Visual Abstract

Thymic epithelial cells (TECs) are critical for the development and generation of functionally competent T cells. Until now, the mechanism that regulates the survival of TECs is poorly understood. In the current study, we found that Tsc1 controls the homeostasis of medullary TECs (mTECs) by inhibiting lysosomal-mediated apoptosis pathway in mice. TEC-specific deletion of Tsc1 predominately decreased the cell number of mTECs and, to a lesser content, affected the development cortical TECs. The defect of mTECs caused by Tsc1 deficiency in mice impaired thymocyte development and peripheral T cell homeostasis. Mechanistically, Tsc1 deficiency did not affect the cell proliferation of mTECs but increased the apoptosis of mTECs significantly. RNA-sequencing analysis showed that pathways involved in lysosomal biogenesis, cell metabolism, and apoptosis were remarkably elevated in Tsc1-deficient mTECs compared with their wild-type counterparts. Tsc1-deficient mTECs exhibited overproduction of reactive oxygen species and malfunction of lysosome, with lysosome membrane permeabilization and the release of cathepsin B and cathepsin L to the cytosol, which then lead to Bid cleaved into active truncated Bid and subsequently intrinsic apoptosis. Finally, we showed that the impaired development of mTECs could be partially reversed by decreasing mTORC1 activity via haploinsufficiency of Raptor. Thus, Tsc1 is essential for the homeostasis of mTECs by inhibiting lysosomal-mediated apoptosis through mTORC1-dependent pathways.

Thymus is a primary immune organ for the development and generation of functionally competent T cells that are critical components of the adaptive immune system (1). Thymic epithelial cells (TECs) are essential for T cell development and repertoire selection (1). Based on their function, phenotypes, and localization, TECs are divided into cortical TECs (cTECs) and medullary TECs (mTECs) (2). cTECs are responsible for the homing of T cell precursors through the expression of CXCL12 and CCL25, T cell lineage commitment via DL4-dependent activation of Notch1, and positive selection of thymocytes based on the proper affinity with MHC molecules (36). In contrast, mTECs provide proper microenvironment for the negative selection and maturation of thymocytes as well as the generation of thymic regulatory T (Treg) cells (7, 8).

The development, proliferation, and survival of TECs are regulated by various signaling pathways. It has been established that early TEC specification is dependent on bone morphogenetic protein signaling (1, 9, 10). Recently, two groups reported that NOTCH signaling is essential for the specification of TEC common progenitors into mTEC lineage (11, 12). The subsequent differentiation and maturation of immature mTECs into mature mTECs is regulated by NF-κB signaling pathway (1315). TECs exhibit relative high turnover rates of several weeks (16). The wingless-type MMTV integration site family signaling is critical for the proliferation and expansion of TECs (17). The keratinocyte growth factor promotes thymus regeneration by enhancing the proliferation of TECs (18). Recently, we and another group found that mTOR signaling in TECs controls the proliferation of TECs (19, 20). Although many progresses have been made in understanding the development and proliferation of TECs, the molecular mechanisms that control the survival of TECs are still poorly understood. Several years ago, the STAT3 signaling has been shown to promote the survival and maintenance of mTECs (21, 22). Recently, a group initially reported that the prosurvival protein MCL-1, but not BCL-2 or Bcl-xL, is essential for TEC survival (23). Later, they further revealed that the death of TEC is not mediated by death receptor or necroptosis pathways but by intrinsic apoptosis pathway (also called the mitochondrial pathway) (24). More studies need to be done to further understand the molecular regulation of TEC survival.

Tuberous sclerosis complex (TSC) is an autosomal dominant disorder that affects multiple organ systems and is caused by loss-of-function mutation in either Tsc1 or Tsc2 gene (25). The TSC is composed of TSC1 and TSC2 protein and serve as the negative regulator of mTOR signaling (25). Many pathological alterations of the disease caused by Tsc1/2 gene mutation could be ascribed to the hyperactivation of mTOR signaling pathway. The activity of mTOR signaling is delicately controlled in organisms (26). Hyperactivation of mTOR signaling due to Tsc1 ablation is implicated in the dysfunction of many immune cells. Tsc1 play critical role in the survival of naive T cells, the differentiation of Th cells and memory CD8+ T cells (2730). Tsc1 ablation is also associated with impaired NK cell development, invariant NKT cell differentiation and abnormal macrophage polarization (3133). However, the importance of Tsc1 for TEC biology is unknown. Using mouse models with TEC-specific deletion of Tsc1, we found that Tsc1 ablation predominately impaired the development and survival of mTECs and caused severe thymus atrophy. We further showed that Tsc1 ablation led to lysosomal-mediated apoptosis of mTECs because of hyperactivation of mTORC1 activity. Thus, Tsc1 is essential for the homeostasis of mTECs by inhibiting lysosomal-mediated apoptosis via mTORC1-dependent pathway.

TEC-specific Tsc1 knockout (Tsc1 cKO) mice were obtained by crossing Tsc1flox/flox mice with Foxn1-Cre mice (34). We further crossed Tsc1 cKO mice with Tsc1flox/floxRaptorflox/wt mice to obtain Tsc1 cKO Raptor cHE mice. Foxn1-Cre negative littermates served as controls. Foxn1-Cre mice were the generous gifts of Dr. Y. Zhang from Peking University Health Science Center, Beijing, China. Tsc1flox/flox mice and Raptorflox/flox mice were kindly provided by Dr. H. Zhang (Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China). The primers used to identify the genetic information of mice were listed in Table I. All mice used in the study were bred under pathogen–free conditions. All animal experiments were performed in accordance with the approval of the animal Ethics Committee of Institute of Zoology, Beijing, China.

