 1. Metabolic Acid Production.

Acid-generating glycolytic flux[2] is elevated in tumors due to both anaerobic and aerobic transcriptional induction of key enzymes by hypoxia inducible factor 1 alpha (HIF1α, HIF1α targets: blue)[91] and oncogenic MYC (MYC targets: red),[92] respectively. (Shared targets of MYC and HIF1α: purple). Further, MYC stimulates mitochondrial biogenesis and function, increasing glutamine oxidation and oxidative phosphorylation,[10] resulting in the production of carbonic acid (H2CO3) from carbon dioxide (CO2) and water. GLUT1 (Glucose transporter 1); HK½ (Hexokinase ½); GPI (Phosphoglucose isomerase); PFK (Phosphofructokinase); ALDA (Fructose-1,6-bisphosphate aldolase); GAPDH (Glyceraldehyde-3-phosphate dehydrogenase); PGK1 (Phosphoglycerate kinase); ENO1 (Enolase); PKM (Pyruvate kinase); PDK (Pyruvate dehydrogenase kinase); LDHA (Lactate dehydrogenase); ATP (Adenosine triphosphate); ADP (Adenosine diphosphate); NAD (Nicotinamide adenine dinucleotide, oxidized); NADH (Nicotinamide adenine dinucleotide, reduced); H+ (proton), HCO3i+ (bicarbonate).
Oncogenes also drive acid-generating glycolytic flux independent of oxygen tensions. Most notably, the commonly activated oncogenic transcription factor MYC also transcriptionally induces glycolysis under non-hypoxic conditions. This contributes to the propensity for cancers to produce lactic acid from glucose even in the presence of oxygen—the so-termed Warburg effect.[10] Activation of other oncogenes, such as AKT, PI3K, BCR-ABL, KRAS, and EGFR, or loss of tumor suppressors, such as TSC2 or LKB1, also has the potential to enhance aerobic glycolysis either post-translationally or through signaling that converges on MYC and HIF (Figure 1).[11, 12] Both anaerobic and aerobic glycolytic flux thus contribute to diminished tumor pH. Hindered perfusion also contributes by slowing the efflux of acid out of the tumor microenvironment for its ultimate elimination by the lungs and kidneys.[5]
Intracellular cytoplasmic pH (pHi) in normal cells is roughly pH 7.2 and protected from further acidification by multiple reinforcing proton-extruding pathways.[13] As HIFs orchestrate the switch from respiration to anaerobic glycolysis in hypoxia, they simultaneously induce monocarboxylate transporters (e.g. MCT4, a symporter for lactate and H+) and carbonic anhydrases (e.g. CA9 and CA12) to permit extracellular venting of the anticipated additional proton load. Other ion channels, carriers, and pumps important to homeostatic pHi maintenance include Na+/H+ exchangers (NHEs), H+-ATPases, and bicarbonate transporters.[5]
Through the use of pH probes and spectroscopy, solid tumors have been documented in vivo to be acidic with pHe as low as pH 5.6 with mean values generally between 6.6 to 7.0.[4, 14] For example, 31P NMR spectroscopy permits measurements of pHi via chemical shifts of energetic phosphorus species (phosphate, phosphocreatine, nucleotide phosphates) in response to protonation. Other approaches, including magnetic resonance imaging of infused hyperpolarized carbon-13 bicarbonate, have revealed tumor pHe as low as 6.5 in experimental tumors.[1] The pHi in human cancers tends to be on average slightly less than pH 7.2 (typically in the range 7.0–7.2), indicating that the redundant pathways for extruding protons successfully allow for maintenance of homeostatic pHi at the expense of acidifying pHe in most cells.[14] The main NHE isoform in most cells, NHE1, becomes more active in response to mitogens.[15, 16] Classic experiments revealed cells expressing mutant NHE isoforms incapable of alkalinizing the cytoplasm in response to mitogens failed to reenter the cell cycle until pHi was elevated with alkaline or bicarbonate-containing media.[15] As such, the prevailing paradigm is that cancers resist pHi acidification to sustain cellular function and pro-growth pathways. This avid extrusion of H+ is thought to generate a more acidic pHe and more alkaline pHi, thereby inverting the normal directionality of the H+ gradient across the plasma membrane.[17] However, a careful study of 31 canine cancers in vivo revealed a wide range of pHi and pHe and significant tumor to tumor variations.[18] Contrary to dogma, pHe was found to be greater than pHi in over 22% of cases, with pHi noted as low as 6.7.
These in vivo data suggest that sudden, sustained, or substantial acid-generating stresses (i.e. hypoxia or oncogene-driven glycolysis) or confrontation by acidic pHe can overwhelm proton extrusion pathways and intracellular buffering capacity. Indeed, such cytoplasmic acidification in response to acidic media exposure is particularly well documented in vitro.[15, 19, 20] Moreover, reported near normal average tumor pHi values do not preclude intratumoral pHi spatial and temporal heterogeneity—potential features which remain relatively underexplored. Additionally, the subcellular topography of proton concentrations within the cytoplasm or across cell or organelle membranes, particularly after stresses such as hypoxia that introduce a proton load, remain unexplored.
3. Sensors of intracellular acidification include both rectifiers and responders
A change in environmental or intracellular pH has myriad effects on cells and tissues, and multiple sensors and signaling cascades have been variably ascribed a contributory role in these effects.[13] Protonation of biological molecules and changes in transmembrane proton gradients are two conceivable means of sensing pH changes. Histidine residues appear to play a dominant role in pH sensing in many different proteins owing to their near neutral pKa.[21] However, in addition to histidine (pKa = 6.5), other functional groups with pKa close to physiological range, such as terminal amino groups (pKa ~ 8), and cysteine (pKa = 8.5), have the potential to “sense” pH through protonation when their pKa values are nudged closer to physiologic pH in the context of protein structure.[22] Perhaps the most illustrative example of such histidine-mediated pH-sensing is the Bohr effect. Under low pH, a salt bridge is formed between a protonated histidine residue and an aspartate residue that allosterically lowers the affinity of hemoglobin for oxygen, enhancing oxygen delivery to hypoxic acidic tissues.
pHI and pHe are believed to be sensed independently. Sensors of acidic pHe, including G-protein coupled receptors (GPCRs), acid-sensitive ion channels (ASICs), and transient receptor potential vanilloid (TRPV) family members, signal to diverse downstream pathways and are reviewed elsewhere.[13, 23–25] Here we discuss pHi sensing.
Because changes in pHi threaten to affect protein folding, macromolecule function or localization, and transmembrane proton gradients (such as those across mitochondrial membranes), several redundant means of H+ efflux act to protect pHi, including bicarbonate exchangers, H+-ATPases, carbonic anhydrases, and NHEs. Although also activated by mitogens as discussed above, the primary stimulus for NHE1 is acidic pHi, which allosterically activates the exchanger through a C-terminal cytoplasmic cluster of histidine resides.[26] In this manner, NHE1 serves as a rectifier of pHi, becoming more active in response to lowering of pHi from the physiologic set point.
