Skip to main content
PLOS Biology logoLink to PLOS Biology
. 2022 Mar 31;20(3):e3001594. doi: 10.1371/journal.pbio.3001594

Folliculin promotes substrate-selective mTORC1 activity by activating RagC to recruit TFE3

Kristina Li 1,2,#, Shogo Wada 1,#, Bridget S Gosis 1, Chelsea Thorsheim 1, Paige Loose 1, Zolt Arany 1,*
Editor: Anne Simonsen3
PMCID: PMC9004751  PMID: 35358174

Abstract

Mechanistic target of rapamycin complex I (mTORC1) is central to cellular metabolic regulation. mTORC1 phosphorylates a myriad of substrates, but how different substrate specificity is conferred on mTORC1 by different conditions remains poorly defined. Here, we show how loss of the mTORC1 regulator folliculin (FLCN) renders mTORC1 specifically incompetent to phosphorylate TFE3, a master regulator of lysosome biogenesis, without affecting phosphorylation of other canonical mTORC1 substrates, such as S6 kinase. FLCN is a GTPase-activating protein (GAP) for RagC, a component of the mTORC1 amino acid (AA) sensing pathway, and we show that active RagC is necessary and sufficient to recruit TFE3 onto the lysosomal surface, allowing subsequent phosphorylation of TFE3 by mTORC1. Active mutants of RagC, but not of RagA, rescue both phosphorylation and lysosomal recruitment of TFE3 in the absence of FLCN. These data thus advance the paradigm that mTORC1 substrate specificity is in part conferred by direct recruitment of substrates to the subcellular compartments where mTORC1 resides and identify potential targets for specific modulation of specific branches of the mTOR pathway.


How does the mTORC1 complex, which influences myriad cellular processes, achieve specificity? This study shows that substrate specificity is in part conferred by modulating the recruitment of substrates, in this case the transcription factor TFE3, to mTORC1.

Introduction

The ability of a cell to sense and respond to the intracellular and extracellular environment is vital for it to maintain metabolic homeostasis. Doing so is also fundamentally necessary for the cell to align its metabolic programming to ongoing cellular physiological needs. A major component of sensory integration occurs at the mechanistic target of rapamycin complex I (mTORC1) kinase complex [15]. This multisubunit complex integrates numerous inputs, including signals from growth factors, ambient levels of various amino acids (AAs), the cellular energy state, and hypoxia and DNA damage. In turn, it regulates multiple metabolic programs, for example, promoting anabolic processes such as lipid and protein synthesis, while inhibiting catabolic processes such as autophagy and lysosome biogenesis [15].

The mTORC1 complex, nucleated around the adaptor protein Raptor, is recruited to the lysosome membrane upon AA sufficiency and then activated by Rheb in response to growth factors, achieved by relieving the repression of Rheb by the TSC complex [15]. AA sensing by mTORC1 is complex, including sensing of leucine by Sestrin and sensing of arginine by SLC38A9. In response to these integrated inputs, mTOR phosphorylates a myriad of targets, including p70S6K and 4EBP1 to promote protein translation and ribosome biogenesis, ULK1 to suppress autophagy, Lipin1 to promote lipid synthesis, and the TFE3/B transcription factors to suppress lysosome biogenesis [15]. The mTORC1 pathway is thus often depicted as monolithic, acting as a single on/off switch that senses dozens of upstream informational inputs and integrates them into the single response of phosphorylating its multiple targets [15]. However, such a monochromatic model of central control of cellular homeostasis is highly unlikely to be accurate.

We have recently identified a substrate-specific branch of mTORC1 signaling, providing the first example of specific regulation of different branches of mTORC1 signaling [6,7], subsequently also reported by the Zoncu and Ballabio groups [8,9]. In this pathway, the protein folliculin (FLCN) regulates mTORC1-mediated phosphorylation of only TFE3/B, while not affecting phosphorylation of other canonical substrates such as S6K and 4EBP1. Thus, deletion of FLCN completely abrogates phosphorylation of TFE3, releasing it from 14-3-3 binding and cytoplasmic sequestration and allowing its nuclear translocation to drive genes of lysosome and mitochondria biogenesis. In contrast, deletion of FLCN does not disable phosphorylation of canonical substrates like S6K and 4EBP1 [6,7]. Understanding how, mechanistically, FLCN confers this substrate specificity onto the mTORC1 complex is thus of significant interest.

FLCN is a GTPase-activating protein (GAP) and thus stimulator of the small G-proteins RagC and D, which are active in their GDP-bound state [10]. RagC and D heterodimerize with RagA or B to incorporate into the mTORC1 complex and positively regulate mTORC1 activity. Structures elucidated by cryoEM reveal FLCN to bind directly to RagC/D [8,11], confirming earlier coprecipitation studies [12], and prior work has indicated that RagC binds to TFE3 [13]. We thus hypothesized here that the mechanism by which FLCN modulates only the TFE3/B arm of mTORC1 signaling is by activating RagC to recruit TFE3 to the mTORC1 complex, i.e., achieving substrate specificity via specific recruitment of substrate to the complex. While the work that we report here was being finalized, the Ballabio group reported overlapping findings with TFEB [9].

Results

TFE3 phosphorylation is responsive to AAs, via the GATOR complex

To begin to investigate the specific regulation of TFE3 phosphorylation, we tested the impact on TFE3 phosphorylation by known upstream regulators of canonical mTORC1 activity: growth factors and AAs. C2C12 cells were grown in complete media containing growth factor-rich 10% fetal bovine serum (FBS). The cells were then changed for 60 minutes into either complete media, media lacking AAs but containing dialyzed FBS (dFBS), media with AAs but no FBS, or media with neither. As seen in the “NTC” columns of Fig 1A, phosphorylation of TFE3 at S320, the mTORC1-targeted site, detected with a phospho-specific antibody, was seen in full media and media lacking serum, but not in media lacking AAs (quantification in S1 Fig). Consistent with its dephosphorylation, TFE3 translocated to the nucleus in the absence of AAs (Fig 1B, “NTC”). Thus, TFE3 phosphorylation depends more on AA sensing than on growth factor sensing.

Fig 1. TFE3 phosphorylation is responsive to AAs, via the GATOR complex.

Fig 1

(A, B) Control C2C12 cells (NTC) or cells lacking Flcn, Tsc2, or Depdc5, as indicated, were switched from complete medium to media lacking serum and/or AAs, as indicated, for 60 minutes, followed by immunoblotting for TFE3, phospho-TFE3, S6K, and phospho-S6K (A) or immunohistochemistry for subcellular localization of TFE3 (B). (C) The same cells as in A, after 60 minutes in medium lacking AAs, were returned to complete media for the indicated time points and immunoblotted for S6K, and phospho-S6K. Values were normalized to the 15-minute time point of each line. (D, E) C2C12 cells lacking Flcn, Depdc5, or both, as indicated, were evaluated as in A and B. The data underlying all the graphs shown in the figure is included in S1 Data. AA, amino acid; FLCN, folliculin; KO, knockout.

Canonical mTORC1 signaling senses growth factor signals via the inactivation of the repressive TSC complex and senses the presence of leucine via inactivation of the repressive GATOR1 complex [1]. Thus, CRISPR/Cas-9–mediated deletion of either Tsc2 (an obligatory component of TSC) or of Depdc5 (an obligatory component of GATOR1) led to constitutive phosphorylation of the canonical target S6K, even in the absence of AAs or serum (Fig 1A, “Tsc2KO” and “Depdc5KO”). In contrast, only deletion of Depdc5 led to constitutive phosphorylation of TFE3, while deletion of Tsc2 did not. These data lead to 2 conclusions: First, growth factor signaling via TSC inhibition cannot promote phosphorylation of TFE3, thus separating canonical and noncanonical signals. Second, phosphorylation of TFE3 in response to AAs is mediated in large part via DEPDC5.

We have shown previously that FLCN regulates the phosphorylation of TFE3 [6,7], as first described by the Linehan group [14]. Consistent with this, under all conditions tested, cells lacking Flcn also lacked any detection of TFE3 phosphorylation (Fig 1A, “FlcnKO”). In contrast, deletion of Flcn had no impact on S6K phosphorylation under any of the conditions tested. To investigate if FLCN may have a specific role in the kinetics of S6K phosphorylation in response to AAs, we also performed a time course after AA replenishment (Fig 1C). At no time point, however, was S6K phosphorylation altered in the cells lacking Flcn (Fig 1C). We conclude that AA sensing is intact in cells lacking Flcn and that FLCN is entirely dispensable for canonical AA signaling to S6K, again separating canonical and noncanonical signals. Of note, while concordant with our prior observations in other cell types [6,7,9], these findings differ from those of Tsun and colleagues [10], perhaps reflecting our use of complete knockout via CRISPR, in contrast to the siRNA approach taken by Tsun and colleagues.

Finally, to test if AA sensing via GATOR promotes TFE3 phosphorylation via FLCN, we generated cells lacking both Depdc5 and Flcn (dKO cells). As seen in Fig 1D, Flcn was epistatic to Depdc5, i.e., loss of Depdc5 failed to activate phosphorylation of TFE3 in the absence of FLCN, whether in the presence or absence of AAs and serum. Together, these data demonstrate that the presence of AAs is necessary and sufficient to promote phosphorylation of TFE3 by mTORC1, and does so via GATOR1 and FLCN, while growth factor signaling via TSC inhibition does not promote phosphorylation of TFE3, in sharp contrast to canonical phosphorylation of S6K.

