SUMMARY
The mechanistic target of rapamycin complex-1 (mTORC1) coordinates regulation of growth, metabolism, protein synthesis, and autophagy1. Its hyper-activation contributes to disease in many organs including the heart1,2, though broad mTORC1 inhibition risks interference with its homeostatic roles. Tuberin (TSC2) is a GTPase-activating protein and prominent intrinsic regulator of mTORC1 by modulating Rheb (Ras homolog enriched in brain). TSC2 constitutively inhibits mTORC1, but this activity is modified by phosphorylation from multiple signaling kinases to in turn inhibit (AMPK, GSK3β) or stimulate (Akt, ERK, RSK-1) mTORC1 activity3–9. Each kinase requires engagement of multiple serines, impeding analysis of their role in vivo. Here, we reveal phosphorylation or gain-or-loss of function mutations at either of two adjacent serines in TSC2 (S1365/1366 mouse; 1364/1365 human), with no prior known function, is sufficient to bi-directionally potently control growth-factor and hemodynamic-stress stimulated mTORC1 activity and consequent cell growth and autophagy. Basal mTORC1 activity, however, is unchanged. In heart, myocytes, and fibroblasts, phosphorylation occurs by protein kinase G (PKG), a primary effector of nitric oxide and natriuretic peptide signaling whose activation is protective against heart disease10–13. PKG suppression of hypertrophy and stimulation of autophagy in myocytes requires TSC2 phosphorylation. Homozygous knock-in (KI) mice expressing a phospho-silenced TSC2 (S1365A) mutation develop far worse heart disease and mortality from sustained pressure-overload (PO) due to hyperactive mTORC1 that cannot be rescued by PKG stimulation. Heterozygote SA-KI are rescued, and KI-mice expressing a phospho-mimetic (S1365E) mutation are protected. Neither KI model alters resting mTORC1 activity. Thus, TSC2 phosphorylation is both required and sufficient for PKG-mediated cardiac protection against pressure-overload. These newly identified serines provide a genetic tool to bi-directionally regulate the amplitude of stress-stimulated mTORC1 activity.
Hearts subjected to sustained PO develop pathological growth and reduced function (Extended Data 1a), accompanied by mTORC1 activation shown by increased phosphorylation of p70S6K and 4EBP1 (elF4E binding protein-1) stimulating gene transcription/translation, and Ulk-1 (Unc-51-like kinase-1) reducing autophagy14 (Figure 1a, Extended Data 1b). PKG activation by orally administered sildenafil (phosphodiesterase-type-5 inhibitor) suppresses these changes, also increasing LC3-II (microtubule-associated protein light-chain 3-II) while reducing p62 protein expression (Figure 1b, Extended Data 1c) and myocardial protein aggregation (Extended Data 1d), consistent with enhanced autophagy. These effects are mirrored by everolimus (Evl), a relatively selective mTORC1 inhibitor. In isolated cardiac myocytes stimulated by endothelin-1 (ET1), cGMP activation of PKG increases autophagic-flux, demonstrated by increased red-puncta (auto-lysosomes) in cells expressing a fluorescent reporter (TF-LC3)15 (Figure 1c), and by more enhanced LC3-II expression in bafilomycin-A1 treated cells (Extended Data 2a). PKG anti-hypertrophic effects are blunted by gene silencing of autophagy related 5 (Atg5) (Extended Data 2b). Thus, PKG activation suppresses cardiac mTORC1 signaling, blunting growth and enhancing autophagy.
To determine the mechanism for mTORC1 suppression by PKG, adult rat myocytes were exposed to cGMP for 15 min, and lysates analyzed by phospho-proteomics. Among mTORC1-complex and regulating proteins, mass-spectroscopy identified two adjacent serines (hsS1364/65-human; mmS1365/66-mouse) in a highly conserved activation domain of TSC2, upstream of GSK-3β and AMPK phospho-sites (Figure 1d, Extended data 2c). PKG is among the top three kinases predicted to modify hsS1364 (PhosphoNET, Kinexus; link), though it may also modify hsS1365. Databases report phosphorylation of hsS136416,17. There are no reported human mutations in the hsS1364, but are two children with an hsS1365L mutation, each with seizures but no tumors.
A commercial antibody for mouse phospho-mmS1365 was manufactured, but had no prior validation. Mouse embryonic fibroblasts (MEFs) show low-levels of basal signal that increases with cGMP stimulation and is blocked by PKG inhibition (DT3) (Figure 1e upper, Extended Data 2d). Antibody signal is present in myocardium in vivo, increasing with PO and further with PO+Sil but not PO+Evl co-treatment (Figure 1f). This correlates with PKG activity (Figure 1g). Analogous phosphorylation at hsS1364 is found in non-failing human myocardium that increases in dilated heart failure (Figure 1h). To test antibody specificity, myocytes were transfected with hsTSC2-WT, hsTSC2-HA-S1364A, or hsTSC2-HA-S1364E at matched expression levels (Extended data 2e), and cells stimulated with endothelin-1 (ET1) to activate PKG (Extended data 3a, 3b) and induce TSC2-phosphorylation. Antibody signal increased only if WT TSC2 was expressed (Figure 1i, Extended data 3c). Identical results were obtained if hsTSC2-S1365 was mutated (Extended data 3d), so interference with either serine prevents PKG phosphorylation.
PKG directly phosphorylates TSC2 as demonstrated by radiolabeling of TSC2 upon co-incubation of recombinant PKG1α (primary cardiovascular isoform) with TSC2-HA, and [γ−32P]-ATP (Figure 1j). This is also found when hsTSC2-HA-S1364A (Figure 1j) or hsTSC2-FLAG-S1365A is used (Extended data 3e) indicating additional sites exist. We also tested if PKG1α directly modifies TSC2 in the presence of whole cell proteins using mutated PKG1α18 (M438G) that uniquely binds modified/enlarged sulfonated-ATP19. This assay revealed TSC2-thiphosphate ester (PKG phosphorylation) in WT and both SA mutations (Extended Data 3f) as well.