Given that glycolysis is the main acid source in tumors, it makes teleological sense that a similar feedback loop might exist to downregulate glycolysis in response to dropping pHi. To such an end, acidification rapidly induces thioredoxin-interacting protein (TXNIP) transcription, largely due to activation of the MondoA/Mlx heterodimeric transcription factor, a member of the extended MYC protein family.[27, 28] TXNIP, in turn, strongly suppresses glycolysis by suppressing transcription of GLUT1 and promoting GLUT1 internalization.[29] Failure of this feedback loop would be predicted to unbridle acid-generating glycolytic flux. Indeed, lactic acidosis was the presenting symptom for a recently characterized family with homozygous TXNIP mutation.[30] Others have described a concomitant increase in glutamine metabolism and associated ammonia production when glycolysis is inhibited by acid.[31] These metabolic responses indicate that cellular efforts to maintain pHi can profoundly influence the metabolic state of cells and contribute to tumor spatial and temporal metabolic heterogeneity. Intra- and intertumoral heterogeneity in pHe can potentially promote this pHi-driven heterogeneity by influencing the proton gradient that H+ venting pathways must confront.[5]
In addition to molecules that serve as rectifiers of pHi, several other molecules sense pHi changes and respond by signaling to a diverse array of downstream outputs to alter cell biology without necessarily acting to restore pH homeostasis. Perhaps the best characterized effects are those on the actin cytoskeleton, which varying degrees of evidence suggest influence cell migration, invasion, and metastasis.[32] For example, the actin filament disassembly activity of cofilins is suppressed through association with phosphatidyl inositol at acidic pH.[13] Likewise, protonation of residues in talin increases its association with focal adhesions, slowing focal adhesion turnover and thereby motility in low pH.[21] Similarly, histidine protonation plays an important role in regulating additional cancer-relevant proteins involved in cytoskeleton regulation and signaling, such as β-catenin and RasGRP1.[33, 34] Metabolites themselves can also be influenced by acidification. Recent work describes how protonation of alpha-ketoglutarate in hypoxia allows it to serve as a substrate for lactate dehydrogenase and malate dehydrogenase to generate the oncometabolite L-2-hydroxyglutarate.[35]
In addition to affecting the cytoskeleton and metabolism, low pHi has been reported to directly affect the physicochemical properties of the cytoplasm of yeast by triggering a fluid- to solid-like state transition.[36] This phase transition is chiefly a result of pHi effects on formation of higher order protein structures, but macromolecular crowding and reduced metabolic activity may make contributions.[36] In fact, metabolic activity in bacteria is thought to trigger local molecular agitation and transition the cytoplasm from a glassy diffusion-limiting substance to a more fluid state.[37] In yeast, energy depletion by metabolic inhibitors can render pHi acidic and induce the solid-like state required for survival (dormancy) under nutrient-deprived conditions.[36] Macromolecular assembly that drives this phase transition appears to be a result of global changes in protein charges. The distribution in charges of cytoplasmic proteins is multimodal but with a significant peak at about −4e (negatively charged) at pH 7.0. At pH 6, average protein charge becomes 0e (neutral), which portends increased protein aggregation due to reduction in repulsive forces.[38] While further work is needed to explore the effect pHi has on the cytoplasmic properties of mammalian cells, these observations are an additional example of how organisms sense environmental conditions through pHi allowing them to respond with appropriate alterations of their metabolism to endure stresses.
4. mTOR is a sensor of intracellular pH through lysosome positioning
In most cultured cells, lysosomes tend to cluster around the perinuclear microtubule organizing center (MTOC).[39] Reversible dispersion of lysosomes to the periphery of mouse macrophages and chick fibroblasts in low pHe was first observed by Heuser in 1989.[40] Although subsequently documented in many other cell types in vitro,[41–43] a functional significance of this phenomenon remained lacking.[44] However, in the course of our recent studies of the circadian clock (Box 1), we unexpectedly found ourselves amidst this biology.[45]
Box 1 – The molecular circadian clock governs homeostatic metabolism.
Circadian clocks allow organisms to anticipate cyclical (daily) events and coordinate optimal timing of physiologic processes.[93] In mammals, clocks in the suprachiasmatic nucleus of the hypothalamus constitute a central pacemaker tuned to the appropriate phase by retina-sensed light. Through neuronal and hormonal (e.g. glucocorticoid) outputs, rhythmic control of behaviors such as eating, and other signaling pathways, the central clock synchronizes the oscillation of autonomous peripheral clocks found in nearly every cell.[94, 95]
The mechanistic basis for both central and peripheral clocks is an interlocking network of feedback loops centered on the heterodimer CLOCK-BMAL1.[96] CLOCK-BMAL1 drives the expression of many genes containing E-box consensus sequences in their regulatory regions. Among these target genes are those encoding period (PER) and cryptochrome (CRY) proteins, which feedback to inhibit CLOCK-BMAL1 posttranslationally, forming the core negative feedback loop. A second negative feedback loop arises from CLOCK-BMAL1–mediated transcription of genes encoding REV-ERB proteins, which bind the RORE motif in the promoter for BMAL1 and negatively regulate its transcription. Together these feedback loops cause circadian oscillation of CLOCK-BMAL1 activity and, consequently, directly or indirectly, the circadian expression of thousands of downstream clock-controlled genes (CCGs) in a given tissue type. Circadian rhythmicity in protein levels, enzymatic activities, metabolites, and cellular processes follow, particularly those with metabolic roles.[97–104]
Our recent work has revealed hypoxia to suppress the circadian clock and circadian transcriptome.[45] This is mediated by a decrease in translation of clock proteins due to mTOR inhibition by intracellular acidification, a consequence of hypoxia-inducible factor (HIF)-driven anaerobic glycolysis. (See text.) Work in flies and mice demonstrating connections between both central and peripheral clocks and mTOR[105–109] indicate this ability of mTOR to modulate the clock in response to sensed environmental stresses may be well-conserved.

Given the central role of the circadian clock in metabolism, we initiated our studies seeking to understand if tumor hypoxia impacted the circadian clock of cancer cells. Indeed, we observed that circadian oscillation of the molecular clock was markedly suppressed in hypoxia. We came to understand that this suppression was mediated by intracellular acidification, a consequence of HIF-mediated upregulation of anerobic glycolysis in hypoxia. Correspondingly, exposure to acidic media was sufficient to decrease pHi and thereby disrupt the clock, while buffering against HIF-mediated acidification or inhibiting HIF-driven glycolytic flux with inhibitors of lactate dehydrogenase (LDH) was sufficient to rescue the clock. Importantly, we found acidification inhibited translation of clock proteins through potent inhibition of mechanistic target of rapamycin complex 1 (mTORC1) signaling, which governs biosynthetic pathways in part through positive regulation of cap-dependent translation (Box 2).[20]. Immunofluorescence and live cell imaging approaches further indicated that mTOR retained proper lysosome localization despite the remarkable redistribution of lysosomes to the periphery of cells in response to pHi acidification (Figure 2). We surmised that this spatial redistribution of lysosomes might be sufficient to inhibit mTOR. Indeed, further experiments using inhibitors or viral activators of motor proteins that traffic lysosomes supported that mTORC1 senses pHi through lysosome positioning.[45]
Box 2 – mTOR senses stress signals to appropriately limit anabolic processes.