RagC, but not RagA, promotes TFE3 phosphorylation in response to AAs

FLCN is a GAP for the highly similar Rags C and D, either of which heterodimerizes with either RagA or B to activate mTORC1 in response to AAs. RagC and D are active in the GDP-bound form, while RagA and B are active in the GTP-bound form. To test which Rag type primarily drives TFE3 phosphorylation, we expressed in C2C12 or 293T cells HA-tagged wild type (WT) or constitutively active RagA (66L mutant, mimicking GTP-bound state) or RagC (75L, mimicking GDP-bound state) [15]. In the absence of AAs, neither WT construct was able to rescue phosphorylation of either S6K (canonical signal) or TFE3 (noncanonical) (Fig 2A, S2A Fig). Constitutively active RagA (66L) efficiently reactivated phosphorylation of S6K in C2C12 cells (but not in 293T cells, reflecting cell-specific effects), while having little impact on TFE3 phosphorylation (Fig 2A). In sharp contrast, constitutively active RagC (75L) reactivated TFE3 phosphorylation in both cell types, while having no impact on S6K, despite the relatively lower expression of RagC 75L protein (Fig 2A, S2A Fig). The latter in part reflected higher protein instability of RagC 75L protein, as revealed by treating cells with the proteasome inhibitor MG132 (S2B Fig). Coexpression of RagC 75L and RagA 66L had no impact on TFE3 phosphorylation beyond that conferred by RagC 75L alone (S2A Fig). Consistent with these findings, only RagC 75L promoted cytoplasmic sequestration of TFE3 in the absence of AA (Fig 2B).

Fig 2. RagC, but not RagA, promotes TFE3 phosphorylation in response to AAs.

Fig 2

(A, B) C2C12 cells expressing HA-tagged WT or constitutive active RagA (GTP) or RagC (GDP) were switched from complete medium to media lacking AAs, as indicated, followed by immunoblotting for TFE3, phospho-TFE3, S6K, phospho-S6K, and HA (A) or immunohistochemistry for subcellular localization of TFE3 (B). Quantification of cytoplasmic-to-nuclear ratio of TFE3 is shown below the images. Scale bar: 20 μm, ****p < 0.0001 by Student t test. (C) The same cells as in A, subjected to a time course after withdrawal of AAs, followed by immunoblotting for phospho-TFE3, phospho-4EBP, and phospho-S6K. Densitometric quantification is shown below. The data underlying all the graphs shown in the figure is included in S1 Data. AA, amino acid; dFBS, dialyzed FBS; WT, wild type.

To evaluate the kinetics of this process, we treated cells with complete media, switched the cells to media lacking AAs and then evaluated dephosphorylation of TFE3, S6K, and 4EBP serially over 120 minutes (Fig 2C, S2C Fig). Removal of AAs led to 90% dephosphorylation of TFE3 by 30 minutes, and 80% dephosphorylation of S6K and 4EBP by 45 minutes. Expression of constitutively active RagC 75L delayed dephosphorylation on TFE3, while having little impact on S6K and 4EBP. Conversely, expression of constitutively active RagA 66L largely maintained phosphorylation on S6K and 4EBP, while having little impact on TFE3. The latter is consistent with the observation in Fig 1D that activation of RagA, conferred by deletion of its inhibitor DEPDC5, is not sufficient to rescue TFE3 phosphorylation in the absence of FLCN. These data thus demonstrate clearly separable Rag-mediated pathways, whereby RagA is sufficient to promote canonical signaling to S6K and 4EBP in response to AA stimulation, while RagC is sufficient to promote signaling to TFE3 without simultaneous additional activation of RagA beyond its preexisting baseline activity.

Active RagC rescues TFE3 phosphorylation in the absence of FLCN, while active RagA does not

To test if RagC confers specificity on noncanonical signaling to TFE3, WT and constitutively active RagA and C were expressed in C2C12 cells lacking Flcn and grown in complete media (Fig 3A). Despite the presence of AAs, TFE3 remained unphosphorylated, reflecting the absence of Flcn. Strikingly, only expression of RagC 75L could rescue phosphorylation of TFE3 in these cells (Fig 3A). Consistent with this, only RagC 75L could promote cytoplasmic sequestration of TFE3 in FLCN knockout cells (Fig 3B). When activated and nuclear, TFE3 is known to drive a broad genetic program, including upregulating expression of lysosome proteins (Mcoln1, Neu1, Hexa, Atp6v0e, Ctsa, Ctsb, and Gpnmb) [13], regulators of mitochondrial biogenesis (Ppargc1a) [16], and a positive feedback loop to mTORC1 via Ragd [7,17]. All of these genes were dramatically induced in the absence of FLCN (Fig 3C). Furthermore, only the expression of RagC 75L prevented their induction, consistent with the phosphorylation and cytoplasmic sequestration of TFE3 (Fig 3C). Thus, we conclude that RagC is epistatic to FLCN, i.e., that FLCN promotes TFE3 phosphorylation via RagC and that RagC confers substrate specificity to the mTORC1 complex.

Fig 3. Constitutively active RagC, but not RagA, rescues TFE3 phosphorylation in the absence of FLCN.

Fig 3

(A, B) Control C2C12 cells and cells lacking Flcn were transduced with HA-tagged WT or constitutive active RagA (66L) or RagC (75L), followed by immunoblotting for FLCN, TFE3, phospho-TFE3, S6K, and phospho-S6K (A), immunohistochemistry for subcellular localization of TFE3 (B), or quantitative PCR evaluation of expression of the indicated genes (normalized to the average expression of HPRT, TBP, and 36B4 as controls) (C). Scale bar: 10 μm, **p < 0.01, ***p < 0.001, ****p < 0.0001 by Student t test. The data underlying all the graphs shown in the figure is included in S1 Data. FLCN, folliculin; KO, knockout; WT, wild type.

AA stimulation drives transient localization of TFE3 to lysosome via FLCN and RagC

To begin to evaluate how RagC confers substrate specificity to the mTORC1 complex, we evaluated TFE3 subcellular localization in response to AA stimulation. Puertollano’s group reported that, in nutrient-replete cells, TFE3 transiently translocates to the lysosome, where it is phosphorylated by mTORC1, followed by binding to 14-3-3 and cytoplasmic sequestration [13]. The transient translocation to the lysosome can be captured by inhibiting mTORC1 activity with Torin1 (Fig 4A, top panel, costaining TFE3 with LAMP2, a lysosome marker). Strikingly, in cells lacking Flcn, TFE3 entirely fails to translocate to the lysosome (Fig 4A, bottom panel). Thus, FLCN serves the critical function of recruiting substrate (TFE3) to mTORC1. Equally strikingly RagC 75L entirely rescued the translocation of TFE3 to the lysosome in cells lacking Flcn (Fig 3B). These data demonstrate that activation of RagC, occurring physiologically via FLCN GAP activity, recruits TFE3 to the lysosome, leading to its phosphorylation and cytoplasmic retention.

Fig 4. AA stimulation drives transient localization of TFE3 to lysosome via FLCN and RagC.

Fig 4

(A) Control C2C12 cells and cells lacking Flcn were maintained for 60 minutes in medium lacking AAs and then returned to complete media in the presence of Torin1 for 15 minutes, followed by immunohistochemistry for subcellular localization of TFE3 and LAMP2, a marker of the lysosome. Right: correlation by Pearson’s R of LAMP2 and TFE3 staining. (B) Cells lacking Flcn were transduced with HA-tagged WT or constitutive active RagA (66L) or RagC (75L), followed by immunohistochemistry as in A. ***p < 0.001 by Student t test (n = 3). Scale bar: 10 μm. The data underlying all the graphs shown in the figure is included in S1 Data. AA, amino acid; FLCN, folliculin; KO, knockout; WT, wild type.

RagC is necessary and sufficient for AA-stimulated TFE3 localization to lysosome and subsequent phosphorylation

The data above demonstrated the sufficiency of activated RagC to drive TFE3 lysosome localization and phosphorylation. To test if RagC is required for this process, we generated by CRISPR/Cas-9 C2C12 cells lacking RagC (S3A Fig). These cells revealed a near complete block of TFE3 phosphorylation in response to AA stimulus (Fig 5A). In clear contrast, canonical phosphorylation of S6K in response to AA was entirely unaffected in these RagC knockout cells (Fig 5A). No compensatory induction of RagD was appreciated in the absence of RagC (S3B Fig). Thus RagC is required for AA signaling to TFE3, but dispensable for AA signaling to S6K, clearly separating the 2 arms of mTORC1 signaling. Note that despite being dispensable, there is evidence that overexpression of RagC mutants can suppress S6K phosphorylation, likely working in a dominant-negative fashion [18]. Evaluation of TFE3 subcellular localization in response to AA stimulus revealed that RagC was equally required for the recruitment of TFE3 to the lysosome (Fig 5B). Thus, we find that activation of RagC by FLCN is both necessary and sufficient to recruit TFE3 to the lysosome and to promote its phosphorylation, without simultaneous additional activation of RagA.

Fig 5. RagC is necessary for AA-stimulated TFE3 phosphorylation and localization to lysosome.

Fig 5

(A) Control C2C12 cells and cells lacking RagC were maintained for 60 minutes in medium lacking AAs and then returned to complete media for the indicated times, followed by immunoblotting as indicated. Phospho-TFE3/totalTFE3 and phospho-S6K/totalS6K were quantified and normalized to the 15-minute time point of NTC (control). (B) Control C2C12 cells and cells lacking RagC were cultured with 250 nM Torin1 for 15 minutes, followed by immunohistochemistry for subcellular localization of TFE3 and LAMP2, a marker of the lysosome. (Scale bar: 10 μm). On the right: correlation by Pearson’s R of LAMP2 and TFE3 staining. (C) Quantitative PCR evaluation of expression of the indicated genes (normalized to the average expression of HPRT, TBP, and 36B4 as controls). **p < 0.01, ***p < 0.001, ****p < 0.0001 by Student t test. The data underlying all the graphs shown in the figure is included in S1 Data. AA, amino acid; KO, knockout.