For flow cytometry staining, the samples were first incubated with 2.4G2 for 20 min at 4°C. Fluorescein-labeled (FL-1061; Vector Laboratories) Ulex europaeus agglutinin I (UEA I) was purchased from Vector Laboratories. Ki-67-PE (556027), Active caspase-3–PE (550914) and allophycocyanin rat anti-mouse CD107a (LAMP1) (560646) were purchased from BD Biosciences. Alexa Fluor 647–conjugated phospho-S6 ribosomal protein (Ser235/236) (D57.2.2E, 4851) and Alexa Fluor 647–conjugated phospho-Akt (Ser473) (193H12) Rabbit mAb were purchased from Cell Signaling Technology. The following Abs were purchased from eBioscience or BioLegend: PerCP/Cy5.5, FITC, PE, PE/Cy7, allophycocyanin, allophycocyanin/Cy7, Brilliant Violet 421 or Alexa Fluor 647–conjugated anti-CD45 (clone 30-F11; BioLegend), CD326 (clone G8.8; BioLegend), CD40 (clone 3/23; BioLegend), CD80 (clone 16-10A1; eBioscience), Ly-51 (clone 6C3; BioLegend), autoimmune regulator (Aire; clone 5H12; eBioscience), CD4 (clone GK1.5; BioLegend), CD8 (BioLegend; clone 53-6.7), CD24 (clone M1/69; eBioscience), CD62L (clone MEL-14; eBioscience), CD25 (clone PC61.5; eBioscience), Foxp3 (clone FJK-16s; eBioscience), CD45RB (clone C363.16A; eBioscience), CD44 (clone IM7; BioLegend), CD69 (clone H1.2F3; eBioscience), TCRB (clone H57-597; BioLegend), and cytochrome C (clone 6H2.B4; BioLegend). Surface staining of cell suspensions was performed in PBS containing 0.1% BSA and 0.02% NaN3 at 4°C for 30 min. Intracellular staining for Ki67, Fxop3, Aire, Bim, Bcl-2, and Mcl-1 was performed using fixation buffer (00-5123-43 and 00-5223-56; eBioscience) and permeabilization buffer (00-8333-56; eBioscience) according to previous experience (3537). For the staining of Phospho-S6 (4851; Cell Signaling Technology), Phospho-Akt (2337; Cell Signaling Technology), and cytochrome C (612310; BioLegend), after staining with cell surface marker, cells were fixed and permeabilized with the BD Biosciences Cytofix/Cytoperm and Perm/Wash solutions. After cell surface staining, the staining of active caspase-3 (550914; BD Pharmingen), JC-1 (C2006; Beyotime Biotechnology), annexin V and propidium iodide (ZP327; ZOMANBIO) were performed according to the manufacturer’s instructions. The flow cytometry was performed with a Gallios Flow Cytometer (Beckman Coulter, Brea, CA).

Thymic stromal cells were obtained according to our previously publications (35, 38). Briefly, freshly isolated thymi were cut into small pieces, then washed the small pieces with DMEM(containing 2% FBS) thoroughly. After the small pieces settled to the bottom of the pipe, we added 2 ml of 1 mg/ml collagenase/dispase (11097113001; Sigma-Aldrich) with 20 U/ml DNAse I (D5025; Sigma-Aldrich) to resuspend these pieces and incubated at 37°C for 45 min. At the end of the digestion, cell suspensions were gently agitated and then 5 ml of PBS containing 1% FBS and 5 mM EDTA was added to neutralize the digestion. At last, cells were centrifuged and resuspended in DMEM (containing 2% FBS) for flow cytometry analysis.

For immunohistology analysis of the thymus, thymi from Tsc1 cKO mice and littermate control mice were fixed in 4% formalin and embedded in paraffin blocks. Sections (6 μm) were stained with H&E and examined by Vectra Automated Multispectral Imaging System (PerkinElmer). The statistical analysis of the area of cortex and medulla was performed using inForm.

For immunofluorescence analysis, serial sections (6 μm) from OCT-embedded frozen thymi or in vitro–cultured TECs were fixed with 4% polyoxymethylene (P1110; Solarbio Life Science) and blocked with donkey serum, washed in PBS–0.05% Tween (60305ES76; TeaSen) and incubated with Abs. The primary Abs were as follows: rat anti–cytokeratin 8 (K8) (Troma-I; Developmental Studies Hybridoma Bank [DSHB]) diluted by 1:200, rabbit anti-K5 (clone AF 138; Covance) diluted by 1:400, LAMP1 (1D4B; DSHB) diluted by 1:100, and cathepsin B (clone D1C7Y; Cell Signaling Technology) diluted by 1:400. The secondary Abs were as follows: Alexa Fluor 488–conjugated donkey anti-rat IgG (H + L) (712-546-150; Jackson ImmunoResearch Laboratories) diluted by 1:400 and Alexa Fluor 594–conjugated donkey anti-rabbit IgG (H + L) (711-586-152; Jackson ImmunoResearch Laboratories) diluted by 1:400. Nuclei were stained with DAPI (D9542; Sigma-Aldrich). Images were acquired with a laser scanning N-SIM Super-Resolution Confocal Microscope (Nikon, Tokyo, Japan).

The organs of 10-mo-old wild-type (WT) and Tsc1 cKO mice were harvested and fixed in 4% paraformaldehyde, embedded in paraffin (39601095; Leica), sectioned (5 μm), and stained with H&E. For the detection of autoantibodies, sera from 10-mo-old WT and Tsc1 cKO mice were prepared, diluted with PBS (1:30), and incubated with frozen sections of rag2−/− mice, followed by Alexa Fluor 488–conjugated donkey anti-mouse IgG (H + L) Abs (715-546-150; 1:300; Jackson ImmunoResearch Laboratories).

After isolation of TECs from pooled thymi by method mentioned above, TECs or mTECs were enriched using anti-mouse CD45 microbeads (130052301; Miltenyi Biotec) and then sorted with a MoFlo XDP cell sorter (Beckman Coulter, Brea, CA). Total RNA was extracted using MicroElute Total RNA Kits (R6831; Omega Bio-Tek), and reverse transcription was performed with SuperScript III Reverse Transcriptase (18080-093; Invitrogen) according to the manufacturer’s instructions. Real-time PCR was performed using multiple kits (TaKaRa SYBR Premix Ex Taq, RR420) on a CFX96 apparatus (Bio-Rad Laboratories, Hercules, CA). The primers using in this study are listed in Table II.

Four to six thymi of 4-wk-old WT and Tsc1 cKO mice were pooled and digested to isolate thymic stromal cells. The isolated stromal cells were enriched using anti-mouse CD45 microbeads (130052301; Miltenyi Biotec) then sorted for mTECs using a Fusion cell sorter (BD Biosciences). Total RNA were extracted using TRIzol agent, and first strand cDNA synthesis and cDNA amplification was performed using Smart-Seq2 method. DEGseq was used to identify the differentially expressed genes between WT and Tsc1-deficient mTECs. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was performed on all the different genes of each cell using the KOBAS online search tool (http://kobas.cbi.pku.edu.cn/). The RNA-sequencing (RNA-seq) datasets for this study can be found in the National Center for Biotechnology Information Sequence Read Archive database (http://www.ncbi.nlm.nih.gov/sra/) with the accession number PRJNA723503.

Because of the rarity of freshly isolated mTECs, Simple Western was used to detect the expression of Bid and some other proteins. mTECs were sorted by cell sorter then were lysed with radioimmunoprecipitation assay (RIPA). Simple Western was performed using 12–230 kDa Separation Module (SM-W002-1; ProteinSimple) according to the manufacturer’s instructions.