mTOR complex 1 (mTORC1), a multi-protein assembly composed in part by the kinase mTOR, regulates cell growth, matching anabolic processes to periods of substrate sufficiency and growth-affirmative humoral signals.[46] Such coordination emerges from multiple parameters indicative of suitability for growth converging on two populations of GTP-binding proteins immediately upstream of mTORC1, Ras-related GTP-binding (RAG) and Ras homolog enriched in brain (RHEB) proteins. Sensors for arginine, leucine, and methionine signal amino acid sufficiency to lysosome-localized RAG heterodimers through a series of protein interactions that ultimately control RAG nucleotide binding states.[110–114] Correct GTP/GDP loading of RAG proteins recruits mTORC1 to the lysosomal surface. In the widely accepted model, RHEB is anchored to the lysosomal surface thus allowing activation of amino-acid-recruited mTOR by RHEB.[53] RHEB is GTP-bound and thus capable of activating recruited mTOR so long as the tuberous sclerosis complex (TSC), which contains TSC2, the GTPase-activating protein (GAP) for RHEB, is spatially disengaged from RHEB in response to growth factors (e.g. signals relayed through phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling)[55] and kept inactive through absence of TSC2-activating stress signals (e.g. energy insufficiency sensed by adenosine monophosphate (AMP)-sensitive AMP kinase (AMPK)).[115]

Active mTORC1 positively regulates translation through mTOR phosphorylation of downstream substrates.[116, 117] Phosphorylation of ribosomal S6 kinase (S6K) stimulates S6K phosphorylation of ribosomal protein S6, a component of the 40S ribosomal subunit, driving cell growth through debated mechanisms.[118] mTOR also phosphorylates inhibitory binding partners of the eukaryotic initiation factor 4E (eIF4E), the 4EBP proteins, preventing their engagement with eIF4E and thereby releasing suppression of cap-dependent translation. mTORC1 thus senses stress and reserves energy-costly translation and downstream biosynthetic processes for periods in which reserves of energy and building blocks can support these demands. In addition to driving biosynthesis through activation of cap-dependent translation, mTOR also positively regulates lipid and nucleotide biosynthesis to support growth, as well as metabolic pathways such as glycolysis and production of reducing equivalents that support these anabolic processes[115].

Figure 2. Acid-mediated dispersion of lysosomes inhibits mTORC1 signaling.

Amino acids through RAG proteins recruit mTOR to the surface of lysosomes allowing mTOR to contact RHEB localized to a non-lysosomal endomembrane. RHEB’s activation of mTOR is licensed by growth factors and replete energy stores. When intracellular pH falls, lysosomes disperse to the periphery of cells which inhibits mTOR through separation from RHEB. Lysosome dispersion likely results from an imbalance between dynein-mediated centripetal forces and kinesin-mediated centrifugal forces in acid. Inhibition of mTOR decreases phosphorylation (p-) of downstream S6 and 4EBP1 proteins, which suppresses synthesis of circadian clock proteins through inhibition of cap-dependent translation. (See also Box 2.)
mTORC1 integrates environmental signals including levels of amino acids and growth factors to limit translation and other biosynthetic activities to periods of replete resources.[46] It therefore seems appropriate that mTOR activity is similarly gated by pHi to restrict protein synthesis to times when pH is conducive to proper protein folding. Additionally, as acid stress is often coupled to hypoxic stress, mTOR sensitivity to pHi may have evolved to curb energy costly protein synthesis during periods of energy stress. Furthermore, as mTORC1 positively regulates glycolysis, in part through driving HIF translation,[47, 48] the ability of mTORC1 to sense glycolysis-generated acid may permit a feedback loop to tune glycolytic flux to maintain homeostatic pHi.
Peripheral redistribution of lysosomes in acidic pHi may additionally help rectify pHi through fusion of lysosomes with the plasma membrane. As cytoplasmic protons are pumped into lysosome lumens through the action of the lysosomal H+ pump V-ATPase, “lysosome exocytosis” could permit dumping of protons into the extracellular space. Additionally, this would place V-ATPase on the plasma membrane where it could continue to extrude cytoplasmic protons. Indeed, cells chronically adapted to acidic (pH 6.7) media display strong plasma membrane staining for the lysosomal transmembrane protein LAMP2 consistent with lysosome exocytosis.[17]
Trafficking of lysosomes on microtubules in cells involves opposing activities of two classes of ATP-dependent motor proteins. Dynein traffics lysosomes toward the nucleus of cells, while members of the large family of kinesins largely participate in centrifugal movement toward the plus ends of microtubules (Figure 2).[44] The mechanism by which acid drives lysosomes to the periphery remains unsettled but is generally speculated to be an upsetting of the normal balance between centripetal and centrifugal forces. Indeed, small molecule inhibition of dynein dispersed lysosomes and diminished mTORC1 activity in neutral pH conditions.[45] Correspondingly, knockdown of KIF5B, an abundant cellular kinesin and component of kinesin-1, reduced centrifugal lysosome movement in acid.[42] and partially increased mTORC1 activity under low pHe.[45] Intriguingly, kinesin has been shown in vitro to be inactive in neutral pH through inhibitory interaction of its light chain (LC) with its heavy chain (HC) in a manner that blocks HC microtubule binding. Lowering the pH in vitro below 7.2 protonates a negatively charged domain within the LC and permits microtubule binding.[49, 50] Alternatively, axonemal (flagellar) dynein is appreciated to be inhibited by intracellular acidification, which is believed to keep spermatozoa flagella inactive during storage in the epididymis,[51] and pH-dependence of cytoplasmic dynein has been likewise proposed and attributed to histidines at dynein LC’s dimer interface.[52] These observations suggest that a rapid response to low pH through kinesin activation or dynein inhibition may disperse lysosomes to cellular peripheries and thereby rapidly inhibit mTORC1 signaling. However, further work is necessary to elucidate the exact mechanism underlying this three-decade-old phenomenon.[40]