Discussion

The mechanisms by which mTORC1 integrates upstream signals and transmits them downstream has been extensively and elegantly characterized [1]. However, how such a complex integrator of multiple inputs achieves specificity in its outputs has received little attention. We first demonstrated clearly that one branch of mTORC1 output could be independently regulated from another, i.e., we showed that loss of FLCN, a RagC/D GAP, abrogated mTORC1-mediated phosphorylation of TFE3 while having no impact on canonical phosphorylation of S6K and 4EBP [6,7,19]. The impact in vivo of such selective regulation in different cell types included beiging of adipocytes and chronic activation of monocytes. Lacking from these studies, however, was a clear mechanistic understanding of how FLCN confers substrate specificity on mTORC1. We elucidate here this mechanism of substrate specificity, whereby FLCN activates RagC to its GDP-bound form via its GAP activity; activated RagC then physically recruits TFE3 to lysosome surface, thereby promoting its phosphorylation by mTORC1; phosphorylated TFE3 is then bound to 14-3-3 and sequestered in the cytoplasm, thus suppressing TFE3 target pathway activation in the nucleus. Similar findings were recently reported for the regulation of TFEB [9]. Importantly, these events occur independently of Rheb and RagA-mediated regulation of canonical phosphorylation of S6K and 4EBP. Disruption of GATOR1, a GAP of RagA/B, renders mTORC1 insensitive to AA withdrawal, maintaining TFE3 as well as canonical substrates phosphorylated even in the absence of AAs. Concomitant loss of FLCN selectively blunted TFE3 phosphorylation while phosphorylation status of canonical substrates remains insensitive to AA withdrawal, further supporting the separable branches of mTORC1.

Heterozygous loss-of-function germline mutations in FLCN cause Birt–Hogg–Dubé (BHD) syndrome, which is marked by chronic development of lung cysts, abundant benign dermal hamartoma-like tumors, and a high incidence of renal cell carcinoma (RCC) [20]. Both the dermal tumors and RCC are characterized by high canonical mTORC1 activity and yet occur in the context of loss of heterozygosity (LOH), i.e., loss of FLCN-mediated activation of mTORC1. The existence of the substrate-specific mechanism described here helps to explain this seeming paradox: loss of FLCN unleashes TFE3 to the nucleus, but has no direct impact on canonical mTORC1 signaling. Moreover, as we have shown before [7], an indirect positive feedback loop explains how in some cell types canonical mTORC1 activity in fact increases in the absence of FLCN: Nuclear TFE3 strongly induces gene expression of RagD [17], which can drive canonical mTORC1 phosphorylation of S6K even in the absence of FLCN [7]. The mechanistic separation of mTORC1 signaling into FLCN-independent (canonical) and FLCN-dependent (noncanonical) arms thus explains the apparent paradoxical development of tumors with high mTORC1 activity in BHD patients.

We note evidence of 2 reciprocal feedback loops between these 2 arms of mTORC1 signaling. On the one hand, inactivation of FLCN can lead to RagD-mediated activation of canonical S6K phosphorylation, as described above. Conversely, we also note that constitutive activation of canonical signaling, achieved via deletion of Tsc2, leads to reciprocal partial suppression of TFE3 phosphorylation (Fig 1). This observation is consistent with a previous study, in which unbiased genetic screens revealed TSC to act upstream of FLCN and TFE3 in the regulation of exit from pluripotency in embryonic stem cells [21]. The mechanism for this second feedback loop remains unclear.

TFE3 is member of a small family of bHLH-ZIP-type transcription factor that includes TFEB, TFEC, and MITF [22]. Interestingly, TFE3 translocations and gene duplications (i.e., gain-of-function variants) are a relatively common cause of kidney cancer, associated with high mTORC1 activity, thus mimicking the effects of FLCN deletion in BHD syndrome [23]. TFEB and MITF mutations have also been noted in kidney cancers, albeit more rarely. Genetic deletion of Flcn in the kidney in mice yields severe polycystic disease, but not frank cancer, indicating that additional genetic hits are likely required to develop cancer. Ballabio’s group recently showed that codeletion of Tfeb rescues the polycystic phenotype of kidney-specific Flcn deletion [9]. In the same study, the authors show similar effects of RagC on TFEB as we show here on TFE3. There is thus likely a fair amount of similarity between TFE3 and TFEB pathways. The fact that deletion of either Tfeb or Tfe3 abrogates the effect of Flcn deletion suggests that TFE3 and TFEB may heterodimerize, although such interaction has not been reported to date. Alternatively, TFE3 and TFEB perform different functions in different tissues, as suggested by, for example, the lethality of whole-body deletion of Tfeb, while Tfe3 knockout mice are viable, with little baseline phenotype [24,25].

In summary, we elucidate here the mechanistic basis by which FLCN confers substrate specificity upon the mTORC1 complex: FLCN activates RagC to physically recruit TFE3 to the mTORC1 complex, promoting TFE3 phosphorylation while having little impact on canonical substrates such as S6K. Our work, combined with similar work with TFEB [9], mechanistically exposes the first clear example of parsing of mTORC1 signaling.

Materials and methods

Cell culture

Mouse C2C12 myoblasts were cultured in Gibco Dulbecco’s Modified Eagle Medium (DMEM) with high glucose and GlutaMAX (Invitrogen 10569010, MA, USA) supplemented with 10% FBS and 1% penicillin streptomycin (Invitrogen 15140122). Cells were incubated in 37°C and 5% CO2. DMEM media was changed every 2 days and split with trypsin (Invitrogen 25200056) when cells reached 95% confluence.

Nutrient withdrawal and restimulation experiment

For AA withdrawal experiments, cells were washed with sterile PBS and AA-free DMEM with 10% dFBS was placed on cells for specified times. For restimulation experiments, AA free media was replaced with complete media (DMEM with 10% FBS).

Antibodies

Phospho-TFE3 (Ser320) antibody was a gift from Dr. Rosa Puertollano and previously described [13]. Other antibodies used are as follows: total TFE3 (Cell Signaling Technology, 14779, MA, USA), phospho-p70S6K (Thr389) (Cell Signaling Technology, 9234), HA tag (Cell Signaling Technology, 2367), LAMP2 (Abcam, ab13524, MA, USA), FLCN (Abcam, ab124885), total p70S6K (Cell Signaling Technology, 2708), beta-actin (Cell Signaling Technology, 4970), and 14-3-3 (Cell Signaling Technology, 8312)

Gene deletion by CRISPR/Cas-9 system

lentiCRISPR version 2 was a gift from Feng Zhang (Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA) (Addgene, plasmid 52961). The guide RNAs (gRNAs) were designed using the Optimized CRISPR Design website (http://crispr.mit.edu) from Zhang Lab (Cambridge, MA, USA). Mouse C2C12 myoblast cells were infected with lentivirus encoding for the Cas-9/gRNA, selected using puromycin, and validated using western blot before being used as populations. The gRNA sequence was as follows (the pam sequence is excluded): mouse nontarget control (5′-ATTGTTCGACCGTC TACGGG-3′), mouse flcn (5′-TCCGTGCAGAAGAGCGTGCG-3′), mouse tsc2 (5′-TTGATGCAATGTATTCGTCA-3′), mouse depdc5 (5′-GACAAGTTTGTAGACCTTTG-3′), and mouse RagC (5′-GGACTTCGGCTACGGCGTGG-3′).

Lenti and retro virus production

Lenti transfer plasmid, psPAX2 (Addgene, plasmid 12260), and pMD2.G (Addgene, plasmid 12259) (both gifts from Didier Trono, School of Life Sciences, EcolePolytechnique Federale de Lausanne, Lausanne, Switzerland) were cotransfected onto HEK293T cells using Lipofectamine 3000 (Thermo Fisher Scientific, MA, USA). Plasmids were removed after 18 hours on HEK293T cells, and media was replenished. After 48 hours, conditioned media was collected and passed through a low protein binding 0.45-μm syringe filter to remove cell debris. Mouse C2C12 myoblasts were plated on 6-well multiwell plates and infected via spinfection method. Polybrene was added to the virus containing media (final concentration of 8 μg/mL) and added on top of the C2C12 cells. Each 6-well plate was centrifuged for 90 minutes at 1,000 g (spinfection), and media was replaced. Stable cells were selected 24 hours postspinfection with the appropriate antibiotic.

Western blot

Cell culture samples were lysed with RIPA buffer with a proteinase inhibitor (Complete miniproteinase inhibitor cocktail, Roche, BS, CH) and a phosphatase inhibitor (PhosSTOP, Roche). Samples were sonicated and spun down to remove lipid and insoluble debris. A BCA protein assay kit (Thermo Fisher Scientific) was used to quantify and normalize protein concentrations. The same amount of protein (10 to 20 μg) was loaded on to a 4% to 20% gradient Tris-glycine polyacrylamide gel (Bio-Rad, CA, USA) and electrophoresed (SDS-PAGE). Samples were transferred to PVDF membrane (MilliporeSigma, MA, USA) and blocked with 5% milk for 1 hour and incubated with primary antibody overnight. The following day, membranes were washed with TBS-T and incubated in appropriate HRP-conjugated secondary antibody for 60 minutes. Images were taken using the ImageQuant LAS 4000 (GE Healthcare Life Sciences, NJ, USA).

Immunoprecipitation

Cells were washed with cold PBS and lysed with IP Lysis Buffer (Thermo Fisher Scientific) with proteinase inhibitor (Complete miniproteinase inhibitor cocktail, Roche) and a phosphatase inhibitor (PhosSTOP, Roche). Samples were sonicated and centrifuged. Supernatant was added to Pierce Anti-HA Magnetic Beads (Thermo Fisher Scientific) and incubated at room temperature for 30 minutes with end-over-end mixing. Samples were washed 3 times with TBS-T, reconstituted in 1xSDS RIPA, and boiled at 95°C for 5 minutes. HA beads were removed.