The primary and secondary Abs used for Simple Western are as follows: human/mouse BID Ab (AF860; R&D Systems), cleaved caspase-3 (Asp175) (5A1E) Rabbit mAb (9664; Cell Signaling Technology), human/mouse caspase-8 Ab (AF1650; R&D Systems), and mouse/rat cathepsin L Ab (AF1515; R&D Systems) and anti-goat secondary HRP Ab (043-522; Simple Western), anti-mouse secondary HRP Ab (042-205; Simple Western), and anti-rabbit Secondary HRP Ab (042-206; Simple Western).

mTECs were sorted from pooled thymi and lysed in RIPA. Briefly, cells were washed with cold PBS and lysed in RIPA buffer (50 mM Tris-HCl [pH 7.4], 1% NP-40 [ab142227; Abcam], 0.25% Na-deoxycholate [S817543; Macklin], 150 mM NaCl, and 1 mM EDTA [pH 7.4]) with a protease inhibitor mixture (P8340; Sigma-Aldrich). Protein concentration was determined using a bicinchoninic acid assay. Proteins were analyzed by SDS-PAGE (L5750; Sigma-Aldrich) and transferred onto PVDF membranes (IPFL00010; Merck Millipore). The PVDF membranes were blocked with 5% nonfat dried milk (LP0031; Oxoid) for 1 h and then incubated with primary Abs overnight on a shaker at 4°C. The HPR-coupled anti-mouse (074-1806; KPL International) or anti-rabbit (070-1506; KPL International) secondary Abs were then added and were detected through chemiluminescence (WBKLS0500; Merck Millipore).

The following primary Abs were used: phospho-s6 ribosomal protein (Ser240/244) (D68F8) Rabbit mAb (5364; Cell Signaling Technology) diluted by 1:10000, β-Tubulin (T5201; Sigma-Aldrich) diluted by 1:1000, LAMP1 (AB 528127, 1D4B; DSHB) diluted by 1:1000, cathepsin B (31718, D1C7Y; Cell Signaling Technology) diluted by 1:1000, and mouse/rat cathepsin L Ab (AF1515; R&D Systems) diluted by 1:1000.

Cellular fractionation was performed using cellular fractionation kit–HT (ab109718; Abcam) according to the protocol described by Huai et al. (39). Briefly, cells were digested, washed once with buffer A, and centrifuged. The pellet was resuspended buffer A at the concentration of 6 × 106 cells/ml, added equal volume of buffer B, and then incubated at room temperature for 7 min on a rotator and centrifuged sequentially at 5,000 × g and 10,000 × g at 4°C for 1 min each. The supernatant was collected as cytosolic fraction. To get the lysosome fraction, the cytosolic fraction was further centrifuged at 100,000 × g for 1 h. The pellet represented the lysosomal fraction.

The detection of reactive oxygen species (ROS) in freshly isolated mTECs was performed using CellROX Deep Red Flow Cytometry Assay Kit (C10491; Thermo Fisher Scientific) according to manufacturers’ instruction. The detection of ROS in cultured TECs was performed using Reactive Oxygen Species Assay Kit (S0033; Beyotime Biotechnology) following manufacturers’ instruction.

For in vitro TEC culture, thymi from WT and Tsc1 cKO neonatal mice were digested as mentioned above. Small thymic fragments from each step were collected and pooled. Fragments were allowed to settle and washed twice with Thymic Epithelial Cell Medium (3911; ScienCell). The remaining thymic explants were plated in 24-well plates with Thymic Epithelial Cell Medium and cultured at 37°C with 5% CO2. TECs will grow in this medium, and cells gradually die. After ∼8 d, TECs were digested with tyrosine for annexin V and propidium iodide staining.

The lysosome of live cells was stained by LysoTracker Red (C1046; Beyotime Biotechnology) according to the manufacturer’s instructions.

To test the activity of cathepsin B, Magic Red Cathepsin assays was performed. Freshly isolated thymic stromal cells were incubated with the cathepsin B substrate (6133; ImmunoChemistry Technologies) at 37°C for 1 h; after staining with surface Abs, the activity of cathepsin B was examining by flow cytometry.

All data are presented as the means ± SD. A Student unpaired t test for comparison of means was used to compare groups. A p value <0.05 was considered statistically significant.

To investigate the role of Tsc1 in TECs, we generated mice with conditional inactivation of Tsc1 in TECs (Foxn1-Cre; Tsc1flox/flox; designated as Tsc1 cKO mice in brief; the age-matched Tsc1fl/fl mice were used as control mice; Table I). The efficient inactivation of Tsc1 locus was confirmed by real-time PCR (Supplemental Fig. 1A, 1B). We first assessed the effect of TEC-specific inactivation of Tsc1 on the development of thymus at embryonic and postnatal stages. After TEC-specific deletion of Tsc1, the thymus size was smaller in comparison with age-matched controls (Supplemental Fig. 1C). Consistently, total thymocyte cellularity reduced significantly from embryonic day 17.5 (E17.5) to 4-wk-old mice compared with WT littermate controls (Fig. 1A). We next investigated the impact of Tsc1 cKO on thymic epithelia. The cell numbers of TECs isolated from Tsc1 cKO mice decreased significantly at E17.5, newborn, and postnatal 1, 2, and 4 wk (Fig. 1B), although the frequencies of TECs only decreased obviously at 2 and 4 wk (Supplemental Fig. 1D, 1E). TECs are composed of cTECs and mTECs, which are responsible for the positive and negative selection of thymocytes, respectively (1). The percentages and cell numbers of mTECs decreased dramatically in Tsc1 cKO mice in comparison with littermate Tsc1f/f mice (Fig. 1C, 1D, Supplemental Fig. 1F). In contrast, the absolute numbers of cTECs were almost unaffected by the loss of Tsc1, although the relative frequencies of cTECs increased significantly (Fig. 1E, 1F, Supplemental Fig. 1F).

FIGURE 1.

TEC-specific Tsc1 deficiency predominately impairs mTEC compartment. The cellularity of thymocytes (A) and TECs (B) from WT and Tsc1 cKO mice at E17.5, newborn (NB), and postnatal 1, 2, and 4 wk. For each time points, at least three mice of each genotype were analyzed. Statistical analysis of the frequency (C) and the cellularity (D) of mTECs from WT and Tsc1 cKO mice at indicated time points. For each time point, at least three mice of each genotype were analyzed. mTECs were defined as CD45EpCAM+UEA I+Ly-51. Statistical analysis of the frequency (E) and cellularity (F) of cTECs from WT and Tsc1 cKO mice at indicated time points. For each time point, at least three mice of each genotype were analyzed. cTECs were defined as CD45EpCAM+UEA ILy-51+. (G) The representative histological thymus sections and the ratio of medulla area to cortex area in 4-wk-old WT and Tsc1 cKO mice. Original magnification, 5×. Three sections per thymus of four WT mice and three Tsc1 cKO mice were analyzed. (H) Frozen thymic sections derived from 4-wk-old WT and Tsc1 cKO mice for the expression of K5 (red) and K8 (green). Scale bars, 500 μm. (I) Immunofluorescence staining showing the expression of K5 in thymi of 4-wk-old WT and Tsc1 cKO mice. Scale bars, 100 μm. (J) The structure of CMJ revealed by the staining of K5 (red) and K8 (green) in thymi of 4-wk-old WT and Tsc1 cKO mice. Scale bars, 500 μm. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 1.