5. Where is RHEB?
In acidic conditions, lysosomes dispersed peripherally and such a spatial change appeared necessary and sufficient for mTORC1inhibition by acid.[45] However, why this change in localization of lysosome-bound mTOR should be inhibitory to mTORC1 signaling was not clear initially. In the widely accepted model of mTORC1 signaling, mTORC1 is activated when amino acid sufficiency is sensed via RAG proteins resulting in recruitment of mTORC1 to the surface of lysosomes where it can be activated by lysosome-localized RHEB.[53] In acidic conditions, mTOR retained its proper lysosome localization yet mTOR signaling was strongly inhibited. Even deleting TSC2 (the upstream inhibitory GTPase activating protein (GAP) for RHEB) to ensure active RHEB (Box 2) failed to rescue mTOR signaling in acid.[45] Given this perplexing ability of acid to strongly inhibit mTORC1 signaling without hindering mTOR lysosomal localization or activating TSC2, we surmised that perhaps RHEB’s contact with mTOR was hindered. In surprising agreement with this prediction, RHEB immunostaining revealed that RHEB maintained a perinuclear distribution in acid despite centrifugal dispersion of lysosomes. This rapid separation of lysosome-bound mTORC1 from its activator RHEB appeared to underpin inhibition of mTORC1, as strongly overexpressing RHEB to intentionally cause its mislocalization throughout the cytoplasm rescued mTORC1 signaling and the circadian clock.[45]
Retention of RHEB in a perinuclear position in acid suggested RHEB might be localized to a non-lysosomal endomembrane compartment. Indeed, high resolution images of immunofluorescence co-staining for endogenous RHEB, lysosomal protein LAMP1, and mTOR at neutral conditions revealed high coincidence in spatial distribution of LAMP1 and mTOR but distinct patterning of RHEB suggestive of RHEB’s occupancy of a spatially adjacent but separate compartment.[45] This led us to propose that RHEB normally contacts and activates lysosome-localized mTOR from a distinct endomembrane compartment (Figure 2) (a relationship which we termed “transendomembrane” to contrast to the prevailing “cis-” interaction in which both RHEB and mTOR are proposed to both be lysosomal). In other words, amino acids recruit mTOR to lysosomes enriched at the MTOC allowing mTOR to contact RHEB localized to other endomembranes likewise scaffolded by the MTOC.
Our model (Figure 2) differs from the prevailing model of mTORC1 signaling in which RHEB is asserted to be constitutively anchored to lysosomal membranes through its farnesylated tail.[53–55] The ability of farnesylation inhibitors that dislodge RHEB from membranes to inhibit mTOR signaling[56, 57] suggests active RHEB resides on endomembranes. But which membrane? The perinuclear location of RHEB suggests several candidate membranes including Golgi, endoplasmic reticulum (ER), and mitochondria.
Notably, despite the dominant model localizing RHEB to lysosomes,[53] several groups have reported non-lysosomal perinuclear RHEB localization in agreement with our inferences. Using GFP-tagged RHEB and RHEB2 (RHEBL1) constructs, Hanker and colleagues concluded RHEB colocalized with Golgi and ER markers,[56] Moreover, these authors failed to observe colocalization of either RHEB construct with markers of late endosomes or lysosomes, including LAMP2 and LysoTracker. An earlier report by Buerger and colleagues likewise concluded exogenous RHEB constructs localized to the ER and Golgi.[57] Moreover, noted abolition of mTORC1 signaling after treatment with brefeldin A, an ER to Golgi trafficking inhibitor disruptive of Golgi integrity, supported the proposition that RHEB needed to reach Golgi membranes to activate mTORC1. Correspondingly, substitution of RHEB’s endogenous membrane anchoring tail with one targeted to the Golgi, but not the ER, was sufficient to fully activate mTORC1. Notably, these early authors interpreted Golgi localization of RHEB as indicative of mTOR recruitment to the Golgi as lysosome-localization of mTOR had not yet been described.[53] Shortly after publication of our work, we became aware that an independent group had concurrently converged on a similar model of “inter-organelle communication” between Golgi-localized RHEB and lysosome-localized mTOR after recapitulating the findings of Buerger et al.[58] However, all three of these studies relied on exogenously expressed RHEB constructs which run the risk of mislocalization. Indeed, loss of mTOR’s amino acid dependence in experiments by Hao and colleagues seem to betray this confounding issue.
Fortunately, Manifava and colleagues not only independently corroborated this ER/Golgi distribution of exogenous RHEB constructs but also characterized the location of endogenous RHEB, noting very weak lysosomal localization but substantial colocalization with golgi protein giantin.[59] Recent unpublished work by Angarola and Gerguson likewise notes lack of lysosomal enrichment of endogenous RHEB both when assessed with an endogenous antibody or with an anti-HA antibody after CRISPR-mediated introduction of an tag into the endogenous locus.[60] When they turned to exogenous constructs, they, like Buerger and Hao, observed ER localization but perplexing absence of mTOR signaling when RHEB was constitutively ER targeted. Thus, where the active pool of RHEB resides and how it makes activating contact with lysosome-localized mTOR remains an unanswered question. Although RHEB localization to ribosome-studded ER would be a logical place for mTOR contact to be made given mTOR’s regulation of translation, the above evidence seems to argue against statically ER-localized RHEB being the active pool. One possibility given the typical ER to Golgi flow of membrane is that Golgi-localized RHEB is the truly mTOR-activating pool whereas ER-localized RHEB represents more immature RHEB protein en route to the Golgi (Figure 2). Alternatively, the ER may be the site of RHEB-mTOR contact but static targeting of RHEB to the ER may be problematic perhaps due to a need for exchange with the prominent cytosolic pool of RHEB to somehow maintain RHEB in an activated state.[61]
6. Conflicting reports argue peripheral mTOR is active
Our finding that centrifugal lysosome redistribution is both necessary and sufficient for acid’s suppression of mTOR is consistent with prior work indicating dynein[62] and perinuclear clustering of lysosomes[63, 64] support mTORC1 signaling. However, it is notable that Korolchuk and colleagues reported a seemingly inverted spatial dynamic from our findings with starvation (and therefore inhibited mTOR) associated with perinuclear clustering of lysosomes and refeeding (and therefore active mTOR) associated with peripheral redistribution.[41] As starvation-induced mTOR inhibition induces autophagy, this perinuclear clustering of lysosomes may reflect the movement of lysosomes and autophagosomes to the MTOC to facilitate fusion. Indeed, pharmacologic inhibition of mTORC1 and starvation appear to induce tight perinuclear lysosome aggregation.[55, 65] As centrifugal lysosome redistribution is a cause rather than consequence of mTORC1 inhibition in our model, acid-induced silencing of mTORC1 through peripheral redistribution of lysosomes is not incompatible with autophagy-induced retrograde trafficking of lysosomes.
However, further data presented by Korolchuk and colleagues are more difficult to reconcile with our findings. These authors reported that peripheral margination of lysosomes in Hela cells through overexpression of several kinesins and associated adaptors caused augmented mTOR signaling with the reverse true upon knockdown.[41] However, a more recent report in Hela cells did not observe hindered mTORC1 signaling when lysosomes clustered at the MTOC in response to knockout of a different kinesin-associated factor.[66] The reasons for these contradictory results in the same cell line remains unclear. Nonetheless, we cannot exclude the existence of cell-type specific differences as Hela cells appear distinctly wired in several ways. For instance, Korolchuk and colleagues noted that starvation (including serum starvation) alkalinized Hela cell pHi, which they suspected mediated lysosome perinuclear aggregation. Given that it is classically noted that growth factor stimulation causes alkalinization of pHi, owing to the growth-factor responsiveness of H+ exchangers such as NHE1,[15, 16] this is surprising. It is also possible that human papillomavirus 18 (HPV 18) infection of Hela cells endows a distinct biology. Indeed, cytomegalovirus (CMV), another DNA virus, alters trafficking of mTOR to achieve amino acid independent activation that supports biosynthesis of viral progeny.[62, 64] Intriguingly, HPV E6 has been reported to activate mTORC1 and also interact with a component of endosomal cycling.[67, 68] Moreover, knockdown of E6 was noted to result in tighter perinuclear endosome clustering,[67] suggesting E6 may perhaps underlie peripheral spread of mTOR-bound endomembranes in Hela cells.