Immunocytochemistry

Cells were grown on glass coverslips precoated with collagen type I. The following day, cells were fixed with 4% paraformaldehyde (Thermo Fisher Scientific) for 15 minutes. Cells were washed with PBS and incubated in blocking buffer (1XPBS/5% Normal Goat Serum/0.3% Triton X-100) for 60 minutes at room temperature. Primary antibodies were diluted as indicated on respective datasheets in blocking buffer and incubated overnight on cells at 4°C. Cells were washed with PBS and incubated in appropriate Alexa Flour (Thermo Fisher Scientific) secondary antibodies diluted in blocking buffer for 1 hour at room temperature in the dark. After washing with PBS, coverslips were mounted with Prolong Gold Antifade Reagent with DAPI (Cell Signaling Technology). Images were captured using the Zeiss LSM 710 (BW, DE) confocal microscope, and image analysis was done using ImageJ.

Supporting information

S1 Fig. TFE3 phosphorylation is responsive to AAs, via the GATOR complex.

(A) Control C2C12 cells (NTC) or cells lacking Flcn, Tsc2, or Depdc5 were switched from complete medium to media lacking serum and/or AAs for 60 minutes followed by immunoblotting. Images were uploaded into ImageJ, and signal intensity was quantified. Graphed above is the ratio of p-TFE3 to total TFE3 signal. (B) Quantification and graph of ratio of pS6K to total S6K signal. The data underlying all the graphs shown in the figure is included in S1 Data. AA, amino acid; FLCN, folliculin.

(PDF)

S2 Fig. RagC, but not RagA, promotes TFE3 phosphorylation in response to AAs.

(A) Coexpression of active RagA and RagC in 293T cells does not confer further phosphorylation of TFE3 compared to active RagC alone. (B) RagC 75L protein is partially stabilized by inhibition of the proteasome with MG132. (C) To accompany main Fig 2C, immunoblotting for total levels of TFE3, S6K1, and 4E-BP in C2C12 cells expressing HA-tagged WT, or constitutive active RagA (GTP) or RagC (GDP), demonstrate equivalent expression of these proteins at all time points after switching from complete medium to media lacking AAs. AA, amino acid; WT, wild type.

(PDF)

S3 Fig. RagD expression is not increased as compensation for RagC CRISPR KO or siRNA knockdown.

(A) C2C12 cells with RagC CRISPR KO were used for Fig 5A. (B) RagC CRISPR KO (in C2C12s) and RagC siRNA knockdown (in HEK 293Ts) showed no significant compensation of RagD expression. The data underlying all the graphs shown in the figure is included in S1 Data. KO, knockout.

(PDF)

S1 Data. Data underlying graphs in main and Supporting information figures.

(XLSX)

S1 Raw Images. Raw images.

(PDF)

Abbreviations

AA

amino acid

BHD

Birt–Hogg–Dubé

dFBS

dialyzed FBS

DMEM

Dulbecco’s Modified Eagle Medium

FLCN

folliculin

GAP

GTPase-activating protein

gRNA

guide RNA

LOH

loss of heterozygosity

mTORC1

mechanistic target of rapamycin complex I

RCC

renal cell carcinoma

WT

wild type

Data Availability

All relevant data are within the paper and its Supporting Information files.

Funding Statement

SW was supported by a postdoctoral fellowship from the American Diabetes Association, BG was supported by the National Institutes of Health (NIH) (F30) and the Blavatnik Family Foundation, and ZA was supported by the NIH (R01 DK107667). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Liu GY, Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol. 2020;21(4):183–203. Epub 2020/01/16. doi: 10.1038/s41580-019-0199-y ; PubMed Central PMCID: PMC7102936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Condon KJ, Sabatini DM. Nutrient regulation of mTORC1 at a glance. J Cell Sci. 2019;132(21). Epub 2019/11/15. doi: 10.1242/jcs.222570 ; PubMed Central PMCID: PMC6857595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Saxton RA, Sabatini DM. mTOR Signaling in Growth, Metabolism, and Disease. Cell. 2017;168 (6):960–76. doi: 10.1016/j.cell.2017.02.004 ; PubMed Central PMCID: PMC5394987 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kennedy BK, Lamming DW. The Mechanistic Target of Rapamycin: The Grand ConducTOR of Metabolism and Aging. Cell Metab. 2016;23(6):990–1003. Epub 2016/06/16. doi: 10.1016/j.cmet.2016.05.009 ; PubMed Central PMCID: PMC4910876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ben-Sahra I, Manning BD. mTORC1 signaling and the metabolic control of cell growth. Curr Opin Cell Biol. 2017;45:72–82. Epub 2017/04/16. doi: 10.1016/j.ceb.2017.02.012 ; PubMed Central PMCID: PMC5545101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Wada S, Neinast M, Jang C, Ibrahim YH, Lee G, Babu A, et al. The tumor suppressor FLCN mediates an alternate mTOR pathway to regulate browning of adipose tissue. Genes Dev. 2016;30(22):2551–64. Epub 2016/12/04. doi: 10.1101/gad.287953.116 ; PubMed Central PMCID: PMC5159669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Li J, Wada S, Weaver LK, Biswas C, Behrens EM, Arany Z. Myeloid Folliculin balances mTOR activation to maintain innate immunity homeostasis. JCI Insight. 2019;5. Epub 2019/03/08. doi: 10.1172/jci.insight.126939 ; PubMed Central PMCID: PMC6483010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lawrence RE, Fromm SA, Fu Y, Yokom AL, Kim DJ, Thelen AM, et al. Structural mechanism of a Rag GTPase activation checkpoint by the lysosomal folliculin complex. Science. 2019;366(6468):971–7. Epub 2019/11/02. doi: 10.1126/science.aax0364 ; PubMed Central PMCID: PMC6945816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Napolitano G, Di Malta C, Esposito A, de Araujo MEG, Pece S, Bertalot G, et al. A substrate-specific mTORC1 pathway underlies Birt-Hogg-Dube syndrome. Nature. 2020;585(7826):597–602. Epub 2020/07/03. doi: 10.1038/s41586-020-2444-0 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tsun ZY, Bar-Peled L, Chantranupong L, Zoncu R, Wang T, Kim C, et al. The folliculin tumor suppressor is a GAP for the RagC/D GTPases that signal amino acid levels to mTORC1. Mol Cell. 2013;52 (4):495–505. doi: 10.1016/j.molcel.2013.09.016 ; PubMed Central PMCID: PMC3867817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Shen K, Rogala KB, Chou HT, Huang RK, Yu Z, Sabatini DM. Cryo-EM Structure of the Human FLCN-FNIP2-Rag-Ragulator Complex. Cell. 2019;179(6):1319–29 e8. Epub 2019/11/11. doi: 10.1016/j.cell.2019.10.036 ; PubMed Central PMCID: PMC7008705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Petit CS, Roczniak-Ferguson A, Ferguson SM. Recruitment of folliculin to lysosomes supports the amino acid-dependent activation of Rag GTPases. J Cell Biol. 2013;202(7):1107–22. Epub 2013/10/02. doi: 10.1083/jcb.201307084 ; PubMed Central PMCID: PMC3787382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Martina JA, Diab HI, Lishu L, Jeong AL, Patange S, Raben N, et al. The nutrient-responsive transcription factor TFE3 promotes autophagy, lysosomal biogenesis, and clearance of cellular debris. Sci Signal. 2014;7(309):ra9. doi: 10.1126/scisignal.2004754 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hong SB, Oh H, Valera VA, Baba M, Schmidt LS, Linehan WM. Inactivation of the FLCN tumor suppressor gene induces TFE3 transcriptional activity by increasing its nuclear localization. PLoS ONE. 2010;5 (12):e15793. doi: 10.1371/journal.pone.0015793 ; PubMed Central PMCID: PMC3012117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled L, et al. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science. 2008;320(5882):1496–501. Epub 2008/05/24. doi: 10.1126/science.1157535 ; PubMed Central PMCID: PMC2475333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Salma N, Song JS, Arany Z, Fisher DE. Transcription Factor Tfe3 Directly Regulates Pgc-1alpha in Muscle. J Cell Physiol. 2015;230(10):2330–6. Epub 2015/03/05. doi: 10.1002/jcp.24978 ; PubMed Central PMCID: PMC4617629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Di Malta C, Siciliano D, Calcagni A, Monfregola J, Punzi S, Pastore N, et al. Transcriptional activation of RagD GTPase controls mTORC1 and promotes cancer growth. Science. 2017;356(6343):1188–92. Epub 2017/06/18. doi: 10.1126/science.aag2553 ; PubMed Central PMCID: PMC5730647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Yang G, Humphrey SJ, Murashige DS, Francis D, Wang QP, Cooke KC, et al. RagC phosphorylation autoregulates mTOR complex 1. EMBO J. 2019;38(3). Epub 2018/12/16. doi: 10.15252/embj.201899548 ; PubMed Central PMCID: PMC6356064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wada S, Arany Z. Adipose tissue browning: mTOR branches out. Cell Cycle. 2017;16(6):493–4. Epub 2017/02/01. doi: 10.1080/15384101.2017.1285634 ; PubMed Central PMCID: PMC5384586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Schmidt LS, Linehan WM. FLCN: The causative gene for Birt-Hogg-Dube syndrome. Gene. 2018;640:28–42. Epub 2017/10/04. doi: 10.1016/j.gene.2017.09.044 ; PubMed Central PMCID: PMC5682220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Betschinger J, Nichols J, Dietmann S, Corrin PD, Paddison PJ, Smith A. Exit from Pluripotency Is Gated by Intracellular Redistribution of the bHLH Transcription Factor Tfe3. Cell. 2013;153 (2):335–47. doi: 10.1016/j.cell.2013.03.012 ; PubMed Central PMCID: PMC3661979 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.La Spina M, Contreras PS, Rissone A, Meena NK, Jeong E, Martina JA. MiT/TFE Family of Transcription Factors: An Evolutionary Perspective. Front Cell Dev Biol. 2020;8:609683. Epub 2021/01/26. doi: 10.3389/fcell.2020.609683 ; PubMed Central PMCID: PMC7815692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Linehan WM, Schmidt LS, Crooks DR, Wei D, Srinivasan R, Lang M, et al. The Metabolic Basis of Kidney Cancer. Cancer Discov. 2019;9(8):1006–21. Epub 2019/05/16. doi: 10.1158/2159-8290.CD-18-1354 . [DOI] [PubMed] [Google Scholar]
  • 24.Steingrimsson E, Tessarollo L, Reid SW, Jenkins NA, Copeland NG. The bHLH-Zip transcription factor Tfeb is essential for placental vascularization. Development. 1998;125(23):4607–16. Epub 1998/11/10. doi: 10.1242/dev.125.23.4607 . [DOI] [PubMed] [Google Scholar]
  • 25.Yagil Z, Hadad Erlich T, Ofir-Birin Y, Tshori S, Kay G, Yekhtin Z, et al. Transcription factor E3, a major regulator of mast cell-mediated allergic response. J Allergy Clin Immunol. 2012;129(5):1357–66 e5. Epub 2012/03/01. doi: 10.1016/j.jaci.2011.11.051 . [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Ines Alvarez-Garcia