We next tested the functional significance of the serines. Figure 2a and 2b show results from myocytes transfected with WT, hsTSC2-HA-S1364A, or hs-TSC2-HA-S1364E and then further stimulated with 48-hr ET1. Basal Nppb mRNA (a hypertrophy biomarker) was low, regardless of which TSC2 was over-expressed. ET1 increased Nppb most in hsS1364A-TSC2 and least in hsS1364E-TSC2 expressing cells, each compared to WT (Figure 2a). Nppb expression declined with PKG only in TSC2-WT-expressing cells, consistent with the lack of hsS1364 phosphorylation with either site mutated (Figure 1i). Very similar results were obtained with hsS1365A and hsS1365E mutations (Extended Data 4a), so mutations of either serine alone are sufficient to induce a similar biological response. Compared to WT, SA-TSC2 amplified whereas SE-TSC2 attenuated mTORC1 activity stimulated by ET1, neither having an impact at rest. LC3-II protein increased and p62 declined with SE expression supporting enhanced autophagy; the opposite occurring with SA expression. This was observed regardless of which serine was mutated (hsS1364: Figure 2b, Extended Data 4b; hsS1365: Extended Data 4c, 4d). Increased TF-LC3 red-punctae support enhanced autophagic flux with SE expression (Figure 2c). Phenylephrine mimicked ET1 effects (Extended Data 5a), and hsS1364A-TSC2 expression depressed PKG-modulation of LC3-II and p62 (Extended Data 5b). Thus, site-substitution mutants at either of the two adjacent serines on TSC2 either amplify or attenuate stress-stimulated mTORC1 regulation, and PKG effects on growth and autophagy are either markedly impaired (SA) or mimicked (SE) by single point mutations of either one.
TSC2 principally modulates mTORC1 by regulating Rheb-GTP binding, and Rheb gene silencing blocked ET1-stimulated mTORC1 activation in WT- and SA-TSC2 expressing myocytes. It had no effect in SE-TSC2 expressing cells that exhibited low mTORC1 activity regardless (Fig. 2d, Extended Data 5c). The serines are proximate to AMPK and GSK3β targeted residues, the latter requiring AMPK co-activation of TSC29. To test if an SA mutation prevents TSC2-mTORC1 control via AMPK, TSC2-KO MEFs expressing WT, SA, or empty vector were exposed to ET1 and then 2-deoxyglucose (2-DG), the latter inhibiting glycolysis to stimulate AMPK. KO MEFs displayed constitutive mTORC1 activation fairly insensitive to 2-DG. WT- or SA-TSC2 expression reduced this activity, but it was further depressed similarly by 2-DG in concordance with hsTSC2-S1387 phosphorylation (AMPK site) (Figure 2e-2h).
Mice with a global mmTSC2-S1365A (SA) knock-in mutation were generated using CRISPR/Cas9 gene editing (Figure 3a and Extended Data 6a) to assess its role in vivo. They are born in normal Mendelian ratios, grow and develop normally, have similar cardiac structure and function as their littermate controls (Extended Data Table), and levels of myocardial TSC2 protein (Extended Data 6b). Heterozygote (SA/WT), homozygote (SA/SA) and control (WT) mice were subjected to PO, and randomized to vehicle or sildenafil co-treatment. Mortality after PO in vehicle-treated mice was markedly higher in SA/WT and SA/SA versus WT, and sildenafil prevented this in SA/WT but not SA/SA mice (Figure 3b). PO-stimulated heart hypertrophy, dysfunction, and greater lung weight was similar in SA-expressers versus WT, but sildenafil only reversed this in SA/WT and WT (Figure 3c-3d, Extended Data 6c). MTORC1 activity and myocardial protein aggregation were greater and autophagy (p62, LC3-II) depressed following PO in SA-expressers versus WT, and was reversed by sildenafil in all but SA/SA hearts (Figure 3g, 3h, Extended Data 6d, 6e). These findings were similar in males and females. MTOR-complex-2 activation was unaltered by the SA mutation (Extended Data 7a). SA/SA mice randomized to everolimus (vs. vehicle, each starting 3 days before PO) showed full rescue of mortality (Figure 4a) and heart disease (Figure 4b, 4c), with enhanced autophagy (Extended Data 7b), supporting a key role of mTORC1 hyperactivation from SA-expression. Together, they show the mmS1365A mutation acts as an autosomal dominant, amplifying stress-stimulated but not rest mTORC1 activity in vivo, and that pS1365 is required for PKG-mediated mTORC1 suppression and amelioration of PO-induced heart disease.
KI mice expressing mmTSC2-S1365E were also generated (Extended Data 8a) to test if a phosphomimetic mutation confers effects opposite to SA. The mice are born with normal Mendelian ratios, cardiac morphology, and function (Extended Data Table). Opposite to SA, both SE/WT and SE/SE are markedly protected against PO, with minimal hypertrophy and better heart function (Figure 4d, 4e). Baseline mTORC1 activity with SE expression is similar to WT but remains low after PO, while LC3-II increases and p62 declines (Figure 4f, Extended Data 8b). In vivo autophagic flux is somewhat higher in SE and less in SA vs WT (Extended Data 8c). SE-TSC2 expressing mice also have reduced myocardial protein aggregation (Figure 4g).
Both SA and SE TSC2 modifications impact stress-induced but not basal mTORC1 activity or in vivo cardiac physiology. This distinguishes them from myocyte-targeted TSC2-deletion that constitutively activates mTORC1 inducing heart disease20, and genetic mTOR or mTORC1 suppression that induces heart failure and early lethality21,22. Lack of a basal phenotype in both KI models is notable, as these mutations exist in all cells, highlighting an advantage of modulating mTORC1 via TSC2 rather than targeting the mTORC1 complex itself. Prior in vitro studies with TSC2 mutants that silenced (S→A substitution) kinase-targeted phospho-sites for AMPK8, ERK1/24, Akt5, and GSK3β9 each found they altered basal mTORC1 activity. Phospho-mimetic simulation of ERK1/2 phosphorylation stimulated mTORC1 activity and cell growth4. This too differs from hsTSC2-S1364 (or S1365) modulation, that acts more as an amplifier/attenuator of mTORC1 co-stimuli. Moreover, it affords selective control, potently regulating growth factors (or PO, a multi-factorial stress), but not metabolic signaling via AMPK. Similar selectivity may apply to other TSC2 kinase regulators, but this remains unknown. The present KI mice are the first in vivo models altering TSC2 phosphorylation, and whether multi-site silencing/mimetic mutants of other TSC2 regulating kinases yield similarly benign rest phenotypes remains to be determined.