Recent work studying a panel of cell lines, including Hela cells, has demonstrated that cells display two pools of lysosomes, a larger perinuclear pool and a smaller more dynamic peripheral pool.[39] mTOR localization appears to mirror this distribution in the presence of amino acids.[45, 54, 55] We suspect future investigations will clarify that perinuclear lysosome-bound mTORC1 is an active pool and that this location is critical for spatial contact with RHEB. That CMV has evolved a mechanism to actively bring mTOR to the perinuclear region and that doing so allows the virus to maintain mTORC1 activity in the face of inhibitory signals underscore, we believe, the importance of this spatial location to mTORC1 activation.[62, 64] However, in our studies at neutral pH, the periphery of the cell was not devoid of lysosome-bound mTOR and our model need not imply that subpopulations of mTORC1 do not translocate to various niches in the cell to regionally tailor biosynthetic activities. Indeed, in other studies exploring the spatial localization of mTOR in Hela cells—as well as in MEFs, A549, and PANC-1 cells—the majority of mTORC1 localized perinuclearly in serum-stimulated (mTORC1-activated) cells with a much smaller portion localized to plasma membranes.[69] Intriguingly, Hong and colleagues have recently proposed a model in which an ER localized protein, protrudin, makes contact with lysosomes in response to their reception of mTORC1-activating and phosphatidylinositol-3-phosphate (PI(3)P)-generating stimuli, activates kinesin-1 in the process, and thus sends lysosomes peripherally.[70] Such a model might reconcile a portion of discrepant findings, underscoring the critical importance of multi-organelle contact at the MTOC to activation of mTORC1 without disallowing for peripheral translocation of activated mTORC1.
7. Leukocyte mTOR inhibition by tumor acidity may suppress anticancer immunity
Tumor acidity affects not only cancer cell intrinsic biology but also the immune system.[71] Lymphocytes have long been recognized to alkalinize pHi upon stimulation.[72] More recently, mTORC1 has been revealed to play a critical role in the differentiation and activation of CD4+ and CD8+ T cells.[73–75] For instance, pharmacologic or genetic inhibition of mTORC1 signaling prevents differentiation of CD8+ T cells into effector cells[74, 76] and CD4+ T cells into Th1 and Th17 subtypes.[77] Several reports have noted without mechanistic explanation that acidic media reversibly suppresses T cell cytokine production and cytolytic abilities in vitro.[71, 78–81] Correspondingly, the potent suppression of mTORC1 signaling by acid observed by us in CD8+ and CD4+ cells might be the mechanism behind acid’s suppressive effects on T cells.[45] Consistent with this, suppression of IFNγ production in acid was noted to be reversible and posttranscriptional in a manner suggestive of a translational block.[78, 81] Indeed, rapamycin has been used for decades for its immunosuppressive function.
Peripheral redistribution of lysosomes observed in acidic cancer cells suggests microtubule-based vesicle trafficking events critical to T cell function might also be disturbed in the acidic tumor microenvironment.[45] After engaging an antigen presenting cell, the MTOC of the naïve T cell moves beneath the immune synapse (point of T cell receptor (TCR) engagement) and additional TCR receptors are delivered in a dynein-dependent manner. Lytic granules are similarly delivered to the MTOC when a cytolytic T cell encounters a target cell.[82] Moreover, asymmetric partitioning (i.e. microtubule-directed trafficking) of fate-determining proteins amongst daughter cells during antigen-stimulated cell divisions gives rise to memory and effector lineages of CD8+ T cells.[74] Therefore, it is conceivable that acid-mediated antegrade redistribution of vesicles could hinder activation, differentiation, and effector abilities of T cells in both mTOR-dependent and independent ways. Consistent with this, in previous studies of T cell cultured in acid, perforin content was lower[81] and degranulation was also reversibly suppressed.[79] Acid neutralizing tactics might thus be helpful adjuvants to traditional cancer therapeutics and newer anticancer immunotherapies through restoration of mTORC1 signaling and proper vesicle trafficking in immune cells.
8. Restoring mTORC1 signaling through pH neutralization may have therapeutic potential
Administration of HCO3−-supplemented drinking water to tumor-bearing mice has been shown by hyperpolarized H13CO3− magnetic resonance imaging,[1] intravital pH indicator dye,[83] and microelectrode measurements[84] to increase tumor pHe in vivo.[1] Two therapeutic strategies can be envisioned. One, restoring tumor cell pHi and thereby mTOR signaling could drive cells into a state susceptible to metabolic inhibitors, anti-proliferative agents, or other therapies. While restoring mTORC1 signaling in quiescent cancer cells potentially restores their proliferative capacity, the idea is to create an opportunity to then target this awakened metabolic state. Illustrative of this strategy, we and others demonstrated that HCO3−-supplemented drinking water increased mTORC1 signaling within multiple xenograft and allograft tumors.[45, 85] Moreover, Faes and colleagues further demonstrated that this strategy restored rapamycin sensitivity.[85] Two, neutralizing pHe could enhance antitumor immune function. Indeed, HCO3− administration or treatment with a proton pump inhibitor elevated the pHe of B16 melanoma allografts and enhanced response to immune checkpoint inhibitor therapy and adoptive lymphocyte transfer, respectively.[78, 79] These studies suggest that therapeutic strategies to normalize tumor pHe could have clinical promise.[25]
Large unresectable hepatocellular carcinomas (HCCs) are treated with chemoembolization, a tactic intended to deliver concentrated chemotherapy and starve cancer cells through embolization but that is of only questionable benefit.[86] Intriguingly, however, a clinical study of unresectable HCC in 57 non-randomized Chinese patients revealed that the addition of bicarbonate to chemoembolization markedly improved objective response rate (ORR) (100% vs 44%) and reduced residual disease volume (RDV) (7% vs. 46%).[87] Importantly, this was subsequently born out in a smaller randomized controlled trial of 20 patients (ORR 100% vs. 64%, RD 4% vs 28%). While the mechanism for this effect can only be speculated at current, the authors favor the hypothesis based on their prior in vitro data that tumor lactic acidosis exacerbated by embolization pushes cancer cells into a quiescent state of low metabolic demand protective against embolization-induced nutrient deprivation.[88–90] Therefore, it’s intriguing to speculate that bicarbonate-induced reactivation of mTOR may sustain untenable metabolic demand that mediate these remarkable findings. Furthermore, as available survival data also point to sustained benefit (41- vs 14-month nonrandomized mean survival), it’s likewise alluring to speculate that bicarbonate-mediated enhanced immune cell mTOR signaling also contributes. Given these remarkable outcomes, we eagerly await future study revealing the mechanism of this bicarbonate-driven effect and indications for extended use.