4 Mar 2021

Dear Zolt,

Thank you for submitting your manuscript entitled "FLCN promotes substrate-selective mTORC1 activity by activating RagC to recruit TFE3 to the lysosome" for consideration as a Research Article by PLOS Biology. Thank you also for your patience as we completed our editorial process, and apologies for the delay.

Your manuscript has now been evaluated by the PLOS Biology editorial staff as well as by an academic editor with relevant expertise and I am writing to let you know that we would like to send your submission out for external peer review.

However, before we can send your manuscript to reviewers, we need you to complete your submission by providing the metadata that is required for full assessment. To this end, please login to Editorial Manager where you will find the paper in the 'Submissions Needing Revisions' folder on your homepage. Please click 'Revise Submission' from the Action Links and complete all additional questions in the submission questionnaire.

Please re-submit your manuscript within two working days, i.e. by Mar 08 2021 11:59PM.

Login to Editorial Manager here: https://www.editorialmanager.com/pbiology

During resubmission, you will be invited to opt-in to posting your pre-review manuscript as a bioRxiv preprint. Visit http://journals.plos.org/plosbiology/s/preprints for full details. If you consent to posting your current manuscript as a preprint, please upload a single Preprint PDF when you re-submit.

Once your full submission is complete, your paper will undergo a series of checks in preparation for peer review. Once your manuscript has passed all checks it will be sent out for review.

Given the disruptions resulting from the ongoing COVID-19 pandemic, please expect delays in the editorial process. We apologise in advance for any inconvenience caused and will do our best to minimize impact as far as possible.

Feel free to email us at plosbiology@plos.org if you have any queries relating to your submission.

Kind regards,

Ines

--

Ines Alvarez-Garcia, PhD,

Senior Editor

PLOS Biology

ialvarez-garcia@plos.org

Decision Letter 1

Ines Alvarez-Garcia

13 May 2021

Dear Dr Arany,

Thank you very much for submitting your manuscript "FLCN promotes substrate-selective mTORC1 activity by activating RagC to recruit TFE3 to the lysosome" for consideration as a Research Article at PLOS Biology. Thank you also for your patience as we completed our editorial process, and please accept my apologies for the delay in providing you with our decision. Your manuscript has been evaluated by the PLOS Biology editors, an Academic Editor with relevant expertise, and by two independent reviewers.

As you will see, the reviews think that the conclusions are interesting and potentially significant for the field, but they also raise several concerns that would need to be addressed to confirm the findings. Both reviewers suggest several experiments to strengthen the results – including using a different cell line - mention several relevant references that you should add to give proper credit to related literature and ask you to clarify several points.

In light of the reviews (attached below), we will not be able to accept the current version of the manuscript, but we would welcome re-submission of a much-revised version that takes into account the reviewers' comments. We cannot make any decision about publication until we have seen the revised manuscript and your response to the reviewers' comments. Your revised manuscript is also likely to be sent for further evaluation by the reviewers.

We expect to receive your revised manuscript within 3 months.

Please email us (plosbiology@plos.org) if you have any questions or concerns, or would like to request an extension. At this stage, your manuscript remains formally under active consideration at our journal; please notify us by email if you do not intend to submit a revision so that we may end consideration of the manuscript at PLOS Biology.

**IMPORTANT - SUBMITTING YOUR REVISION**

Your revisions should address the specific points made by each reviewer. Please submit the following files along with your revised manuscript:

1. A 'Response to Reviewers' file - this should detail your responses to the editorial requests, present a point-by-point response to all of the reviewers' comments, and indicate the changes made to the manuscript.

*NOTE: In your point by point response to the reviewers, please provide the full context of each review. Do not selectively quote paragraphs or sentences to reply to. The entire set of reviewer comments should be present in full and each specific point should be responded to individually, point by point.

You should also cite any additional relevant literature that has been published since the original submission and mention any additional citations in your response.

2. In addition to a clean copy of the manuscript, please also upload a 'track-changes' version of your manuscript that specifies the edits made. This should be uploaded as a "Related" file type.

3. Resubmission Checklist

When you are ready to resubmit your revised manuscript, please refer to this resubmission checklist: https://plos.io/Biology_Checklist

To submit a revised version of your manuscript, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' where you will find your submission record.

Please make sure to read the following important policies and guidelines while preparing your revision and fulfil the editorial requests:

a) *Published Peer Review*

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

b) *PLOS Data Policy*

Please note that as a condition of publication PLOS' data policy (http://journals.plos.org/plosbiology/s/data-availability) requires that you make available all data used to draw the conclusions arrived at in your manuscript. If you have not already done so, you must include any data used in your manuscript either in appropriate repositories, within the body of the manuscript, or as supporting information (N.B. this includes any numerical values that were used to generate graphs, histograms etc.). For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5

Please provide the data underlying the following figures, and make sure you mention in the corresponding figure legends WHERE THE DATA CAN BE FOUND. Please also ensure that your Data Statement in the submission system accurately describes where your data can be found.

c) *Blot and Gel Data Policy*

Please provide the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare them now, if you have not already uploaded them. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements

d) *Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosbiology/s/submission-guidelines#loc-materials-and-methods

Thank you again for your submission to our journal. We hope that our editorial process has been constructive thus far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Ines

--

Ines Alvarez-Garcia, PhD

Senior Editor

PLOS Biology

ialvarez-garcia@plos.org

----------------------------------------------

Reviewers’ comments

Rev. 1:

This manuscript by Wada et al. describes how FLCN promotes TFE3 recruitment to the lysosome by activating RagC. TFE3 belongs to the MiT-TFE family of transcription factors, which also includes MITF, TFEB, and TFEC. Previous studies have shown that these transcription factors are regulated in a similar manner, through a mechanism that involves mTORC1-mediated control of nucleo-cytoplasmic shuttling. More recent studies by several groups, including the group of authors of the present manuscript, have shown that mTORC1-mediated regulation of TFEB and TFE3 occurs through a "non-canonical" pathway that requires the FLCN-RagC/D axis. In the present manuscript the authors show that an mTORC1 substrate-specific mechanism, which was previously shown for TFEB (PMID: 32612235), also operates for TFE3. This observation is interesting and potentially relevant, considering the evidence that these two transcription factors may have different roles and relevance in both physiological and pathological settings. However, there are several issues, both in the text and figures, that need to be addressed by the authors.

CREDIT TO PREVIOUS STUDIES

The authors tend to ignore or minimize crucial contributions that other groups have previously made to the specific topic of the present manuscript. The following issues must be addressed by acknowledging previous discoveries and properly citing relevant previous papers.

1) A recent manuscript described a substrate-specific mechanism by which mTORC1 differentially phosphorylates its substrates and how this mechanism is relevant for Birt-Hogg-Dube' syndrome (PMID: 32612235). However, the results and conclusions of this previous manuscript have been largely ignored by the authors. Here are some examples: a) In the abstract (line 27) the authors stated "mTORC1 phosphorylates a myriad of substrates, but how different substrate specificity is conferred on mTORC1 by different conditions is largely unknown". This sentence does not take into account the mechanism recently described in ref. PMID: 32612235. b) Line 80. The hypothesis made by the authors at the end of the introduction is strikingly identical to the main conclusions of ref. PMID: 32612235. It looks like the authors ignored the important findings of this study by hypothesizing an identical mechanism… c) Line 291. The authors claim to have elucidated a new mechanism that enables substrate-specific phosphorylation by mTORC1. However, once again, they seem to ignore that this mechanism was previously described for TFEB (PMID: 32612235). d) Lines 306-314. The "BHD mTORC1 hyperactivity paradox" was recently explained in reference PMID: 32612235 . e) Line 344. The sentence "The work mechanistically exposes the first example of parsing of mTORC1 signaling" is incorrect, as it does not account for the mTORC1 substrate-specific mechanism described in ref. PMID: 32612235.

2) Line 66. Although the authors were the first to report the evidence of a "non-canonical" mTORC1 pathway that mediates TFE3 phosphorylation in adipose and myeloid tissue, additional studies reported differential phosphorylation of TFE3 versus canonical mTORC1 substrates in FLCN deficient cells (PMID: 21209915, PMID: 31672913).

3) Line 110. The first evidence that FLCN regulates the phosphorylation of TFE3 was described in ref. PMID: 21209915. This reference should be added.

4) Line 169. It has been well established by multiple studies that FLCN acts upstream of RagC (PMID: 24095279, PMID: 24081491, PMID: 31672913, PMID: 31704029). Such studies should be cited and the sentence "To test if RagC acts upstream or downstream of FLCN..." should be eliminated.