Both SA- and SE-TSC2 modify mTORC1-regulated growth and autophagy, revealed by multiple assays with mutations at either of the two adjacent serines, and in the SA-PO everolimus-rescue study. While PKG modulates pathological growth by other pathways as well11, we believe this is the first study coupling it to autophagy. In SA/SA mice subjected to PO, sildenafil failed to alter LC3-II and p62 expression or protein aggregation, supporting a key role of PKG-TSC2 modulation at mmS1365. Whether genetically preventing autophagy abrogates PKG-amelioration of heart disease in vivo, and if so requires TSC2-hsS1364/S1365, remains to be tested.
Our results identify PKG as a direct mediator of TSC2-hsS1364/1365 post-translational regulation. That PKG does not phosphorylate TSC2 regardless of which is mutated, and that cell behavior from SA or SE mutations in each serine are very similar supports tight inter-regulation. While additional PKG sites on TSC2 are suggested by our studies, the data indicates either of the identified sites is sufficient. Other kinases may also target these residues, as mmS1365 phosphorylation was first reported in a proteomic study of TSC2-suppressors (ERK, p90S6K), and modified by phorbol ester, a non-specific protein kinase C activator16. The study did not perform functional analysis, and importantly, phorbol ester and more specific PKC activators activate mTORC123,24, opposite to PKG. The findings that pathological stress stimulates TSC2 phosphorylation in myocytes and myocardium along with mTORC1 activation, but its prevention by TSC2-SA expression amplifies the pathology, supports a role of this phosphorylation site on negative feedback to mTORC1. Protective effects from TSC2-SE expression or more selective phosphorylation by PKG supports this further. Mammalian cells broadly express TSC2, mTOR, and related mTOR-complex proteins, and many also express PKG. Thus, our data have implications beyond the heart, expanding potential therapeutic roles of PKG activators for disease where altered modulation of mTORC1 is present and/or desired. Furthermore, genetic modulation of the newly identified phospho-sites provides an mTORC1 controller with potential utility for adoptive cell therapies by providing more nuanced and flexible mTORC1 regulation.
METHODS
Animal models
Mice expressing global TSC2 knock-in mutations mmTSC2-S1365A (SA) or mmTSC2-S1365E (SE) (Figure 3A, Extended Data 6a, Extended Data 8a) were newly generated using CRISPR/Cas9 gene editing (Transgenic Core Laboratory, Johns Hopkins University). TSC2 guide RNA was designed using an algorithm developed by Vinod Ranganathan25, and cloned into pSpCas9(BB)-2A-Puro (PX459) V2.0 (a gift from Feng Zhang, Addgene plasmid #62988), and pUC57-sgRNA expression vector (a gift from Xingxu Huang, Addgene plasmid #51132). DNA cleavage was tested in mouse N2a cells using the Surveyor Mutation Detection kit (Integrated DNA Technologies, Coralville, IA) according to the protocol of Ran et al26. In vitro transcription was performed for both Cas9 (from a modified pX330-U6-Chimeric_BB-CBh-hSpCas9 plasmid, originally a gift from Feng Zhang, Addgene plasmid #42230), and guide RNA using the Ambion mMESSAGE mMACHINE kit and NEB HiScribe T7 High Yield RNA Synthesis kit, respectively. ssODN for the S1365A or S1365E point mutations were purchased from Integrated DNA Technologies. C57Bl/6J blastocyst injections were performed by the Johns Hopkins Transgenic Mouse Core Laboratory with a mix consisting of: 25ng/μl Cas9; 12.5ng/μl guide RNA, and 25ng/μl ssODN. We received 5 founders positive for the TSC2 SE transgene and 3 founders positive for the TSC2 SA transgene. Both colonies of mice were backcrossed with mice on a C57Bl/6J background, generating mice with a 98.5% (SE) and 99.3% (SA) purity. The genetic backgrounds of mice were assessed at DartMouse™ Speed Congenic Core Facility at the Geisel School of Medicine at Dartmouth. DartMouse uses the Illumina, Inc. (San Diego, CA) Infinium Genotyping Assay to interrogate a custom panel of 5307 SNPs spread throughout the genome. The raw SNP data were analyzed using DartMouse’s SNaP-MaP™ and Map-Synth™ software, allowing the determination for each mouse of the genetic background at each SNP location. We maintained each colony with the breeding strategy of a heterozygous male and a heterozygous female, yielding KI homozygous mice, KI heterozygous mice, and littermate WT mice at a 25:50:25 ratio.
Pressure overload (PO) model
PO was induced by trans-aortic constriction as previously described10. Sham controls underwent similar surgery without ligature constriction. Age and weight-matched littermates were randomly divided into PO or sham groups with male and female mice equally represented in the data presented. Mice were followed for up to 6 weeks after PO, in some instances, co-treated with everolimus (Evl, Sigma; oral gavage, 10 mg/kg/day), or sildenafil (Sil, Pfizer or Wako Pure Chemical Industries, 200 mg/kg/day mixed in soft diet, Bioserv)27, or appropriate matched vehicle. Drug treatment started either 1-week following PO, or several days prior to PO. All of the protocols were approved by the Johns Hopkins Medical Institutions Animal Care and Use Committee. The studies were in compliance with all ethical regulations.