TEC-specific Tsc1 deficiency predominately impairs mTEC compartment. The cellularity of thymocytes (A) and TECs (B) from WT and Tsc1 cKO mice at E17.5, newborn (NB), and postnatal 1, 2, and 4 wk. For each time points, at least three mice of each genotype were analyzed. Statistical analysis of the frequency (C) and the cellularity (D) of mTECs from WT and Tsc1 cKO mice at indicated time points. For each time point, at least three mice of each genotype were analyzed. mTECs were defined as CD45EpCAM+UEA I+Ly-51. Statistical analysis of the frequency (E) and cellularity (F) of cTECs from WT and Tsc1 cKO mice at indicated time points. For each time point, at least three mice of each genotype were analyzed. cTECs were defined as CD45EpCAM+UEA ILy-51+. (G) The representative histological thymus sections and the ratio of medulla area to cortex area in 4-wk-old WT and Tsc1 cKO mice. Original magnification, 5×. Three sections per thymus of four WT mice and three Tsc1 cKO mice were analyzed. (H) Frozen thymic sections derived from 4-wk-old WT and Tsc1 cKO mice for the expression of K5 (red) and K8 (green). Scale bars, 500 μm. (I) Immunofluorescence staining showing the expression of K5 in thymi of 4-wk-old WT and Tsc1 cKO mice. Scale bars, 100 μm. (J) The structure of CMJ revealed by the staining of K5 (red) and K8 (green) in thymi of 4-wk-old WT and Tsc1 cKO mice. Scale bars, 500 μm. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal
Table I.

The list of primers used for mouse identification

GeneDirectionSequence
Tsc1flox/flox Forward AGGAGGCCTCTTCTGCTACC 
Reverse CAGCTCCGACCATGAAGTG 
Tsc1 deletion Forward GCGGCTCTGGAGGAACACAAT 
Reverse TCGGTCATGCTGTAGCTGTCTG 
Raptorflox/flox Forward CTCAGTAGTGGTATGTGCTCAG 
Reverse GGGTACAGTATGTCAGCACAG 
Foxn1-Cre Forward TGCCTAGACGTTGGTCCTCAT 
Reverse AATGTTGCTGGATAGTTTTTACTGC 
GeneDirectionSequence
Tsc1flox/flox Forward AGGAGGCCTCTTCTGCTACC 
Reverse CAGCTCCGACCATGAAGTG 
Tsc1 deletion Forward GCGGCTCTGGAGGAACACAAT 
Reverse TCGGTCATGCTGTAGCTGTCTG 
Raptorflox/flox Forward CTCAGTAGTGGTATGTGCTCAG 
Reverse GGGTACAGTATGTCAGCACAG 
Foxn1-Cre Forward TGCCTAGACGTTGGTCCTCAT 
Reverse AATGTTGCTGGATAGTTTTTACTGC 
Table II.

The list of primers used in real-time PCR

GeneDirectionSequence
Ctsb Forward AGACCTGCTTACTTGCTGTG 
Reverse GGAGGGATGGTGTATGGTAAG 
Ctsd Forward TGACAAGTCCAGCACCTATG 
Reverse CTCCACCTTGATACCTCTTGC 
Ctsl Forward AGCAAGAACCTCGACCATG 
Reverse TTCCATACCCCATTCACTTCC 
TPP Forward AAGACACAAGTTCCGCCC 
Reverse CGTGGGAAAACATTGCTGAAG 
Hexb Forward GAAGCTCCTGGTCTCCATTAC 
Reverse GTCTCTAAACCTCGTAACGCTC 
Neu1 Forward GCCAAGTTCATCGCCATG 
Reverse CCCTGTGTCTACATCGTTCAC 
Gns Forward GTCATCGCACCAAGAAACAAG 
Reverse TCAACTGAGAGCAGGGTTTG 
Dnase2a Forward CAAGACGGTGTAGGGTACATC 
Reverse GAGGTTGGTCGTTGTAGAGTAG 
Aga Forward CGTCAACACTTGGCCTTTTAAG 
Reverse CATCACACTGCTCCTTCTCAC 
Psap Forward TGGACATGATTAAGGGCGAG 
Reverse CTGTTTCTGGTTTTGCTCGG 
Lamp1 Forward ACAAACCCCACTGTATCCAAG 
Reverse CATTTGGGCTGATGTTGAACG 
Ctns Forward CTGGAAAATGGCAGTTCAACC 
Reverse CGCGAGGCACTATAACTTCATC 
Laptm4b Forward ACTGTGCTTGTCTATCCGAAC 
Reverse CCTTAAGTGTCAAAAGAATGCCG 
Litaf Forward CCACAGGGCTCATTACAGG 
Reverse GTCATAGAAGGAGACAGGCTG 
Atp6v0b Forward CTCTACCTCGGGATCTTTGTG 
Reverse AGATTCGACCACATGAAGGG 
Atp6v0c Forward GCCCTAATCCTCTCCACAAAG 
Reverse GGACACTGCACATTTACAAGAC 
Atp6v0a4 Forward CTAAAACAGACCCTCCCACC 
Reverse ACATCACAGCGAACAGGAAG 
GeneDirectionSequence
Ctsb Forward AGACCTGCTTACTTGCTGTG 
Reverse GGAGGGATGGTGTATGGTAAG 
Ctsd Forward TGACAAGTCCAGCACCTATG 
Reverse CTCCACCTTGATACCTCTTGC 
Ctsl Forward AGCAAGAACCTCGACCATG 
Reverse TTCCATACCCCATTCACTTCC 
TPP Forward AAGACACAAGTTCCGCCC 
Reverse CGTGGGAAAACATTGCTGAAG 
Hexb Forward GAAGCTCCTGGTCTCCATTAC 
Reverse GTCTCTAAACCTCGTAACGCTC 
Neu1 Forward GCCAAGTTCATCGCCATG 
Reverse CCCTGTGTCTACATCGTTCAC 
Gns Forward GTCATCGCACCAAGAAACAAG 
Reverse TCAACTGAGAGCAGGGTTTG 
Dnase2a Forward CAAGACGGTGTAGGGTACATC 
Reverse GAGGTTGGTCGTTGTAGAGTAG 
Aga Forward CGTCAACACTTGGCCTTTTAAG 
Reverse CATCACACTGCTCCTTCTCAC 
Psap Forward TGGACATGATTAAGGGCGAG 
Reverse CTGTTTCTGGTTTTGCTCGG 
Lamp1 Forward ACAAACCCCACTGTATCCAAG 
Reverse CATTTGGGCTGATGTTGAACG 
Ctns Forward CTGGAAAATGGCAGTTCAACC 
Reverse CGCGAGGCACTATAACTTCATC 
Laptm4b Forward ACTGTGCTTGTCTATCCGAAC 
Reverse CCTTAAGTGTCAAAAGAATGCCG 
Litaf Forward CCACAGGGCTCATTACAGG 
Reverse GTCATAGAAGGAGACAGGCTG 
Atp6v0b Forward CTCTACCTCGGGATCTTTGTG 
Reverse AGATTCGACCACATGAAGGG 
Atp6v0c Forward GCCCTAATCCTCTCCACAAAG 
Reverse GGACACTGCACATTTACAAGAC 
Atp6v0a4 Forward CTAAAACAGACCCTCCCACC 
Reverse ACATCACAGCGAACAGGAAG 