9. Conclusions and Outlook
Setting out to assess the state of the molecular circadian clock in hypoxia, we have come away learning principles of fundamental pH sensing and mTOR activation that have ignited our inquiry into diverse biology from molecular motors and endomembrane trafficking to antitumor immunity. We have learned mTOR is a sensor of pHi through lysosome positioning relative to RHEB. As such, mTOR appears to complement other pH sensing mechanisms while also offering an ability to appropriately broaden the metabolic response. But how acid-driven lysosome dispersion occurs at a molecular level remains unanswered. Separation of mTOR and its activator RHEB in acid have revealed RHEB to occupy a non-lysosomal membrane, the identity of which remains under active debate. We surmise that mTOR-coordinated downregulation of the circadian clock and biosynthetic processes represents a powerful stress response that cancers may rely on to confront the exceptional acidity of solid tumors. Solid tumor acidity may thus contribute to tumor dormancy and blunted antitumor immune responses. As such, additional studies are warranted to determine whether relatively straightforward pH correction could enhance a broad range of cancer therapeutic approaches.
Acknowledgements.
This work is partially supported by NCI grants R01CA051497 (CVD), R01CA57341 (CVD), F30CA200347 (ZEW), T32CA9140–39 (ZEW), P30CA010815 (D. Altieri); the Patel Scholar Award (ZEW); and the Ludwig Institute for Cancer Research.
Abbreviations:
- HIF
hypoxia-inducible factor
- pHe
extracellular pH
- pHi
intracellular cytoplasmic pH
- mTORC1
mechanistic target of rapamycin complex 1
- RHEB
Ras homolog enriched in brain
- RAG
Ras-related GTP-binding
- NHE
Na+/H+ exchanger
- TXNIP
thioredoxin-interacting protein
- MTOC
microtubule organizing center
Footnotes
Conflict-of-Interest disclosure: We have no financial relationship that creates a conflict with the content of this work. CVD has stocks in Agios, is a consultant for Cellworks, Inc; Rafael Pharm, Inc.; Dracen Pharm, Inc; Polaris Pharm, Inc., and member of the board of directors of Rafael Pharmaceuticals.
References
- [1].Gallagher FA, Kettunen MI, Day SE, Hu DE, Ardenkjaer-Larsen JH, Zandt R, Jensen PR, Karlsson M, Golman K, Lerche MH, Brindle KM, Nature 2008, 453, 940. [DOI] [PubMed] [Google Scholar]
- [2].Divakaruni AS, Paradyse A, Ferrick DA, Murphy AN, Jastroch M, Methods Enzymol 2014, 547, 309. [DOI] [PubMed] [Google Scholar]
- [3].Carmeliet P, Jain RK, Nature 2011, 473, 298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Vaupel P, Kallinowski F, Okunieff P, Cancer Res 1989, 49, 6449. [PubMed] [Google Scholar]
- [5].Swietach P, Vaughan-Jones RD, Harris AL, Hulikova A, Philos Trans R Soc Lond B Biol Sci 2014, 369, 20130099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Pasteur L, Robb DC, Faulkner F, Studies on fermentation The diseases of beer, their causes, and the means of preventing them, Macmillan & co., London: 1879. [Google Scholar]
- [7].Semenza GL, J Clin Invest 2013, 123, 3664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Kim JW, Tchernyshyov I, Semenza GL, Dang CV, Cell Metab 2006, 3, 177. [DOI] [PubMed] [Google Scholar]
- [9].Xie H, Simon MC, J Biol Chem 2017, 292, 16825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Stine ZE, Walton ZE, Altman BJ, Hsieh AL, Dang CV, Cancer Discov 2015, 5, 1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].DeBerardinis RJ, Chandel NS, Science advances 2016, 2, e1600200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Vander Heiden MG, DeBerardinis RJ, Cell 2017, 168, 657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Damaghi M, Wojtkowiak JW, Gillies RJ, Front Physiol 2013, 4, 370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Gillies RJ, Raghunand N, Karczmar GS, Bhujwalla ZM, J Magn Reson Imaging 2002, 16, 430. [DOI] [PubMed] [Google Scholar]
- [15].Pouyssegur J, Franchi A, L’Allemain G, Paris S, FEBS Lett 1985, 190, 115. [DOI] [PubMed] [Google Scholar]
- [16].Moolenaar WH, Annu Rev Physiol 1986, 48, 363. [DOI] [PubMed] [Google Scholar]
- [17].Damaghi M, Tafreshi NK, Lloyd MC, Sprung R, Estrella V, Wojtkowiak JW, Morse DL, Koomen JM, Bui MM, Gatenby RA, Gillies RJ, Nat Commun 2015, 6, 8752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Prescott DM, Charles HC, Poulson JM, Page RL, Thrall DE, Vujaskovic Z, Dewhirst MW, Clin Cancer Res 2000, 6, 2501. [PubMed] [Google Scholar]
- [19].McBrian MA, Behbahan IS, Ferrari R, Su T, Huang TW, Li K, Hong CS, Christofk HR, Vogelauer M, Seligson DB, Kurdistani SK, Mol Cell 2013, 49, 310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Balgi AD, Diering GH, Donohue E, Lam KK, Fonseca BD, Zimmerman C, Numata M, Roberge M, PLoS One 2011, 6, e21549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Webb BA, Chimenti M, Jacobson MP, Barber DL, Nat Rev Cancer 2011, 11, 671. [DOI] [PubMed] [Google Scholar]
- [22].Platzer G, Okon M, McIntosh LP, J Biomol NMR 2014, 60, 109. [DOI] [PubMed] [Google Scholar]
- [23].Justus CR, Dong L, Yang LV, Front Physiol 2013, 4, 354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Ludwig MG, Vanek M, Guerini D, Gasser JA, Jones CE, Junker U, Hofstetter H, Wolf RM, Seuwen K, Nature 2003, 425, 93. [DOI] [PubMed] [Google Scholar]
- [25].Pillai SR, Damaghi M, Marunaka Y, Spugnini EP, Fais S, Gillies RJ, Cancer Metastasis Rev 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Webb BA, White KA, Grillo-Hill BK, Schonichen A, Choi C, Barber DL, J Biol Chem 2016, 291, 24096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Chen JL, Merl D, Peterson CW, Wu J, Liu PY, Yin H, Muoio DM, Ayer DE, West M, Chi JT, PLoS Genet 2010, 6, e1001093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Wilde BR, Ye Z, Lim TY, Ayer DE, Elife 2019, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Wu N, Zheng B, Shaywitz A, Dagon Y, Tower C, Bellinger G, Shen CH, Wen J, Asara J, McGraw TE, Kahn BB, Cantley LC, Mol Cell 2013, 49, 1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Katsu-Jimenez Y, Vazquez-Calvo C, Maffezzini C, Halldin M, Peng X, Freyer C, Wredenberg A, Gimenez-Cassina A, Wedell A, Arner ESJ, Diabetes 2019, 68, 709. [DOI] [PubMed] [Google Scholar]
- [31].Xie L, Pilbrough W, Metallo C, Zhong T, Pikus L, Leung J, Aunins JG, Zhou W, Biotechnology and bioengineering 2002, 80, 569. [DOI] [PubMed] [Google Scholar]