Conscious mouse echocardiography
Intact heart morphology and function were measured in conscious mice by serial M-mode transthoracic echocardiography (VisualSonics Vevo 2100, 18–38 MHz transducer; SanoSite Inc.). Images were obtained and analyzed using VisualSonics image software, by an individual blinded to the animal condition.
TSC2 expression plasmids and adenovirus
A FLAG-tagged human TSC2 expression plasmid was provided by Brendan Manning (Harvard University). Based on this, we further generated 5 new constructs, one in which FLAG was replaced by an HA sequence (both at the C-terminus), and then each WT vector further modified to generate hsTSC2-HA-S1364A, hsTSC2-HA-S1364E, hsTSC2-FLAG-S1365A, and hsTSC2-FLAG-S1365E mutants. Adenovirus was also developed expressing the FLAG-tagged WT, hsS1365A, or hsS1365E (Welgen, Inc., Worcester, MA) and used for studies involving TSC2-KO and TSC2-WT mouse embryonic fibroblasts MEFs. These cells lines were provided by Dr. Brendan Manning as first generated by David Kwiatkowski28.
Neonatal rat cardiomyocyte studies (NRCMs)
NRCMs were isolated and cultured at 1M cells/well in 6-well plates for 24 hours in DMEM with 10% FBS and antibiotics prior to study, described10. Hypertrophy was stimulated with endothelin 1 (ET1, 10 nM, Sigma), phenylephrine (PE, 100 μM, Sigma) for either 15 min or 48 hours in serum-free DMEM supplemented with 0.1% Insulin-Transferrin-Selenium (Life Technologies). Studies were also performed in NRCMs first transfected with plasmids expressing TSC2-WT, SA, or SE mutations (hsS1364 or hsS1365) (5 μg/well), using Takara Clontech Xfect reagent per manufacturer’s protocol. Cells were provided 24 hours prior to exposure to stimulation with ET1 or vehicle in combination with other reagents as described in the study.
Mouse Embryonic Fibroblasts (MEF) and HEK TSC2-KO cells
MEFs were cultured in DMEM supplemented with 10% FBS and 1% antibiotics until reaching 50–60% confluence at which time the cells were transfected with TSC2 constructs or empty vector, or infected with adenovirus expressing WT-, SA- or SE-TSC2. Expression vectors used a CMV promoter. Following an additional 24 hours of culturing, transfected MEFs were treated with various reagents: 8-Bromo-cGMP (100 μM), DT3 (1 μM), 2-deoxyglucose (100 μM), and vehicle control.
Human myocardium analysis
Human myocardium was obtained in accordance with institutional review board approvals at Johns Hopkins University and the University of Pennsylvania. The use of human subject material was in compliance with all ethical regulations. Failing human hearts were obtained at time of explant surgery, and non-failing controls at time of other organ harvesting. LV free wall tissue was collected at the University of Pennsylvania under ice-cold cardioplegia and rapidly frozen in liquid nitrogen. Informed consent was obtained from failing heart human tissue donors. The family or legal representative provided consent for organ harvesting from deceased donor controls. For the non-failing controls, the age was 52.8 ±15.4, 6 males, 6 females, no clinical history of heart failure; for the heart failure group, the mean age was 51.3 ±12.1, 8 males, 4 females (p=NS for sex distribution between groups), and all were non-ischemic dilated cardiomyopathy patients with severe LV dysfunction who then underwent cardiac transplantation.
Immunoblot analysis
Whole cell lysate was extracted (Cell Signaling Technology #9803) and protein concentration determined by BCA method (Pierce). Samples were prepared in SDS Tris-Glycine buffer (Life Technologies) and run on Novex 8–16% Tris-Glycine Gels (Life Technologies) or TGX 7.5% and 4–20% Tris-Glycine Gels (Bio-Rad) and blotted onto a nitrocellulose membrane. The following primary antibodies were used in this study: phospho-Akt (S473) #9271 lot 14 used at 1:1,000, Akt #9272 lot 28 used at 1:1,000, phospho-70 S6K (T389) #9205 lot 21 used at 1:1,000, p70 S6K #9202 lot 20 used at 1:1,000, phospho-4EBP1 (S65) #9451 lot 14 used at 1:1,000, 4EBP1 #9452 lot 12 used at 1:1,000, phospho-Ulk-1 (S757) #1420 clone D706U lot 4 used at 1:1,000, Ulk-1 #8054 clone D8H5 lot 5 used at 1:1,000, phospho-FoxO1/3 #9464 lot 7 used at 1:2,500, FoxO1 #2880 clone C29H4 lot 11 used at 1:3,000, phospho-NRDG-1 #3217 clone D98G11 lot 3 used at 1:5,000, NRDG-1 #9395 clone D6C2 lot 1 used at 1:1,000, GAPDH #2118 clone 14C10 lot 10 used at 1:1,000, Rheb #13879 clone E1G1R lot 1 used at 1:1,000, phospho-TSC2 (S1387) #5584 lot 5 used at 1:1,000, TSC2 #3612 clone D93F12 lot 5 used at 1:1,000, and α-tubulin #3873 clone DM1A lot 12 used at 1:1,000 (Cell Signaling Technology), phospho-TSC2 (S1365) #120718 lot NFSA12072OAH used at 1:500 (NovoPro Labs), LC3 #ab192890 lot GR321–3 used at 1:1,000, thiophosphate ester #ab92570 lot GR237393–18 used at 1:5,000, and p62 #ab109012 lot GR12843–70 used at 1:1,000 (Abcam), FLAG #F3165 lot SLBK1346V used at 1:1,000, ubiquitin #SAB4503053 lot 310385 used at 1:1,000 (Sigma), and a total protein stain #926–11016 lot C80522–02 used at 5 ml/membrane (Li-Cor). Antibody binding was visualized by infrared imaging (Odyssey, Licor) and quantified with Licor Image Studio Software 3.1.