We then examined the anatomical changes of the thymus in Tsc1 cKO mice. We found that the area of the medullary compartment of Tsc1 cKO mice was significantly smaller compared with WT mice as showed by reduced medulla to cortex ratio (Fig. 1G). The medulla and cortex express its specific marker K5 and K8, respectively (1). A histological examination revealed a smaller medulla region in mutant mice as expected (Fig. 1H). What is more, the thymus medullary region in Tsc1 cKO mice appeared tattered and was sparsely dispersed in cortical region (Fig. 1H). The mutant mice also displayed a medulla of damaged architecture compared with controls (Fig. 1I). In contrast, the mutant mice had a cortex with regular architecture that was indistinguishable in comparison with control mice (Supplemental Fig. 2A). In contrast to well-demarcated cortico-medullary junction (CMJ) in WT thymi, the typical circumscribed boundary at CMJ was disrupted in thymi of Tsc1 cKO mice (Fig. 1J). These data collectively indicated that the mTEC compartment was severely impaired after TEC-specific ablation of Tsc1.

We further investigate whether the reduction of mTECs is intrinsic or due to a defect of progenitors. More and more evidence indicated mTECs were derived from progenitors expressing molecular traits specific for cTECs, including β5t, CD205, and IL-7 (4046). We then evaluated the expression of CD205 on TECs at E17.5; the results showed that the expression of CD205 on TECs is similar between WT mice and Tsc1 cKO mice (Supplemental Fig. 3A). So, the reduction of mTECs may not be due to early stage of mTEC lineage commitment.

We then focused our studies on mTECs. As the maturation of mTECs, the expression of MHC class II (MHCII), CD80, CD40, and Aire is upregulated (47). The percentage of CD40+ mTECs was unaffected by inactivation of Tsc1 (Fig. 2A). mTECs are heterogeneous with regard to the expression of MHCII and CD80, enabling their subdivision into MHCIIloCD80lo mTEClo immature subsets and MHCIIhiCD80hi mTEChi mature subsets (1). Similar to the expression of CD40, the frequency of mTEChi and mTEClo subsets was unaltered after conditional Tsc1 cKO (Fig. 2C, 2D). Aire is responsible for the expression of many tissue-restricted Ags (TRAs) and the induction of immune tolerance (1). Tsc1 ablation in TECs did not change the expression of Aire in mTECs (Fig. 2E). However, because of the overall reduction of mTECs, the absolute cell numbers of CD40+, MHCIIhi, mTEClo, and Aire+ mTECs are all decreased dramatically (Fig. 2B, 2D, 2F). In contrast, both the percentage and the cell numbers of MHCIIhi cTECs were unchanged after Tsc1 ablation (Supplemental Fig. 2B, 2C, 2D). We further examined the impact of Tsc1 deletion on the differentiation of embryonic mTECs, and the results showed that the expression of MHCII on mTECs at E17.5 was comparable between WT and Tsc1 cKO mice (Supplemental Fig. 3B), indicating the primitive mTEC maturation is not obviously affected by Tsc1 deletion. These results collectively demonstrated that the ablation of Tsc1 in TECs did not affect the differentiation and maturation of mTECs but resulted in the overall reduction of mTECs.

FIGURE 2.

The maturation and differentiation of mTECs are unaffected after TEC-specific ablation of Tsc1. (A) Flow cytometric profiles and average frequency of CD40+ mTECs from 4-wk-old WT and Tsc1 cKO mice. (B) The absolute cell numbers of CD40+ mTECs of 4-wk-old WT (n = 7) and Tsc1 cKO mice (n = 7). (C) Flow cytometric profiles and average frequency of MHCIIlow and MHCIIhigh mTECs from 4-wk-old WT and Tsc1 cKO mice. (D) The absolute cell numbers of MHCIIlow and MHCIIhigh mTECs of 4-wk-old WT (n = 10) and Tsc1 cKO mice (n = 10). (E) Flow cytometric profiles and average frequency of Aire+ mTECs from 4-wk-old WT and Tsc1 cKO mice. (F) The absolute cell numbers of Aire+ mTECs of 4-wk-old WT (n = 7) and Tsc1 cKO mice (n = 7). ***p < 0.001.

FIGURE 2.

The maturation and differentiation of mTECs are unaffected after TEC-specific ablation of Tsc1. (A) Flow cytometric profiles and average frequency of CD40+ mTECs from 4-wk-old WT and Tsc1 cKO mice. (B) The absolute cell numbers of CD40+ mTECs of 4-wk-old WT (n = 7) and Tsc1 cKO mice (n = 7). (C) Flow cytometric profiles and average frequency of MHCIIlow and MHCIIhigh mTECs from 4-wk-old WT and Tsc1 cKO mice. (D) The absolute cell numbers of MHCIIlow and MHCIIhigh mTECs of 4-wk-old WT (n = 10) and Tsc1 cKO mice (n = 10). (E) Flow cytometric profiles and average frequency of Aire+ mTECs from 4-wk-old WT and Tsc1 cKO mice. (F) The absolute cell numbers of Aire+ mTECs of 4-wk-old WT (n = 7) and Tsc1 cKO mice (n = 7). ***p < 0.001.