5) Line 177. The first evidence of an mTORC1-TFEB/TFE3-RagC/D feedback loop was reported in reference PMID: 28619945. Reference 7 only refers to the genes that are regulated by TFE3. Thus, reference PMID: 28619945 should be added when referring to the feedback loop (see also discussion Line 309).

TECHNICAL AND CONCEPTUAL ISSUES

6) Based on the data shown in Figure 2, the authors conclude that activation of RagA is required for the phosphorylation of S6K, but dispensable for the phosphorylation of TFE3. However, this conclusion cannot be drawn from the data shown in this manuscript for the following reasons: in Fig 2A the levels of TFE3 and S6K phosphorylation in starved cells expressing either active RagA or C are very weak. To claim that RagA activity is not relevant for TFE3 phosphorylation, the authors should compare the levels of TFE3 phosphorylation in cells expressing active RagC only, with those observed in cells expressing both active RagA and C. In Fig 2B a staining for RagA/C should be used to show which cells express active RagC/A. Furthermore, based on data in Fig 2B the authors claim that active RagC, but not active RagA, promotes cytosolic sequestration of TFE3 in the absence of AA. However, from these panels it appears to me that amino acid withdrawal induces TFE3 nuclear translocation in both active RagA- and active RagC-expressing cells. The authors should also add quantifications for these IF data. In Figure 2C loading controls are missing, making quantifications unreliable. RagA/C immuno-blotting data are also missing. My overall impression from Fig 2C is that both active RagA and active RagC show a very similar effect on TFE3 phosphorylation in starved cells. In addition to the aforementioned technical issues, the statement that activation of RagA is dispensable for TFE3 phosphorylation conflicts with the data in Figure 1A showing constitutive phosphorylation of TFE3 in cells lacking DEPDC5, a specific inhibitor of RagA/B.

7) Line 221. The authors state that "RagC is dispensable for AA signaling to S6K". However, Rag GTPases are known to work as dimers of either RagA or B bound to RagC or D. No published evidence suggests that RagA/B could work as monomers. The authors' conclusion that RagC is dispensable for S6K phosphorylation is based on the data in Fig 5a showing that phosphorylation of S6K is only marginally affected in RagC-KO cells. However, RagC and RagD are well known to be able to compensate for each other. Therefore, in Fig 5A the authors should assess the levels of RagD (as well as those of RagC), to determine whether a possible RagD compensation mechanism exists in RagC-KO cells. In addition, to claim that RagC is dispensable for S6K phosphorylation, the authors should assess the phosphorylation of S6K in a RagC/D-double KO cell line or in RagC-KO cells silenced for RagD.

8) Based on the data of Figure 6 the authors claim that anchoring TFE3 to different cytosolic compartments rescues its phosphorylation in FLCN-deficient cells. The mechanism by which such rescue occurs is unclear. In addition Figure 6 contains several important issues that need to be addressed (see below).Alternatively, the authors may consider eliminating this figure from the paper. Points to be addressed in Figure 6:

Fig6A: the levels of TFE3-Rheb15 in FLCN-KO cells are huge compared to the levels of either WT-TFE3 or TFE3-CAAX observed in the same cells (compare lane 4 with lanes 2 and 6). With such a huge difference in expression levels, it is impossible to come to any conclusion on the phosphorylation status of the different constructs in FLCN-KO cells.

Fig6E: Similarly, the levels of deltaNLS-TFE3 in FLCN-KO cells are massively higher than the levels of WT-TFE3 in the same cells (compare lane 6 with lane 2). Once again, it is impossible to make any comparison between these two samples.

Fig 6D: The authors need to compare on the same blot the phosphorylation of TFE3 fusion proteins in WT and FLCN-KO cells with the phosphorylation levels of WT TFE3 in the same cells. Are these fusion proteins efficiently phosphorylated compared to WT TFE3 in control cells? Is there any difference in the phosphorylation of WT vs chimeric TFE3 (expressed at similar levels) in FLCN-KO cells? Without this comparison it is impossible to assess if there is any "rescue" of TFE3 phosphorylation by linking it to specific cellular compartments in FLCN-KO cells.

Fig6B-C: the localization of TFE3-Rheb15 appears diffuse with no evident colocalization of TFE3 with either lysosomes, Golgi or ER. Similarly, I cannot appreciate any co-localization between TFE3-Tmem192 with LAMP2 (lysosomes), or between TFE3-Sec61b and PDI (ER). I suggest to perform higher quality IF analysis for co-localization of the chimeric protein with the specific cellular compartments and to perform, in addition, biochemical fractionation of the different organelles to confirm these data.

9) Related to point 7), the title of the paragraph "RagC, but not RagA, rescues TFE3 phosphorylation in the absence of FLCN" (lane 168) is misleading. The authors may want to change it with "Active RagC rescues TFE3 phosphorylation in the absence of FLCN, while active RagA does not".

10) The levels of total TFE3 in Fig 1A seem to vary considerably in the different treatments/cell lines. The authors should quantify their data and the levels of pTFE3 should be normalized with total TFE3.

11) Figure 3: In panel A the authors should clearly label WT and FLCN-KO cells; in panel B a staining for RagA/C should be used to show which cells express active RagC/A. Panel C: Due to the huge difference in the expression levels of TFE3 targets in WT and FLCN-KO cells, the graph in Fig 3C is hard to visualize. The transcript levels of TFE3 targets should be represented as fold change of mRNA levels in FLCN-KO cells VS NTC or the authors should separate the data of the two cell lines to allow a better comparison.

12) Figure legends lack the information regarding the statistics for each experiment and the number of times (n) experiments were performed. In addition, scale bars are missing in Fig 1, Fig 2, Fig 3, Fig 5 and Fig 6.

Rev. 2:

It was previously shown that mTORC1-dependent phosphorylation of the transcription factor TFE3 is lost in FLCN deficient cells. Here, the authors study the mechanism of TFE3 phosphorylation by mTORC1. In response to amino acids, FLCN activates RagC, which in turn recruits TFE3 to the lysosome. The authors show that in FLCN or RagC deficient cells, TFE3 is active in the nucleus. This phenotype is suppressed by overexpression of constitutively active RagC. Phosphorylation of the mTORC1 substrate S6K is unperturbed in FLCN or RagC deficient cells. Finally, the authors claim that by expelling TFE3 from the nucleus, by forcing localization to the lysosome, ER, Golgi or cytoplasm, TFE3 phosphorylation is restored even in the absence of FLCN. Specific comments are as follows.

1) The authors study TFE3 phosphorylation in cells cultured in different media: full medium, amino acid deficient, serum deficient and lacking both amino acids and serum. Based on this, the authors claim that growth factor signaling is dispensable for TFE3 phosphorylation (lines 105 -106 and 134), despite the fact that mTORC1 is normally growth factor dependent. Nevertheless, in Fig.1A TFE3 phosphorylation is reduced in serum starved cells compared to full medium. Also, surprisingly, in TSC2 deficient cells, TFE3 phosphorylation is reduced in all conditions. How do the authors explain these phenotypes? The authors should provide an explanation for the loss of TFE3 phosphorylation upon serum starvation (if mTORC1 is not the TFE3 kinase). They should also provide a more compelling explanation for why TFE3 phosphorylation is reduced in TSC2 deficient cells. In general, there are contradictory and confusing claims on the role of mTORC1 in TFE3 phosphorylation.

2) The experiments were performed exclusively in C2C12 cells. Do their findings apply to other cell lines?

3) Fig. 6. The authors force TFE3 localization to the cytoplasm, ER, Golgi or lysosome. Under these conditions, the authors claim that TFE3 is phosphorylated by mTORC1 even in the absence of the mTORC1 activator FLCN. How do the authors explain this observation?

4) Regarding the concluding statement in lines 341-342, the author claim to "elucidate the mechanistic basis by which FLCN confers substrate specificity upon the mTORC1." This statement is too strong. There are still open questions on the exact regulation (see point 1). Moreover, the authors fail to mention a publication from the Ballabio group (G. Napolitano et al, Nature, 2020) claiming a substrate-specific mechanism of TFEB phosphorylation by mTORC1. The authors should rephrase their concluding statement.

5) The authors claim that S6K phosphorylation is unperturbed by deletion of the FLCN gene. However, it has been shown that in FLCN deleted cells S6K phosphorylation is either lost or increased compared to control (e.g. Z. Tsun et al, Molecular Cell, 2013 and Y. Hasumi et al, Human Molecular Genetics, 2014). How can these different phenotypes be explained by the authors' findings? On a similar note, the authors claim that RagC deletion has no effect on S6K phosphorylation. However, the James group (e.g. G. Yang et al, EMBO, 2018) showed a reduction in S6K phosphorylation in RagC shRNA treated cells. The manuscript would benefit from inclusion of these two points the Discussion.

6) Related to statement of lines 196-197, the authors use Torin to increase intracellular amino acids level while blocking mTORC1 activity. The authors should explain how Torin, an mTORC1 and mTORC2 inhibitor, increases nutrients in cells.

7) Fig. 3C. Graphs in this image lack error bars.

8) Introduction. The Introduction would benefit from more citations. Lines 48-52 and 56-64 completely lack references where some should be provided.

9) Figures. To improve clarity, the authors should always add treatment information (e.g., drug concentration, time and media condition) in figure legends. Figures should be self-explanatory. For example, in Fig. 6 the authors should indicate which gene is KO'ed (FLCN). Moreover, it is difficult to distinguish colors in dot bar graphs. The authors should make graphs clearer. Finally, the authors should add axis labels to Fig. 1C and Fig. 2C.

10) The blots are generally over-exposed which might mask important differences. The total TFE3 antibody gives a different pattern in Fig 2A.

Decision Letter 2

Ines Alvarez-Garcia

1 Dec 2021

Dear Dr Arany,

Thank you for submitting a revised version of your manuscript "FLCN promotes substrate-selective mTORC1 activity by activating RagC to recruit TFE3" for consideration as a Research Article at PLOS Biology. This revised version of your manuscript has been evaluated by the PLOS Biology editors, the Academic Editor and the two original reviewers.