Gene expression by quantitative RT-PCR
Total RNA isolated from left ventricular myocardium or cultured NRCMs (Trizol Reagent, Invitrogen), was reverse transcribed to cDNA (High Capacity RNA-to-cDNA Kit, Applied Biosystems, Life Technologies), and underwent PCR amplification using TaqMan probes for atrial natriuretic peptide (Nppa) (mouse #Mm01255747_g1, rat #Rn00664637_g1), B-type natriuretic peptide (Nppb) (mouse # Mm01255770_g1, rat #Rn00580641_m1), regulator of calcineurin-1 (Rcan-1) (mouse #Mm01213406_m1, rat #01458494_m1), tuberous sclerosis complex 2 (tuberin, TSC2) (mouse #Mm00442004_m1, rat #Rn00562086_m1), or glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (mouse #99999915_g1, rat #Rn01775763_g1) (Applied Biosystems). The threshold cycle value was determined using the crossing point method, and samples normalized to the GAPDH for each run.
Bafilomycin A1 autophagy-flux assay
For in vitro studies, NRCMs were cultured as described above, stimulated with cGMP (50 μM) for 15 minutes, and then with either vehicle or bafilomycin A1 (BFA, 1 μM) (Sigma) for 3 hours. For in vivo studies, WT, SA/SA, and SE/SE mice received two injections of BFA (3 μM/kg, IP) 90 minutes apart. Myocardium was collected 90 minutes after the second injection, and protein extract examined by immunoblot for LC3-II. The relative increase in expression with versus without BFA indexes autophagic flux.
Protein Aggregation Assays
Protein aggregation was measured using Proteostat (Enzo #ENZ-51023) following manufacturer’s instructions. Ventricular myocardial lysate (Cell Signaling lysis buffer) was obtained, protein concentration assayed, and 10 μg protein loaded into a 96 well microplate and protein aggregates analyzed using the Proteostat® assay kit (Enzo Life Sciences) following manufacturer’s instructions. After background subtraction, values were normalized to WT sham.
For a second assay, 1 mmol/L phenylmethanesulfonyl fluoride and a phosSTOP™ tablet (Sigma-Aldrich/Roche) were added to myocardial lysates. This was centrifuged at 8,000g in 4°C for 10 minutes, supernatant (soluble fraction) extracted, and the pellet then resuspended in insoluble extraction buffer (40 mmol/L Tris HCl [pH 8.8], 1% SDS, 8% glycerol), boiled for 5 minutes, and re-centrifuged at 3,000g for 5 minutes. 2.5 μg of the supernatant (insoluble fraction) was filtered through nitrocellulose membrane (pore diameter, 0.22 μm, Millipore) using a dot-blot apparatus (BioRad), and immunoprobed for ubiquitin and α-tubulin.
Tandem fluorescent LC3 autophagic flux assay
NRCMs were infected with an adenovirus (10 MOI) expressing a tandem fluorescent (GFP-RFP) tagged LC315. This expresses LC3 with both green and red fluorescence as the autophagosomal membrane is forming; but upon merging with the acidic lysosome (autolysosome), the GFP signal is quenched, leaving RFP. The rise in RFP provides a marker of autophagic flux. In some studies, myocytes were further transfected with plasmid encoding for WT-TSC2, or SA or SE mutant TSC2, and further stimulated for 48 hours with endothelin 1 (10 nM). Dot counts for both colors/cell were determined using Image J software (Ver 1.52a, NIH).
In vitro protein kinase G activity
PKG activity was assessed by in vitro colorimetric assay (Cyclex, Cat #CY-1161, Nagano, Japan) following the manufacturer’s instructions. The assay provides cGMP substrate, and a kinase-specific peptide-target to assess phosphorylation activity.
Proteomic analysis of PKG phospho-kinome
Freshly isolated adult cardiac myocytes were obtained from male Wistar rats as described29, and divided into two aliquots, each relaxed in Tyrode buffer (140 mM NaCl, 5 mM KCL, 10 mM HEPES, 1 mM glucose, 1 mM MgCl2, 1 mM Ca2+, pH 7.45). Cells were then exposed to 1 mM 8-Br-cGMP or Tyrode solution for 10 minutes to stimulate intracellular PKG activity. Cells were then centrifuged for 1 minute at 1000g, the supernatant removed, and the pellet frozen in liquid nitrogen and stored at −80°C. Frozen samples (n=3/group) were then lysed in an 8M Urea, 0.5% SDS solution with brief sonication, and protein concentration determined by the BCA method. For each sample, 200 μg of total protein was digested with trypsin/Lys-C protease mixture (Promega), samples were desalted on 10 mg Oasis HLB cartridges (Waters) and eluted in 300 μL of 80% acetonitrile (ACN), 5% trifluoroacetic acid, 1 M glycolic acid and enriched by titanium dioxide (TiO2). Enriched peptides were desalted as above but eluted in 200 μL of 80% ACN, 0.1% formic acid (FA) and dried under vacuum. Dried peptides were re-suspended in 20 μL of 0.1% FA for LC-MS/MS analysis. Samples (4 μL) were injected in duplicate onto an EASY-nLC 1000 (mobile phase A was 0.1% FA in water and mobile phase B was 0.1 % FA in ACN) connected to a Q-Exactive Plus (Thermo) equipped with a nano-electrospray ion source. Raw MS/MS data was searched using the Sorcerer 2TM-SEQUEST® algorithm (Sage-N Research) using default peak extraction parameters. Post-search analysis was performed using Scaffold 4 (Proteome Software, Inc.) with protein and peptide probability thresholds set to 95% and 90%, respectively, and one peptide required for identification, and spectra manually validated. Phospho-site localization was determined using Scaffold PTM version 2.1.3 and phospho-sites had to have probabilities greater than 90%.