- [32].White KA, Grillo-Hill BK, Barber DL, J Cell Sci 2017, 130, 663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].White KA, Grillo-Hill BK, Esquivel M, Peralta J, Bui VN, Chire I, Barber DL, J Cell Biol 2018, 217, 3965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Vercoulen Y, Kondo Y, Iwig JS, Janssen AB, White KA, Amini M, Barber DL, Kuriyan J, Roose JP, Elife 2017, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Intlekofer AM, Dematteo RG, Venneti S, Finley LW, Lu C, Judkins AR, Rustenburg AS, Grinaway PB, Chodera JD, Cross JR, Thompson CB, Cell metabolism 2015, 22, 304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Munder MC, Midtvedt D, Franzmann T, Nuske E, Otto O, Herbig M, Ulbricht E, Muller P, Taubenberger A, Maharana S, Malinovska L, Richter D, Guck J, Zaburdaev V, Alberti S, Elife 2016, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Parry BR, Surovtsev IV, Cabeen MT, O’Hern CS, Dufresne ER, Jacobs-Wagner C, Cell 2014, 156, 183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Weiller GF, Caraux G, Sylvester N, Proteomics 2004, 4, 943. [DOI] [PubMed] [Google Scholar]
- [39].Jongsma ML, Berlin I, Wijdeven RH, Janssen L, Janssen GM, Garstka MA, Janssen H, Mensink M, van Veelen PA, Spaapen RM, Neefjes J, Cell 2016, 166, 152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Heuser J, J Cell Biol 1989, 108, 855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Korolchuk VI, Saiki S, Lichtenberg M, Siddiqi FH, Roberts EA, Imarisio S, Jahreiss L, Sarkar S, Futter M, Menzies FM, O’Kane CJ, Deretic V, Rubinsztein DC, Nat Cell Biol 2011, 13, 453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Dykes SS, Gray AL, Coleman DT, Saxena M, Stephens CA, Carroll JL, Pruitt K, Cardelli JA, Oncotarget 2016, 7, 31037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Glunde K, Guggino SE, Solaiyappan M, Pathak AP, Ichikawa Y, Bhujwalla ZM, Neoplasia 2003, 5, 533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Pu J, Guardia CM, Keren-Kaplan T, Bonifacino JS, J Cell Sci 2016, 129, 4329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Walton ZE, Patel CH, Brooks RC, Yu Y, Ibrahim-Hashim A, Riddle M, Porcu A, Jiang T, Ecker BL, Tameire F, Koumenis C, Weeraratna AT, Welsh DK, Gillies R, Alwine JC, Zhang L, Powell JD, Dang CV, Cell 2018, 174, 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Saxton RA, Sabatini DM, Cell 2017, 168, 960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, Vander Heiden MG, MacKeigan JP, Finan PM, Clish CB, Murphy LO, Manning BD, Mol Cell 2010, 39, 171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Brugarolas JB, Vazquez F, Reddy A, Sellers WR, Kaelin WG Jr., Cancer Cell 2003, 4, 147. [DOI] [PubMed] [Google Scholar]
- [49].Verhey KJ, Lizotte DL, Abramson T, Barenboim L, Schnapp BJ, Rapoport TA, J Cell Biol 1998, 143, 1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Wong YL, Rice SE, Proc Natl Acad Sci U S A 2010, 107, 11781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Nishigaki T, Jose O, Gonzalez-Cota AL, Romero F, Trevino CL, Darszon A, Biochem Biophys Res Commun 2014, 450, 1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Mohan PM, Barve M, Chatterjee A, Hosur RV, Protein Sci 2006, 15, 335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, Sabatini DM, Cell 2010, 141, 290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled L, Sabatini DM, Science 2008, 320, 1496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Menon S, Dibble CC, Talbott G, Hoxhaj G, Valvezan AJ, Takahashi H, Cantley LC, Manning BD, Cell 2014, 156, 771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Hanker AB, Mitin N, Wilder RS, Henske EP, Tamanoi F, Cox AD, Der CJ, Oncogene 2010, 29, 380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Buerger C, DeVries B, Stambolic V, Biochem Biophys Res Commun 2006, 344, 869. [DOI] [PubMed] [Google Scholar]
- [58].Hao F, Kondo K, Itoh T, Ikari S, Nada S, Okada M, Noda T, J Cell Sci 2018, 131. [DOI] [PubMed] [Google Scholar]
- [59].Manifava M, Smith M, Rotondo S, Walker S, Niewczas I, Zoncu R, Clark J, Ktistakis NT, Elife 2016, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Ferguson SM, Angarola B, bioRxiv 2019, 513473. [Google Scholar]
- [61].Kovacevic M, Klein CH, Rossmannek L, Konitsiotis AD, Stanoev A, Kraemer AU, Bastiaens P, bioRxiv 2018, 472241. [Google Scholar]
- [62].Clippinger AJ, Alwine JC, Genes Dev 2012, 26, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Rainero E, Howe Jonathan D., Caswell Patrick T., Jamieson Nigel B., Anderson K, Critchley David R., Machesky L, Norman Jim C., Cell Reports 2015, 10, 398. [DOI] [PubMed] [Google Scholar]
- [64].Clippinger AJ, Maguire TG, Alwine JC, J Virol 2011, 85, 9369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Li X, Rydzewski N, Hider A, Zhang X, Yang J, Wang W, Gao Q, Cheng X, Xu H, Nat Cell Biol 2016, 18, 404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Pu J, Schindler C, Jia R, Jarnik M, Backlund P, Bonifacino JS, Dev Cell 2015, 33, 176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Ganti K, Massimi P, Manzo-Merino J, Tomaic V, Pim D, Playford MP, Lizano M, Roberts S, Kranjec C, Doorbar J, Banks L, PLoS Pathog 2016, 12, e1005854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Spangle JM, Munger K, J Virol 2010, 84, 9398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Saci A, Cantley LC, Carpenter CL, Mol Cell 2011, 42, 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Hong Z, Pedersen NM, Wang L, Torgersen ML, Stenmark H, Raiborg C, J Cell Biol 2017, 216, 4217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Huber V, Camisaschi C, Berzi A, Ferro S, Lugini L, Triulzi T, Tuccitto A, Tagliabue E, Castelli C, Rivoltini L, Semin Cancer Biol 2017, 43, 74. [DOI] [PubMed] [Google Scholar]
- [72].Gerson DF, Kiefer H, Eufe W, Science 1982, 216, 1009. [DOI] [PubMed] [Google Scholar]
- [73].Powell JD, Pollizzi KN, Heikamp EB, Horton MR, Annu Rev Immunol 2012, 30, 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Pollizzi KN, Patel CH, Sun IH, Oh MH, Waickman AT, Wen J, Delgoffe GM, Powell JD, J Clin Invest 2015, 125, 2090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Pollizzi KN, Sun IH, Patel CH, Lo YC, Oh MH, Waickman AT, Tam AJ, Blosser RL, Wen J, Delgoffe GM, Powell JD, Nat Immunol 2016, 17, 704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Rao RR, Li Q, Odunsi K, Shrikant PA, Immunity 2010, 32, 67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, Xiao B, Worley PF, Powell JD, Nat Immunol 2011, 12, 295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Pilon-Thomas S, Kodumudi KN, El-Kenawi AE, Russell S, Weber AM, Luddy K, Damaghi M, Wojtkowiak JW, Mule JJ, Ibrahim-Hashim A, Gillies RJ, Cancer Res 2016, 76, 1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Calcinotto A, Filipazzi P, Grioni M, Iero M, De Milito A, Ricupito A, Cova A, Canese R, Jachetti E, Rossetti M, Huber V, Parmiani G, Generoso L, Santinami M, Borghi M, Fais S, Bellone M, Rivoltini L, Cancer Res 2012, 72, 2746. [DOI] [PubMed] [Google Scholar]