Close modal

The generation, survival, and development of thymocytes depend largely on the thymic epithelial microenvironment (1). We next evaluated the thymopoiesis of 4-wk-old Tsc1fl/fl and Tsc1 cKO mice. The frequencies of CD4+CD8 single-positive (CD4SP) and CD4CD8+ single-positive (CD8SP) thymocytes decreased mildly in Tsc1 cKO mice when compared with Tsc1fl/fl mice and the frequencies of CD4CD8 double-negative (DN) and CD4+CD8+ double-positive (DP) thymocytes had on obvious change (Fig. 3A). However, the absolute numbers of DN, DP, CD4SP, and CD8SP thymocytes all reduced dramatically in Tsc1 cKO mice compared with Tsc1fl/fl controls (Fig. 3B). The frequencies of CD69+TCRβint/hi thymocytes showed a tendency to decrease (Supplemental Fig. 4A), indicating the positive selection of thymocytes is potentially impaired in Tsc1 cKO mice. The positively selected thymocytes subsequently migrate into medullary region to undergo negative selection (1). Thymocytes that survive negative selection proceed further maturation in the thymus before they export to the periphery as naive T cells (48). This postselection maturation is reflected by phenotypic and functional changes, including the increased surface expression of CD62L and downregulated expression of CD24 (49, 50). The frequencies of mature (CD24CD62L+) CD4SP and CD8SP thymocytes decreased significantly in Tsc1 cKO mice compared with littermate controls (Fig. 3C). These observations suggested that the postselection maturation of thymocytes was impaired in mice with TEC-specific Tsc1 ablation.

FIGURE 3.

TEC-specific ablation of Tsc1 impairs the development of thymocytes. The representative flow cytometric plots and average frequency (A) and cellularity (B) of DN, DP, CD4SP, and CD8SP thymocytes of 4-wk-old WT (n = 7) and Tsc1 cKO (n = 7) mice. (C) Flow cytometry profiles and frequency of the CD24 and CD62L expression in CD4+CD8 or CD4CD8+ thymocytes of 4-wk-old WT (n = 3) and Tsc1 cKO (n = 3) mice. The representative flow cytometric profiles (D), frequency (D), and cell numbers (E) of CD4+CD8Foxp3+ tTreg cells in the thymus of Tsc1 cKO mice (n = 3) and WT littermates (n = 3). (F) Flow cytometric plots and frequency for the staining of tTreg precursors (CD4+CD8CD25+Foxp3) of 4-wk-old WT (n = 3) and Tsc1 cKO mice (n = 3). (G) The ratio of tTreg precursor (CD4+CD8CD25+Foxp3) to mature tTreg (CD4+CD8CD25+Foxp3+) cells in the thymus of Tsc1 cKO mice (n = 3) and WT littermates (n = 3). Flow cytometry plots and frequency (H) and absolute cell numbers (I) of CD4+ and CD8+ T cells in the spleen of 4-wk-old WT (n = 5) and Tsc1 cKO mice (n = 6). *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 3.

TEC-specific ablation of Tsc1 impairs the development of thymocytes. The representative flow cytometric plots and average frequency (A) and cellularity (B) of DN, DP, CD4SP, and CD8SP thymocytes of 4-wk-old WT (n = 7) and Tsc1 cKO (n = 7) mice. (C) Flow cytometry profiles and frequency of the CD24 and CD62L expression in CD4+CD8 or CD4CD8+ thymocytes of 4-wk-old WT (n = 3) and Tsc1 cKO (n = 3) mice. The representative flow cytometric profiles (D), frequency (D), and cell numbers (E) of CD4+CD8Foxp3+ tTreg cells in the thymus of Tsc1 cKO mice (n = 3) and WT littermates (n = 3). (F) Flow cytometric plots and frequency for the staining of tTreg precursors (CD4+CD8CD25+Foxp3) of 4-wk-old WT (n = 3) and Tsc1 cKO mice (n = 3). (G) The ratio of tTreg precursor (CD4+CD8CD25+Foxp3) to mature tTreg (CD4+CD8CD25+Foxp3+) cells in the thymus of Tsc1 cKO mice (n = 3) and WT littermates (n = 3). Flow cytometry plots and frequency (H) and absolute cell numbers (I) of CD4+ and CD8+ T cells in the spleen of 4-wk-old WT (n = 5) and Tsc1 cKO mice (n = 6). *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

In addition to providing the molecular cues for the development of conventional T cells, mTECs also provide niches for the development of thymic Treg (tTreg) cells (8). The dramatic reduction of mTECs in Tsc1 cKO mice promoted us to examine whether tTreg cell development was impaired. Both the percentage and absolute cell number of CD4+CD8Foxp3+ tTreg cells reduced considerably in Tsc1 cKO mice compared with WT littermate controls (Fig. 3D, 3E). The mature CD4+CD8CD25+Foxp3+ tTreg cells develop from CD4+CD8CD25+Foxp3 precursors in the thymus (51). We found that the percentage of CD4+CD8CD25+Foxp3 tTreg precursors and the ratio of CD4+CD8CD25+Foxp3 precursors to mature CD4+CD8CD25+Foxp3+ tTreg cells elevated significantly in mice with TEC-specific Tsc1 deletion (Fig. 3F, 3G). These results demonstrated that the development of CD4+CD8CD25+Foxp3+ tTreg cells was blocked at the tTreg precursor stage because of the impaired thymic medulla microenvironment in Tsc1 cKO mice.

The mature naive T cells generated in the thymus populate the peripheral lymphoid organs to perform their functions (52). To assess the thymus output function, we evaluated the recent thymic emigrants that are defined as CD4+CD62LhiCD45RBint splenocytes (53). The mutant mice displayed a significant, albeit limited, reduction of recent thymic emigrants (Supplemental Fig. 4B). Consistently, both the cell numbers of CD4+ and CD8+ T cells considerably decreased in the spleen of Tsc1 cKO mice compared with controls (Fig. 3I), although the frequency the CD4+ and CD8+ T cells only mildly decreased (Fig. 3H). Furthermore, there was noticeable increment of CD44+CD62L+ or CD44+CD62L effector/memory-like CD4 and CD8 T cells and reduction of CD44CD62L+ naive T cells in Tsc1 cKO mice (Supplemental Fig. 4C, 4D). The percentage of Treg cells in the spleen slightly elevated in Tsc1 cKO mice; however, the absolute cell number of Treg cells diminished substantially in Tsc1 cKO mice compared with littermate controls (Supplemental Fig. 4E–G). These data demonstrated the homeostasis of peripheral T cells was disturbed in Tsc1 cKO mice.

mTECs are essential for the establishment of central immune tolerance by expressing the Aire-dependent and Aire-independent TRAs and by providing proper niches for tTreg development (8, 54, 55). The defect of mTEC compartment and the reduction of Treg cells indicated that Tsc1 cKO mice may develop autoimmune disease. We first dissected the impact of Tsc1 ablation on the expression of Aire-dependent and Aire-independent TRAs by comparing our RNA-seq results with published dataset (5458), and the results showed only a fraction of Aire-dependent and Aire-independent TRAs was disturbed by Tsc1 cKO (Supplemental Fig. 5A, 5B). We then examined the presence of lymphocyte infiltrates and autoantibodies in lung, pancreas, liver, and kidney. The lung of 10-mo-old Tsc1 cKO mice exhibited obvious presence of infiltrated lymphocytes and autoantibodies. The other examined tissues showed no noticeable lymphocyte infiltration and autoantibodies in comparison with age-matched Tsc1fl/fl mice (Supplemental Fig. 5C, 5D). Thus, we concluded that Tsc1 cKO mice develop mild autoimmune disease, which may be due to the less-changed TRAs expression.