Detection of direct PKG phosphorylation of TSC2
These assays utilized a TSC2 KO HEK cell line generated in our laboratory. 1 million HEK293T cells were plated overnight in 6 well plate with antibiotics free DMEM supplemented with 10% FBS. The following day 4 μg of Custom Sanger CRISPR plasmid for TSC2 (All-in-one, ready-to-use Cas9 and guide RNA (U6gRNA-Cas9–2A-GFP) expression plasmid against target site GTCGCGGATCTGTTGCAGCCGG, #HS0000020001 Sigma) was transiently transfected using Lipofectamine 3000 (Invitrogen). The following day, media was replaced with DMEM supplemented with 10% FBS and 1% antibiotics, and day after GFP positive cells were FACS sorted to obtain TSC2 KO HEK293T cells and assessed for KO efficiency via immunoblotting. CRISPR TSC2 KO HEK cells were cultured in DMEM supplemented with 10% FBS and 1% antibiotics. For PKG-TSC2 kinase assays, TSC2 was re-introduced into KO HEK cells grown to 50–60% confluence using adenovirus expressing WT-FLAG or S1365A-FLAG TSC2, cultured for 24 hours, and protein isolated.
TSC2 WT or SA (either hsS1364A or hsS1365A)-TSC2 protein was expressed in TSC2 KO HEK cells delivered by plasmid or adenovirus, respectively, and then immunoprecipitated with FLAG or HA magnetic beads (Sigma). Immune complexes were washed three times in lysis buffer and then washed twice in kinase buffer (25 mM HEPES, 10 mM magnesium acetate, and 50 μM ATP) and incubated with 1 μg purified active PKG (Millipore) and 2 μCi [γ−32P]-ATP or 2 μCi [γ−33P]-ATP in 40 μl kinase buffer for 30 minutes at 30°C. The reactions were stopped by washing twice in kinase buffer and boiling in gel loading buffer. Samples were then resolved in a 7.5% TGX gel (Bio-Rad), transferred to a nitrocellulose membrane, and 33P incorporation detected by autoradiography.
We also used the method developed by the Shokat laboratory19 involving a genetically modified PKG (M438G) first reported and validated by the Rudnick lab18 that enlarges the ATP binding site so a bio-orthogonal sulfonated ATPγS (N6-benzyl-ATP) can preferentially bind. TSC2 KO HEKs were seeded into 10-cm plates, infected with FLAG-TSC2 or HA-TSC2, and lysates obtained 24 hrs later in cell signaling lysis buffer. A 200 μL kinase reaction consisted of 25 mM Tris, 10 mM MgCl2, 1 mM 8-CPT-cGMP, 0.4 mM ATP, 6 mM GTP, 1 mM ATP (N6 benzyl ATPγS), and 100 μL cell lysate30. Reactions further received 2 μg of WT or M438G PKG, or buffer to equal final volume. The reaction was carried out at 30o C for 1 hour, then quenched with 20 mM EDTA, and 5 mM p-nitrobenzyl mesylate to alkylate samples (1 hour rotating at room temperature). Immunoprecipitation was carried out (using either FLAG or HA coated beads) overnight at 4oC, samples eluted by boiling in sample buffer for 3 minutes, and the precipitate analyzed by immunoblot using a thiophosphate ester monoclonal Ab (Abcam).
Statistical Analysis
Data presentation format (e.g. mean, SD, SEM), number of experiments and biological independent samples, analysis including multiple comparisons test, and absolute p values for comparisons are provided for each figure panel in the corresponding legend. The majority of summary data are presented as Mean SEM, with a 1-way ANOVA and Tukey multiple comparisons test. Statistical analysis was performed using Graphpad Prism software (Ver. 7a, 2016).
Extended Data
Extended Data Table 1:
Mice Aged 2–3 Months | ||||||||||
TSC2 WT | n | TSC2S1365E | n | p-value | TSC2 WT | n | TSC2 S1365A | n | p-value | |
Body Weight (g) | 25.0 ± 4.1 | 11 | 23.9 ± 2.9 | 13 | 0.60 | 22.5 ± 3.3 | 12 | 21.8 ± 2.4 | 13 | 0.60 |
Heart Weight (mg) | 119.7 ± 23.6 | 6 | 113.6 ± 12.4 | 7 | 0.88 | 117.0 ± 24.7 | 5 | 114.9 ± 12.7 | 4 | 0.88 |
Left Ventricular Weight (mg) | 84.7 ± 17.6 | 6 | 80.5 ± 9.8 | 7 | 0.44 | 81.2 ± 13.4 | 5 | 75.4 ± 4.8 | 4 | 0.44 |
Lung Weight (mg) | 141.7 ± 14.5 | 6 | 128.3 ±13.0 | 7 | 0.72 | 133.4 ± 15.0 | 5 | 130.2 ± 10.8 | 4 | 0.72 |
Tibial Length (mm) | 17.8 ± 0.8 | 6 | 17.6 ± 0.5 | 7 | 0.30 | 18.2 ± 0.8 | 5 | 17.8 ± 0.3 | 4 | 0.30 |
HW/TL | 6.7 ± 1.1 | 6 | 6.4 ± 0.6 | 7 | 0.92 | 6.4 ± 1.1 | 5 | 6.5 ± 0.6 | 4 | 0.92 |
LVW/TL | 4.7 ± 0.8 | 6 | 4.6 ± 0.5 | 7 | 0.57 | 4.5 ± 0.6 | 5 | 4.3 ± 0.3 | 4 | 0.57 |
LuW/TL | 7.9 ± 0.6 | 6 | 7.3 ± 0.7 | 7 | 1.00 | 7.3 ± 0.7 | 5 | 7.3 ± 0.5 | 4 | 1.00 |