As you will see, both reviewers agree that most of the concerns raised in the previous round have been addressed, but they still mention several issues that need to be confirmed or clarified. One of the main points is the conclusion that S6K can be phosphorylated by mTORC1 in the absence of RagC/D, which is not convincingly demonstrated. Reviewer 1 thinks that you need to perform experiments to show if RagA/B can activate mTORC1 in the absence of RagC/D, and how RagA/B monomers activate mTORC1. The reviewer also suggests several other experiments to confirm your findings and the clarification of some points. Reviewer 2 raises similar issues and asks for several missing controls. After consulting with the Academic Editor and the rest of the team, we have decided to invite you to submit a new revision that should address all the remaining points highlighted by the reviewers. In addition, please remember to add references and make accurate attributions to previous work in the field.

In light of the reviews (attached below), we are pleased to offer you the opportunity to address the remaining points from the reviewers in a revised version that we anticipate should not take you very long. We will then assess your revised manuscript and your response to the reviewers' comments and we may consult the reviewers again.

We expect to receive your revised manuscript within 1 month.

Please email us (plosbiology@plos.org) if you have any questions or concerns, or would like to request an extension. At this stage, your manuscript remains formally under active consideration at our journal; please notify us by email if you do not intend to submit a revision so that we may end consideration of the manuscript at PLOS Biology.

**IMPORTANT - SUBMITTING YOUR REVISION**

Your revisions should address the specific points made by each reviewer. Please submit the following files along with your revised manuscript:

1. A 'Response to Reviewers' file - this should detail your responses to the editorial requests, present a point-by-point response to all of the reviewers' comments, and indicate the changes made to the manuscript.

*NOTE: In your point by point response to the reviewers, please provide the full context of each review. Do not selectively quote paragraphs or sentences to reply to. The entire set of reviewer comments should be present in full and each specific point should be responded to individually.

You should also cite any additional relevant literature that has been published since the original submission and mention any additional citations in your response.

2. In addition to a clean copy of the manuscript, please also upload a 'track-changes' version of your manuscript that specifies the edits made. This should be uploaded as a "Related" file type.

3. Resubmission Checklist

When you are ready to resubmit your revised manuscript, please refer to this resubmission checklist: https://plos.io/Biology_Checklist

To submit a revised version of your manuscript, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' where you will find your submission record.

Please make sure to read the following important policies and guidelines while preparing your revision and fulfil the editorial requests:

a) *PLOS Data Policy*

Please note that as a condition of publication PLOS' data policy (http://journals.plos.org/plosbiology/s/data-availability) requires that you make available all data used to draw the conclusions arrived at in your manuscript. If you have not already done so, you must include any data used in your manuscript either in appropriate repositories, within the body of the manuscript, or as supporting information (N.B. this includes any numerical values that were used to generate graphs, histograms etc.). Please also indicate in each figure legend where the data can be found. For an example see here: http://www.plosbiology.org/article/info%3Adoi%2F10.1371%2Fjournal.pbio.1001908#s5

b) *Published Peer Review*

Please note while forming your response, if your article is accepted, you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

c) *Blot and Gel Data Policy*

Please provide the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare them now, if you have not already uploaded them. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements

d) *Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosbiology/s/submission-guidelines#loc-materials-and-methods

Thank you again for your submission to our journal. We hope that our editorial process has been constructive thus far, and we welcome your feedback at any time. Please don't hesitate to contact us if you have any questions or comments.

Sincerely,

Ines

--

Ines Alvarez-Garcia, PhD

Senior Editor

PLOS Biology

ialvarez-garcia@plos.org

------------------------------------------------------------------

Reviewers' comments

Rev. 1:

The authors addressed most of my previous concerns. However, the following important issues remain unsolved:

1. INDEPENDENCE OF S6K PHOSPHORYLATION FROM THE PRESENCE OF RAGC/D

Based on the data shown in new Supplementary Figure 3, the authors claim that S6K can be phopshorylated by mTORC1 in the absence of RagC/D. However, as I had already pointed out in the first revision of this manuscript (point 7), a clear distinction should be made between the presence and the activity of Rag C/D GTPases. While the authors convincingly show that the activity of RagC/D is dispensable for the phosphorylation of S6K, the data shown in this manuscript are too weak to claim that S6K can be phosphorylated in the absence of RagC/D. Furthermore, there is a vast and robust literature showing that Rag GTPases can only work as heterodimers of RagA/B bound to RagC/D. No evidence so far has ever shown or suggested that RagA/B can work as monomers. The possible discovery that RagA/B can activate mTORC1 in the absence of RagC/D would be a revolutionary finding in the field and, therefore, would need to be unequivocally demonstrated with biochemical, structural, and functional analyses. If true, this would be the focus of an entirely different manuscript in which the authors would need to show how RagA/B monomers are able to activate mTORC1. In any case, I remain very skeptical about this possibility as I find the data shown in this manuscript too weak to make such a claim. Therefore, I suggest removing the data on RagC depletion and to keep the data obtained using active RagC, which explore the activity, rather than the presence, of RagC.

2. INDEPENDENCE OF TFE3 PHOSPHORYLATION FROM THE ACTIVITY OF RAGA/B

In response to my concerns in point 6 of the rebuttal, the authors state that they "do not conclude that activation of RagA is dispensable for the phosphorylation of TFE3" and that "RagA is not sufficient to confer activity on TFE3, but it may well be (and likely is) necessary". While I agree with these statements, the authors also state several times in the manuscript that the activity of RagC is sufficient to promote TFE3 phosphorylation. These two concepts contradict each other. The concept that RagC activation is sufficient to induce TFE3 phosphorylation of course implies that the activity of RagA is dispensable, which is in contrast with the authors' own data and statements. Therefore, the authors should fix these issues and re-phrase their claims about the role of the activity Rag GTPases, specifying that while the activity of RagA is essential for the phosphorylation of both TFE3 and S6K, the activity of RagC is important only for the phosphorylation of TFE3.

3. CYTOPLASMIC LOCALIZATION OF TFE3 IS SUFFICIENT TO RESCUE ITS PHOSPHORYLATION IN THE ABSENCE OF FLCN

The authors failed to address my previous concerns on Figure 6. The data contained in this Figure are too weak to support the authors' claim. Figure 6 contains several issues (e.g. a huge difference in the expression of the different proteins analysed, lack of proper localization of the chimeric proteins, inconsistency of data among different panels) and no conclusions can be drawn from these data. Importantly, no mechanism is provided to explain why TFE3 phosphorylation would be rescued in FLCN-KO by simply forcing its localization to different cytoplasmic compartments. This message is in sharp contrast with the overall message of the manuscript (i.e. inactivation of RagC drives TFE3 de-phosphorylation in FLCN-KO cells). Finally, forcing the localization of TFE3 in several cytoplasmic structures (such as Golgi, ER etc…) does not address the "recruitment of TFE3 to mTORC1", as stated in the title of this section. Thus, in my opinion this section, including Figure 6, should be removed from the manuscript.

Rev. 2:

The authors addressed all concerns raised previously. Specific comments are as follows:

1) Related to previous comment 1. Lines 136-139 (before 132-135) state "Together, these data demonstrate that the presence of AAs is necessary and sufficient to promote phosphorylation of TFE3 by mTORC1, and does so via GATOR1 and FLCN, while growth factors are largely dispensable, in sharp contrast to canonical phosphorylation of S6K." The authors should rephrase as this sentence is misleading. The authors should instead include part of the explanation provided to the reviewer, i.e., that the absence of TSC2 is not sufficient to promote TFE3 phosphorylation.

According to the Supplementary Figure 1A, in untreated cells TFE3 phosphorylation is reduced by over 60% in serum-starved conditions compared to full media conditions, indicating at least a partial regulation of TFE3 phosphorylation by growth factors. If the authors want to conclude that S6K phosphorylation dependents more on growth factors than TFE3 phosphorylation, a quantification of S6K phosphorylation of Figure 1A (similarly to Supplementary Figure 1A) should be provided.

2) Regarding comment 2. The authors should explain in the text why in amino acids-starved 293T cells the expression of RagA-66L is not sufficient to promote S6K phosphorylation as shown in Figure 2A for c2c12 cells. Moreover, the author should repeat the experiment in Supplementary figure 2A with equal expression of RagC-WT and RagC-75L. Finally, Figure 2A and Supplementary Figure 2A have different annotations (GDP/GTP vs. 66L/75L), the authors should correct it and keep the same nomenclature in all figures.

3) Supplementary Figure 3C and lines 232-234. The authors' conclusion is not supported by the data. The authors should provide quantification as it appears that phosphorylated S6K is increased upon RagC and RagC/D RNAi treatment. Also, the author should blot phospho and total TFE3 for the same condition (and quantify) as well as RagC and RagD to confirm proper knock down.

4) Regarding comment 5. It would be helpful if the authors included in the Discussion at least part of the explanation provided to the reviewer.

5) Regarding comment 8. The author should add references for lines 48-52 and 56-63.

6) Regarding comment 9. It would facilitate the readers' understanding if the authors indicate which gene is KO'ed (FLCN) in the following figures: 3A, S3A, S4A (both blots), S4B, S5A, S5B (all three blots), 6A, 6C and 6D

Decision Letter 3

Ines Alvarez-Garcia

8 Feb 2022

Dear Dr Arany,

Thank you for submitting the new revised version of your Research Article entitled "FLCN promotes substrate-selective mTORC1 activity by activating RagC to recruit TFE3" for publication in PLOS Biology. I have obtained advice from one of the original reviewers and have discussed these comments with the Academic Editor. 