We then explored the causes for the reduction of mTECs in Tsc1 cKO mice. We found the proliferation ability of mTECs was comparable between WT and Tsc1 cKO mice assessed by Ki67 staining (Fig. 4A, 4B), and similar phenomenon was observed as to cTECs (Supplemental Fig. 6A). Tsc1 ablation has been reported to associate with impaired cell survival ability (27, 28). We therefore measured the expression of active caspase-3, a hallmark of apoptotic cell death. Both the newborn and 4-wk-old Tsc1-deficient mTECs exhibited significantly increased active caspase-3 expression in comparison with WT mTECs (Fig. 4C, 4D, Supplemental Fig. 6B). In contrast, the apoptosis of 4-wk-old cTECs was unaltered by Tsc1 deletion (Supplemental Fig. 6C). Although the apoptosis of newborn cTECs increased after Tsc1 ablation, the apoptosis level of newborn cTECs was much lower than 4-wk-old cTECs (Supplemental Fig. 6C). Collectively, these results indicated TEC-specific Tsc1 deletion predominantly promoted the apoptosis of mTECs and slightly affected the apoptosis of cTECs at certain time points of mice after birth. Furthermore, by using an in vitro TEC culture system established in our laboratory (59, 60), we found the apoptosis of TEC was also increased after Tsc1 deletion, as showed by increased annexin V+ cells (Fig. 4E). Based on these results, we concluded that the reduction of mTECs in Tsc1 cKO mice was due to increased apoptosis.

FIGURE 4.

Tsc1-deficient mTECs exhibit excessive apoptosis. Flow cytometric profiles (A) and frequency (B) for the expression of Ki67 in mTECs of 4-wk-old Tsc1 cKO mice (n = 7) and WT littermate controls (n = 7). Flow cytometric analysis (C) and frequencies (D) of active caspase-3+ mTECs from newborn Tsc1 cKO mice (n = 7) and WT littermates (n = 7). (E) Annexin V/propidium iodide costaining of in vitro–cultured Tsc1 cKO and WT TECs. (F) Scatterplot comparison of global gene expression profiles between WT and Tsc1-deficient mTECs. Transcripts with logFoldChange >0 and p value >0.05 are heighted (red indicated upregulated and blue indicated downregulated). (G) The top 10% of upregulated KEGG pathways in Tsc1-deficient mTECs versus WT mTECs. (H) The representative significantly changed pathways in Tsc1-deficient mTECs versus WT mTECs analyzed by Gene Set Enriched Analysis (GSEA). ***p < 0.001.

FIGURE 4.

Tsc1-deficient mTECs exhibit excessive apoptosis. Flow cytometric profiles (A) and frequency (B) for the expression of Ki67 in mTECs of 4-wk-old Tsc1 cKO mice (n = 7) and WT littermate controls (n = 7). Flow cytometric analysis (C) and frequencies (D) of active caspase-3+ mTECs from newborn Tsc1 cKO mice (n = 7) and WT littermates (n = 7). (E) Annexin V/propidium iodide costaining of in vitro–cultured Tsc1 cKO and WT TECs. (F) Scatterplot comparison of global gene expression profiles between WT and Tsc1-deficient mTECs. Transcripts with logFoldChange >0 and p value >0.05 are heighted (red indicated upregulated and blue indicated downregulated). (G) The top 10% of upregulated KEGG pathways in Tsc1-deficient mTECs versus WT mTECs. (H) The representative significantly changed pathways in Tsc1-deficient mTECs versus WT mTECs analyzed by Gene Set Enriched Analysis (GSEA). ***p < 0.001.

Close modal

To elucidate the molecular events causing the defect survival capacity of Tsc1-deficient mTECs, we performed RNA-seq analysis of mTECs isolated from 4-wk-old Tsc1fl/fl and Tsc1 cKO mice. The global gene expression profiles comparison between WT and Tsc1-deficient mTECs revealed that 475 probes increased and 380 probes decreased after Tsc1 cKO (Fig. 4F). We then used KEGG analysis method to investigate the affected signaling pathways after Tsc1 ablation by comparing the differentially expressed genes shown in (Fig. 4F. The downregulated top ten pathways included Ag processing and presentation, human T cell lymphotropic virus type 1 infection, and so on (data not shown). The upregulated top 10% pathways included lysosome, metabolic pathway, oxidative phosphorylation, and so on (Fig. 4G). Notably, the apoptosis was among these markedly upregulated pathways (Fig. 4G), which is consistent with the excessive apoptosis of Tsc1-deficient mTECs showed in (Fig. 4C–E. Gene Set Enriched Analysis also revealed the increase of metabolic pathway, oxidative phosphorylation, lysosome, and apoptosis of Tsc1-deficient mTECs compared with Tsc1fl/fl mTECs (Fig. 4H). These results indicated that Tsc1-deficient mTECs displayed increased cell metabolism, oxidative phosphorylation, lysosomal biogenesis, and apoptosis. Based on the increased apoptosis of Tsc1 cKO mTECs (Fig. 4C–E), we further scrutinized the apoptosis KEGG pathway. We found that the increased apoptosis signaling of Tsc1 cKO mTECs was mainly through the cathepsin–Bid axis (Supplemental Fig. 7), which is termed lysosomal-mediated apoptosis (61). Consistently, it is well known that the abnormal lysosomal biogenesis, increased cell metabolism, and oxidative phosphorylation could trigger lysosomal-mediated apoptosis (29, 39, 61, 62).

Given that Tsc1 ablation increased cell metabolism and oxidative phosphorylation, which would lead to increased ROS production (29), we next examined the ROS production in mTECs. The production of ROS in Tsc1 cKO mTECs increased significantly compared with WT mTECs (Fig. 5A). Consistently, the cultured Tsc1-deficient TECs also displayed elevated ROS production in comparison with WT controls (Supplemental Fig. 8A). According to the increased lysosome pathway from RNA-seq analysis, we then examined the changes of lysosome in Tsc1-deficient mTECs. The expression of lysosome marker (LAMP1) increased markedly in Tsc1-deficient mTECs compared with WT mTECs (Fig. 5B). The expression of many lysosomal biogenesis genes also increased obviously in Tsc1 cKO mTECs, as assessed by real-time PCR (Fig. 5C; Table II). The amount and size of lysosome both increased in cultured Tsc1-deficient TECs than WT controls as assessed by immunofluorescence and LysoTracker staining (Supplemental Fig. 8B, 8C). Lysosomes are the major digestive organelles with an acidic interior within cells (63). The function of lysosomes is largely dependent on the lysosomal proteases; among them, the cathepsins are the best-known proteases (63, 64). Cathepsins are mainly responsible for lysosomal protein degradation, but they also involved in lysosomal-mediated cell death (65). We then detected the expression of cathepsin B in Tsc1-deficient mTECs. The results showed that the expression of cathepsin B increased significantly in Tsc1-deficient mTECs compared with WT mTECs (Fig. 5D). Consistently, the activity of cathepsin B also increased as measured by Magic Red Cathepsin staining assay (Fig. 5E). These data demonstrated that the production of ROS and lysosomal biogenesis increased significantly in Tsc1-deficient mTECs relative to WT mTECs.