Heart Rate (bpm) | 680.4 ± 56.6 | 11 | 690.0 ± 67.9 | 13 | 0.60 | 675.8 ± 21.4 | 12 | 683.2 ± 42.9 | 11 | 0.60 |
%Ejection Fraction | 82.0 ± 1.5 | 11 | 83.4 ± 3.7 | 13 | 0.74 | 82.1 ± 1.6 | 12 | 81.8 ± 2.6 | 11 | 0.74 |
%Fractional Shortening | 57.6 ± 3.0 | 11 | 59.5 ± 4.4 | 13 | 0.76 | 57.7 ± 1.9 | 12 | 57.4 ± 2.9 | 11 | 0.76 |
Mice Aged 9–12 Months | ||||||||||
TSC2 WT | n | TSC2S1365E | n | p-value | TSC2 WT | n | TSC2 S1365A | n | p-value | |
Body Weight (g) | 30.2 ± 3.0 | 6 | 28.5 ± 3.5 | 7 | 0.37 | 29.6 ± 3.1 | 8 | 29.2 ± 4.2 | 7 | 0.37 |
Heart Weight (mg) | 163.4 ±2 5.6 | 6 | 147.9 ± 26.3 | 7 | 0.31 | 159.5 ± 25.0 | 8 | 167.3 ± 32.2 | 7 | 0.31 |
Left Ventricular Weight (mg) | 119.2 ± 23.1 | 6 | 109.3 ± 27.1 | 7 | 0.50 | 125.7 ± 25.6 | 8 | 134.8 ± 33.9 | 7 | 0.50 |
Lung Weight (mg) | 149.2 ± 20.9 | 6 | 130.8 ± 17.4 | 7 | 0.11 | 141.4 ± 24.5 | 8 | 161.5 ± 37.5 | 7 | 0.11 |
Tibial Length (mm) | 19.7 ± 1.4 | 6 | 19.6 ± 1.3 | 7 | 0.90 | 19.1 ± 1.1 | 8 | 19.1 ± 1.2 | 7 | 0.98 |
HW/TL | 8.3 ± 0.85 | 6 | 7.5 ± 0.9 | 7 | 0.14 | 8.3 ± 0.9 | 8 | 8.7 ± 1.4 | 7 | 0.51 |
LVW/TL | 6.0 ± 0.9 | 6 | 5.5 ± 1.0 | 7 | 0.37 | 6.5 ± 1.0 | 8 | 7.0 ± 1.6 | 7 | 0.50 |
LuW/TL | 7.6 ± 0.7 | 6 | 6.7 ± 0.7 | 7 | 0.04 | 7.4 ± 0.9 | 8 | 8.4 ± 1.7 | 7 | 0.15 |
Heart Rate (bpm) | 704.2 ± 37.5 | 6 | 693.6 ± 25.3 | 7 | 0.56 | 671.3 ±7 4.3 | 8 | 659.3 ± 86.4 | 7 | 0.56 |
%Ejection Fraction | 77.2 ± 2.9 | 6 | 77.0 ± 2.7 | 7 | 0.90 | 80.8 ± 1.9 | 8 | 68.2 ± 24.6 | 7 | 0.90 |
%Fractional Shortening | 52.3 ± 3.1 | 6 | 52.1 ±2.8 | 7 | 0.89 | 52.3 ± 2.2 | 8 | 46.5 ± 19.4 | 7 | 0.89 |
The upper table provides results for aged 2–3 month mice (age used in study), and lower table for 9–12 month aged mice. The results show negligible impact of the TSC2 KI mutations in the basal state at both ages. TL – tibia length, bpm – beats/minute. P values are for a two-sided unpaired Student’s t-test.
Supplementary Material
Acknowledgements:
Supported by National Institute of Health – National Heart Lung and Blood Institute grants: HL-135827, HL-119012, HL089297, T32-HL-07227 (DAK), HHSN268201000032C (JEVE, DAK), F31-HL134196 (KMK), F31-HL143905 (BLDE), American Heart Association Post-Doctoral Fellowships (MJR, DL, TN), Deutsche Forschungsgemeinschaft OE 688/1–1 (CUO), Fondation Leducq TransAtlantic Network of Excellence, and Abraham and Virginia Weiss Professorship (DAK). Erika J. Glazer Endowed Chair in Women’s Heart Health (JEVE) and Barbra Streisand Women’s Heart Center (JEVE). This work was supported by the NIH grants (R01AI077610 and R01AI091481) to JDP and the Bloomberg~Kimmel Institute for Cancer Immunotherapy to JDP. We thank Philip Eaton for providing the PKG WT, PKG C42S KI mice, and related plasmid constructs. We thank Dr. Junichi Sadoshima for providing the tandem fluorescent LC3 adenovirus, Brendan Manning for the DNA construct of wild type TSC2, and Jeremy T. Kass for assisting with protein kinase bioinformatics analyses.
Footnotes
Author Information
Any reagents developed for this study, including novel plasmids, viral vectors, and the TSC2 KI mouse models can be made available upon direct request to the corresponding author: David A. Kass, M.D., Johns Hopkins Medical Institutions, Ross Research Building 858, 720 Rutland Avenue, Baltimore MD 21205. The authors declare the following financial competing interests: David Kass, Jonathan Powell, Mark Ranek, Kristen Kokkonen-Simon, and Chirag Patel are co-inventors on a patent application (PCT: 448070145WO1) that was filed July 2018 (provisional filed June 2017). The patent relates to the use of TSC2-S1365/S1364 modifications for immunological applications. David Kass, Jonathan Powell, and Mark Ranek are co-founders and shareholders of Meta-T Cellular Inc., a start-up company that aims to develop applications of this IP for immune therapy.
Data Availability Statement
The authors declare that the data supporting the findings of this study are available within the paper and extended data file. Numerical values corresponding to figures detailing results from in vivo model studies are provided in a separate Source Data file for Figures 1F, 1G, 1H, 2A, 3D, 4C, 4E, and Extended Data 1A. Other source data related to the study are available from the Corresponding Author upon reasonable request.