Based on the review (attached below), we will probably accept this manuscript for publication, provided you satisfactorily address the two remaining points raised by this reviewer - mainly the removal of Figure 6 and further clarifications on the text. In addition, we have checked carefully the Introduction and we are still not satisfied with the accuracy of some of the statements mentioned regarding previous publications. We think that Reference 13 should be added in the context of line 66 - “We have recently identified a substrate-specific branch of mTORC1 signaling, providing the first example of specific regulation of different branches of mTORC1 signaling [6, 7], subsequently also reported by the Zoncu group [8]” - as the title of that reference states "A substrate-specific mTORC1 pathway". Please also rephrase the last sentence of the Introduction by deleting 'with our input' to avoid misunderstandings: "While the work that we report here was being finalized, the Ballabio group reported overlapping findings with TFEB, with our input [13]".

Please also make sure to address the data and other policy-related requests stated below.

As you address these items, please take this last chance to review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the cover letter that accompanies your revised manuscript.

We expect to receive your revised manuscript within two weeks.

To submit your revision, please go to https://www.editorialmanager.com/pbiology/ and log in as an Author. Click the link labelled 'Submissions Needing Revision' to find your submission record. Your revised submission must include the following:

-  a cover letter that should detail your responses to any editorial requests, if applicable, and whether changes have been made to the reference list

-  a Response to Reviewers file that provides a detailed response to the reviewers' comments (if applicable)

-  a track-changes file indicating any changes that you have made to the manuscript. 

NOTE: If Supporting Information files are included with your article, note that these are not copyedited and will be published as they are submitted. Please ensure that these files are legible and of high quality (at least 300 dpi) in an easily accessible file format. For this reason, please be aware that any references listed in an SI file will not be indexed. For more information, see our Supporting Information guidelines:

https://journals.plos.org/plosbiology/s/supporting-information  

*Published Peer Review History*

Please note that you may have the opportunity to make the peer review history publicly available. The record will include editor decision letters (with reviews) and your responses to reviewer comments. If eligible, we will contact you to opt in or out. Please see here for more details:

https://blogs.plos.org/plos/2019/05/plos-journals-now-open-for-published-peer-review/

*Early Version*

Please note that an uncorrected proof of your manuscript will be published online ahead of the final version, unless you opted out when submitting your manuscript. If, for any reason, you do not want an earlier version of your manuscript published online, uncheck the box. Should you, your institution's press office or the journal office choose to press release your paper, you will automatically be opted out of early publication. We ask that you notify us as soon as possible if you or your institution is planning to press release the article.

*Protocols deposition*

To enhance the reproducibility of your results, we recommend that if applicable you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Please do not hesitate to contact me should you have any questions.

Sincerely,

Ines

--

Ines Alvarez-Garcia, PhD,

Senior Editor,

PLOS Biology

ialvarez-garcia@plos.org

------------------------------------------------------------------------

DATA POLICY: IMPORTANT - PLEASE READ

You may be aware of the PLOS Data Policy, which requires that all data be made available without restriction: http://journals.plos.org/plosbiology/s/data-availability. For more information, please also see this editorial: http://dx.doi.org/10.1371/journal.pbio.1001797 

Note that we do not require all raw data. Rather, we ask that all individual quantitative observations that underlie the data summarized in the figures and results of your paper be made available in one of the following forms:

1) Supplementary files (e.g., excel). Please ensure that all data files are uploaded as 'Supporting Information' and are invariably referred to (in the manuscript, figure legends, and the Description field when uploading your files) using the following format verbatim: S1 Data, S2 Data, etc. Multiple panels of a single or even several figures can be included as multiple sheets in one excel file that is saved using exactly the following convention: S1_Data.xlsx (using an underscore).

2) Deposition in a publicly available repository. Please also provide the accession code or a reviewer link so that we may view your data before publication. 

Regardless of the method selected, please ensure that you provide the individual numerical values that underlie the summary data displayed in the following figure panels as they are essential for readers to assess your analysis and to reproduce it:

Fig. 1C; Fig. 2B, C; Fig. 3C; Fig. 4A, B; Fig. 5A-C; Fig. 6A-C; Fig. S1A B; Fig. S3B; Fig. S5A and Fig. S6

NOTE: the numerical data provided should include all replicates AND the way in which the plotted mean and errors were derived (it should not present only the mean/average values).

Please also ensure that figure legends in your manuscript include information on WHERE THE UNDERLYING DATA CAN BE FOUND, and ensure your supplemental data file/s has a legend.

Please ensure that your Data Statement in the submission system accurately describes where your data can be found.

-------------------------------------------------------------------------

BLOT AND GEL REPORTING REQUIREMENTS:

We require the original, uncropped and minimally adjusted images supporting all blot and gel results reported in an article's figures or Supporting Information files. We will require these files before a manuscript can be accepted so please prepare and upload them now. Please carefully read our guidelines for how to prepare and upload this data: https://journals.plos.org/plosbiology/s/figures#loc-blot-and-gel-reporting-requirements 

-------------------------------------------

Reviewers' comments

Rev. 1:

The authors have satisfactorily addressed some of my criticisms. However, the remaining points are still problematic and need to be properly addressed:

1) As previously stated by this reviewer, data in Figure 6 are very weak and not suitable for publication. The main problems of this figure are: huge difference in the expression of the different proteins analysed, lack of proper localization of the chimeric proteins and inconsistency of data among the different figure panels. Thus, in my opinion Figure 6 needs to be removed from the manuscript.

2) The authors did not properly addressed point 2 of my report: INDEPENDENCE OF TFE3 PHOSPHORYLATION FROM THE ACTIVITY OF RAGA/B. While the authors state that they agree with this reviewer and that RagC is NOT sufficient for TFE3 phosphorylation, as RagA activity is in fact necessary for this process, they now state that "RagC is sufficient to promote TFE3 phosphorylation without simultaneous additional activation of RagA/B". This would imply that Rag GTPases can recruit and phosphorylate TFE3 even when RagA/B are inactive. This is wrong, as it is well established that activation of RagA/B is required for mTORC1 lysosomal recruitment and activation. The text should be changed accordingly.

Decision Letter 4

Ines Alvarez-Garcia

7 Mar 2022

Dear Dr Arany,

On behalf of my colleagues and the Academic Editor, Anne Simonsen, I am pleased to say that we can in principle accept your Research Article entitled "Folliculin promotes substrate-selective mTORC1 activity by activating RagC to recruit TFE3" for publication in PLOS Biology, provided you address any remaining formatting and reporting issues. These will be detailed in an email that will follow this letter and that you will usually receive within 2-3 business days, during which time no action is required from you. Please note that we will not be able to formally accept your manuscript and schedule it for publication until you have any requested changes.

Please take a minute to log into Editorial Manager at http://www.editorialmanager.com/pbiology/, click the "Update My Information" link at the top of the page, and update your user information to ensure an efficient production process.

PRESS

We frequently collaborate with press offices. If your institution or institutions have a press office, please notify them about your upcoming paper at this point, to enable them to help maximise its impact. If the press office is planning to promote your findings, we would be grateful if they could coordinate with biologypress@plos.org. If you have previously opted in to the early version process, we ask that you notify us immediately of any press plans so that we may opt out on your behalf.

We also ask that you take this opportunity to read our Embargo Policy regarding the discussion, promotion and media coverage of work that is yet to be published by PLOS. As your manuscript is not yet published, it is bound by the conditions of our Embargo Policy. Please be aware that this policy is in place both to ensure that any press coverage of your article is fully substantiated and to provide a direct link between such coverage and the published work. For full details of our Embargo Policy, please visit http://www.plos.org/about/media-inquiries/embargo-policy/.

Thank you again for choosing PLOS Biology for publication and supporting Open Access publishing. We look forward to publishing your study. 

Sincerely, 

Ines

--

Ines Alvarez-Garcia, PhD 

Senior Editor 

PLOS Biology

ialvarez-garcia@plos.org

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. TFE3 phosphorylation is responsive to AAs, via the GATOR complex.

    (A) Control C2C12 cells (NTC) or cells lacking Flcn, Tsc2, or Depdc5 were switched from complete medium to media lacking serum and/or AAs for 60 minutes followed by immunoblotting. Images were uploaded into ImageJ, and signal intensity was quantified. Graphed above is the ratio of p-TFE3 to total TFE3 signal. (B) Quantification and graph of ratio of pS6K to total S6K signal. The data underlying all the graphs shown in the figure is included in S1 Data. AA, amino acid; FLCN, folliculin.

    (PDF)

    S2 Fig. RagC, but not RagA, promotes TFE3 phosphorylation in response to AAs.

    (A) Coexpression of active RagA and RagC in 293T cells does not confer further phosphorylation of TFE3 compared to active RagC alone. (B) RagC 75L protein is partially stabilized by inhibition of the proteasome with MG132. (C) To accompany main Fig 2C, immunoblotting for total levels of TFE3, S6K1, and 4E-BP in C2C12 cells expressing HA-tagged WT, or constitutive active RagA (GTP) or RagC (GDP), demonstrate equivalent expression of these proteins at all time points after switching from complete medium to media lacking AAs. AA, amino acid; WT, wild type.

    (PDF)

    S3 Fig. RagD expression is not increased as compensation for RagC CRISPR KO or siRNA knockdown.

    (A) C2C12 cells with RagC CRISPR KO were used for Fig 5A. (B) RagC CRISPR KO (in C2C12s) and RagC siRNA knockdown (in HEK 293Ts) showed no significant compensation of RagD expression. The data underlying all the graphs shown in the figure is included in S1 Data. KO, knockout.

    (PDF)

    S1 Data. Data underlying graphs in main and Supporting information figures.

    (XLSX)

    S1 Raw Images. Raw images.

    (PDF)

    Attachment

    Submitted filename: response to reviewers.docx

    Attachment

    Submitted filename: response to reviewers.docx

    Attachment

    Submitted filename: Response to reviewers.docx

    Data Availability Statement

    All relevant data are within the paper and its Supporting Information files.


    Articles from PLoS Biology are provided here courtesy of PLOS

    RESOURCES