- [80].Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, Matos C, Bruss C, Klobuch S, Peter K, Kastenberger M, Bogdan C, Schleicher U, Mackensen A, Ullrich E, Fichtner-Feigl S, Kesselring R, Mack M, Ritter U, Schmid M, Blank C, Dettmer K, Oefner PJ, Hoffmann P, Walenta S, Geissler EK, Pouyssegur J, Villunger A, Steven A, Seliger B, Schreml S, Haferkamp S, Kohl E, Karrer S, Berneburg M, Herr W, Mueller-Klieser W, Renner K, Kreutz M, Cell Metab 2016, 24, 657. [DOI] [PubMed] [Google Scholar]
- [81].Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, Gottfried E, Schwarz S, Rothe G, Hoves S, Renner K, Timischl B, Mackensen A, Kunz-Schughart L, Andreesen R, Krause SW, Kreutz M, Blood 2007, 109, 3812. [DOI] [PubMed] [Google Scholar]
- [82].de la Roche M, Asano Y, Griffiths GM, Nat Rev Immunol 2016, 16, 421. [DOI] [PubMed] [Google Scholar]
- [83].Estrella V, Chen T, Lloyd M, Wojtkowiak J, Cornnell HH, Ibrahim-Hashim A, Bailey K, Balagurunathan Y, Rothberg JM, Sloane BF, Johnson J, Gatenby RA, Gillies RJ, Cancer Res 2013, 73, 1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Ibrahim-Hashim A, Robertson-Tessi M, Enriquez-Navas PM, Damaghi M, Balagurunathan Y, Wojtkowiak JW, Russell S, Yoonseok K, Lloyd MC, Bui MM, Brown JS, Anderson ARA, Gillies RJ, Gatenby RA, Cancer Res 2017, 77, 2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Faes S, Duval AP, Planche A, Uldry E, Santoro T, Pythoud C, Stehle JC, Horlbeck J, Letovanec I, Riggi N, Demartines N, Dormond O, Mol Cancer 2016, 15, 78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Oliveri RS, Wetterslev J, Gluud C, Cochrane Database of Systematic Reviews 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Chao M, Wu H, Jin K, Li B, Wu JJ, Zhang GQ, Yang G, Hu X, eLife 2016, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Hu X, Chao M, Wu H, Signal Transduct Target Ther 2017, 2, 16047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Wu H, Ding Z, Hu D, Sun F, Dai C, Xie J, Hu X, J Pathol 2012, 227, 189. [DOI] [PubMed] [Google Scholar]
- [90].Xie J, Wu H, Dai C, Pan Q, Ding Z, Hu D, Ji B, Luo Y, Hu X, Sci Rep 2014, 4, 4927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Keith B, Johnson RS, Simon MC, Nat Rev Cancer 2011, 12, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Hsieh AL, Walton ZE, Altman BJ, Stine ZE, Dang CV, Semin Cell Dev Biol 2015, 43, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Bass J, Takahashi JS, Science 2010, 330, 1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Dibner C, Schibler U, Albrecht U, Annu Rev Physiol 2010, 72, 517. [DOI] [PubMed] [Google Scholar]
- [95].Dibner C, Schibler U, J Intern Med 2015, 277, 513. [DOI] [PubMed] [Google Scholar]
- [96].Partch CL, Green CB, Takahashi JS, Trends Cell Biol 2014, 24, 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Takahashi JS, Nat Rev Genet 2017, 18, 164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Bass J, Nature 2012, 491, 348. [DOI] [PubMed] [Google Scholar]
- [99].Zhang R, Lahens NF, Ballance HI, Hughes ME, Hogenesch JB, Proc Natl Acad Sci U S A 2014, 111, 16219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Pizarro A, Hayer K, Lahens NF, Hogenesch JB, Nucleic Acids Res 2013, 41, D1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Koike N, Yoo SH, Huang HC, Kumar V, Lee C, Kim TK, Takahashi JS, Science 2012, 338, 349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Panda S, Science 2016, 354, 1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Krishnaiah SY, Wu G, Altman BJ, Growe J, Rhoades SD, Coldren F, Venkataraman A, Olarerin-George AO, Francey LJ, Mukherjee S, Girish S, Selby CP, Cal S, Er U, Sianati B, Sengupta A, Anafi RC, Kavakli IH, Sancar A, Baur JA, Dang CV, Hogenesch JB, Weljie AM, Cell Metab 2017, 25, 961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Janich P, Arpat AB, Castelo-Szekely V, Lopes M, Gatfield D, Genome Res 2015, 25, 1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Zheng X, Sehgal A, Curr Biol 2010, 20, 1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Cao R, Gkogkas CG, de Zavalia N, Blum ID, Yanagiya A, Tsukumo Y, Xu H, Lee C, Storch KF, Liu AC, Amir S, Sonenberg N, Nat Neurosci 2015, 18, 855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Cao R, Robinson B, Xu H, Gkogkas C, Khoutorsky A, Alain T, Yanagiya A, Nevarko T, Liu AC, Amir S, Sonenberg N, Neuron 2013, 79, 712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Cao R, Li A, Cho HY, Lee B, Obrietan K, J Neurosci 2010, 30, 6302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Ramanathan C, Kathale ND, Liu D, Lee C, Freeman DA, Hogenesch JB, Cao R, Liu AC, PLoS Genet 2018, 14, e1007369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Wolfson RL, Chantranupong L, Saxton RA, Shen K, Scaria SM, Cantor JR, Sabatini DM, Science 2016, 351, 43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Saxton RA, Knockenhauer KE, Wolfson RL, Chantranupong L, Pacold ME, Wang T, Schwartz TU, Sabatini DM, Science 2016, 351, 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Wang S, Tsun ZY, Wolfson RL, Shen K, Wyant GA, Plovanich ME, Yuan ED, Jones TD, Chantranupong L, Comb W, Wang T, Bar-Peled L, Zoncu R, Straub C, Kim C, Park J, Sabatini BL, Sabatini DM, Science 2015, 347, 188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Gu X, Orozco JM, Saxton RA, Condon KJ, Liu GY, Krawczyk PA, Scaria SM, Harper JW, Gygi SP, Sabatini DM, Science 2017, 358, 813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Wolfson RL, Sabatini DM, Cell Metab 2017, 26, 301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].Dibble CC, Manning BD, Nat Cell Biol 2013, 15, 555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Thoreen CC, Chantranupong L, Keys HR, Wang T, Gray NS, Sabatini DM, Nature 2012, 485, 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Nandagopal N, Roux PP, Translation (Austin) 2015, 3, e983402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Meyuhas O, Int Rev Cell Mol Biol 2015, 320, 41. [DOI] [PubMed] [Google Scholar]