REFERENCES
- 1.Saxton RA & Sabatini DM mTOR Signaling in Growth, Metabolism, and Disease. Cell 168, 960–976, doi: 10.1016/j.cell.2017.02.004 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Sciarretta S, Forte M, Frati G & Sadoshima J New Insights Into the Role of mTOR Signaling in the Cardiovascular System. Circ Res 122, 489–505, doi: 10.1161/circresaha.117.311147 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zhang Y et al. Coordinated regulation of protein synthesis and degradation by mTORC1. Nature 513, 440–443, doi: 10.1038/nature13492 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ma L, Chen Z, Erdjument-Bromage H, Tempst P & Pandolfi PP Phosphorylation and functional inactivation of TSC2 by Erk implications for tuberous sclerosis and cancer pathogenesis. Cell 121, 179–193, doi: 10.1016/j.cell.2005.02.031 (2005). [DOI] [PubMed] [Google Scholar]
- 5.Potter CJ, Pedraza LG & Xu T Akt regulates growth by directly phosphorylating Tsc2. Nat Cell Biol 4, 658–665, doi: 10.1038/ncb840 (2002). [DOI] [PubMed] [Google Scholar]
- 6.Roux PP, Ballif BA, Anjum R, Gygi SP & Blenis J Tumor-promoting phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90 ribosomal S6 kinase. Proc Natl Acad Sci U S A 101, 13489–13494, doi: 10.1073/pnas.0405659101 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Menon S et al. Spatial control of the TSC complex integrates insulin and nutrient regulation of mTORC1 at the lysosome. Cell 156, 771–785, doi: 10.1016/j.cell.2013.11.049 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Inoki K, Zhu T & Guan KL TSC2 mediates cellular energy response to control cell growth and survival. Cell 115, 577–590 (2003). [DOI] [PubMed] [Google Scholar]
- 9.Inoki K et al. TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell 126, 955–968, doi: 10.1016/j.cell.2006.06.055 (2006). [DOI] [PubMed] [Google Scholar]
- 10.Lee DI et al. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease. Nature 519, 472–476, doi: 10.1038/nature14332 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kim GE & Kass DA Cardiac Phosphodiesterases and Their Modulation for Treating Heart Disease. Handbook of experimental pharmacology 243, 249–269, doi: 10.1007/164_2016_82 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Takimoto E et al. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat. Med 11, 214–222 (2005). [DOI] [PubMed] [Google Scholar]
- 13.Kinoshita H et al. Inhibition of TRPC6 channel activity contributes to the antihypertrophic effects of natriuretic peptides-guanylyl cyclase-A signaling in the heart. Circ. Res 106, 1849–1860 (2010). [DOI] [PubMed] [Google Scholar]
- 14.Kim J, Kundu M, Viollet B & Guan KL AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nature cell biology 13, 132–141, doi: 10.1038/ncb2152 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hariharan N, Zhai P & Sadoshima J Oxidative stress stimulates autophagic flux during ischemia/reperfusion. Antioxid Redox Signal 14, 2179–2190, doi: 10.1089/ars.2010.3488 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ballif BA et al. Quantitative phosphorylation profiling of the ERK/p90 ribosomal S6 kinase-signaling cassette and its targets, the tuberous sclerosis tumor suppressors. Proc Natl Acad Sci U S A 102, 667–672, doi: 10.1073/pnas.0409143102 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mertins P et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature 534, 55–62, doi: 10.1038/nature18003 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wong A, Zhang YW, Jeschke GR, Turk BE & Rudnick G Cyclic GMP-dependent stimulation of serotonin transport does not involve direct transporter phosphorylation by cGMP-dependent protein kinase. J Biol Chem 287, 36051–36058, doi: 10.1074/jbc.M112.394726 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Allen JJ et al. A semisynthetic epitope for kinase substrates. Nature methods 4, 511–516, doi: 10.1038/nmeth1048 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Taneike M et al. mTOR Hyperactivation by Ablation of Tuberous Sclerosis Complex 2 in the Mouse Heart Induces Cardiac Dysfunction with the Increased Number of Small Mitochondria Mediated through the Down-Regulation of Autophagy. PloS one 11, e0152628, doi: 10.1371/journal.pone.0152628 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhang D et al. MTORC1 regulates cardiac function and myocyte survival through 4E-BP1 inhibition in mice. J Clin Invest 120, 2805–2816, doi: 10.1172/jci43008 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Shende P et al. Cardiac raptor ablation impairs adaptive hypertrophy, alters metabolic gene expression, and causes heart failure in mice. Circulation 123, 1073–1082, doi: 10.1161/circulationaha.110.977066 (2011). [DOI] [PubMed] [Google Scholar]
- 23.Moschella PC, Rao VU, McDermott PJ & Kuppuswamy D Regulation of mTOR and S6K1 activation by the nPKC isoforms, PKCepsilon and PKCdelta, in adult cardiac muscle cells. J Mol Cell Cardiol 43, 754–766, doi: 10.1016/j.yjmcc.2007.09.015 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Fonseca BD et al. Pharmacological and genetic evaluation of proposed roles of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK), extracellular signal-regulated kinase (ERK), and p90(RSK) in the control of mTORC1 protein signaling by phorbol esters. J Biol Chem 286, 27111–27122, doi: 10.1074/jbc.M111.260794 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ranganathan V, Wahlin K, Maruotti J & Zack DJ Expansion of the CRISPR-Cas9 genome targeting space through the use of H1 promoter-expressed guide RNAs. Nature communications 5, 4516, doi: 10.1038/ncomms5516 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ran FA et al. Genome engineering using the CRISPR-Cas9 system. Nat Protoc 8, 2281–2308, doi: 10.1038/nprot.2013.143 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Takimoto E et al. Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat Med 11, 214–222, doi: 10.1038/nm1175 (2005). [DOI] [PubMed] [Google Scholar]
- 28.Zhang H et al. Loss of Tsc1/Tsc2 activates mTOR and disrupts PI3K-Akt signaling through downregulation of PDGFR. J Clin Invest 112, 1223–1233, doi: 10.1172/JCI17222 (2003). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nakamura T et al. Prevention of PKG1alpha oxidation augments cardioprotection in the stressed heart. J Clin Invest 125, 2468–2472, doi: 10.1172/JCI80275 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Scotcher J et al. Disulfide-activated protein kinase G Ialpha regulates cardiac diastolic relaxation and fine-tunes the Frank-Starling response. Nature communications 7, 13187, doi: 10.1038/ncomms13187 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.