FIGURE 5.

Tsc1 deficiency leads to lysosomal-mediated apoptosis of mTECs. (A) The production of ROS in Tsc1-deficient (n = 3) and WT (n = 3) mTECs was examined by flow cytometry. (B) Flow cytometric profiles (left) and frequency (right) for the expression of LAMP1 in mTECs of 4-wk-old Tsc1 cKO mice (n = 8) and WT controls (n = 7). (C) The expression of representative lysosomal biogenesis genes were examined by real-time PCR. (D) The expression of cathepsin B in Tsc1-deficient and WT mTECs isolated from 4-wk-old mice were examined by Simple Western. (E) Magic Red Cathepsin staining for cathepsin B in Tsc1-deficient and WT mTECs were measured by flow cytometry. (F) The cytosolic and lysosome fraction were separated and the expression of cathepsin B and cathepsin L in cultured Tsc1 cKO and WT TECs were measured by Western blot. (G) The expressions of Bid and tBid in Tsc1-deficient and WT mTECs were examined by Simple Western. (H) Flow cytometric analysis for JC-1 monomers and aggregates in mTECs of 4-wk-old Tsc1 cKO mice (n = 3) and WT littermates (n = 3). *p < 0.05, ***p < 0.001.

FIGURE 5.

Tsc1 deficiency leads to lysosomal-mediated apoptosis of mTECs. (A) The production of ROS in Tsc1-deficient (n = 3) and WT (n = 3) mTECs was examined by flow cytometry. (B) Flow cytometric profiles (left) and frequency (right) for the expression of LAMP1 in mTECs of 4-wk-old Tsc1 cKO mice (n = 8) and WT controls (n = 7). (C) The expression of representative lysosomal biogenesis genes were examined by real-time PCR. (D) The expression of cathepsin B in Tsc1-deficient and WT mTECs isolated from 4-wk-old mice were examined by Simple Western. (E) Magic Red Cathepsin staining for cathepsin B in Tsc1-deficient and WT mTECs were measured by flow cytometry. (F) The cytosolic and lysosome fraction were separated and the expression of cathepsin B and cathepsin L in cultured Tsc1 cKO and WT TECs were measured by Western blot. (G) The expressions of Bid and tBid in Tsc1-deficient and WT mTECs were examined by Simple Western. (H) Flow cytometric analysis for JC-1 monomers and aggregates in mTECs of 4-wk-old Tsc1 cKO mice (n = 3) and WT littermates (n = 3). *p < 0.05, ***p < 0.001.

Close modal

Some recent studies illustrated that lysosomal cathepsins are also involved in cell apoptosis, which pivotally depends on the release of cathepsins into the cytosol, a process known as lysosomal membrane permeabilization (LMP) (66). The overproduction of ROS could induce LMP (61), and the enlarged lysosome size also make lysosomes more prone to LMP (61, 63). The increased ROS production and the abnormal lysosomal biogenesis in Tsc1-deficient mTECs reminded us that these cells may undergo LMP. We thus examined the leakage of cathepsins from lysosome into cytosol in WT and Tsc1 cKO cultured TECs. The results showed that the cytosolic cathepsin B and cathepsin L increased obviously in Tsc1 cKO TECs compared with controls (Fig. 5F), indicating Tsc1 ablation resulted in the leakage of cathepsin B and cathepsin L into cytosol. Furthermore, in comparison with WT TECs, Tsc1 cKO TECs not only exhibited an elevated cathepsin B expression but also showed a redistribution of cathepsin B from perinuclear localization to diffused pattern throughout the cytosol (Supplemental Fig. 8D). These results indicated the Tsc1 ablation leads to obvious LMP in TECs.

Apoptosis signaling after LMP often leads to activation of intrinsic pathway and caspase cascade (66). The cytosolic cathepsins cleavage Bid into its active form truncated Bid (tBid), which then translocates to mitochondria so as to induce mitochondria membrane permeabilization and release of cytochrome C and ultimately trigger intrinsic apoptosis pathway (66, 67). We thus detected the cleavage of Bid into tBid in mTECs after Tsc1 deletion, and the results showed that the cleavage of Bid into tBid increased dramatically in Tsc1-deficient mTECs relative to WT controls (Fig. 5G). It has been established that the cleavage of Bid into tBid is mainly mediated by cathepsins in lysosomal-mediated apoptosis or by caspase-8 in Fas-induced apoptosis (61, 68). The expression of Fas and caspase-8 were comparable between Tsc1-deficient mTECs and WT controls (Supplemental Fig. 8E, 8F). These data demonstrated the increased level of tBid was mainly due to the cleavage of Bid by cathepsins. We next examined the mitochondrial membrane potential of Tsc1-deficient mTECs. The results showed that Tsc1 ablation reduced the mitochondrial membrane potential of mTECs significantly as showed by the decreased JC-1 aggregates to monomers ratio (Fig. 5H). Similarly, the in vitro–cultured, Tsc1-deficient TECs also displayed diminished mitochondrial membrane potential in contrast to WT controls (Supplemental Fig. 8G, 8H). In summary, these results illustrated that Tsc1 ablation led to the overproduction of ROS and the malfunction of lysosome. The elevated ROS caused LMP and the release of cathepsins into cytosol, which then cleaved Bid into active tBid that subsequently induced intrinsic apoptosis.

The most important function of TSC1 is to restrain the activity of mTORC1 (26). To elucidate whether the increased apoptosis of mTECs in Tsc1 cKO mice was due to the hyperactivation of mTORC1, we first evaluated the activity of mTORC1 in Tsc1-deficient mTECs. Indeed, Tsc1 ablation resulted in hyperactivation of mTORC1 as assessed by increased phosphorylation of ribosomal protein S6 at Ser240/244 (Fig. 6A). Next, we generated TEC-specific Tsc1 cKO mice that are heterozygous for Raptor, a scaffold protein of mTORC1 (Foxn1-Cre; Tsc1flox/floxRaptorWT/flox, designated as Tsc1 cKO Raptor cHE mice briefly). The genetic reduction of mTORC1 activity in Tsc1 cKO mice markedly increased the size, weight, and cellularity of the thymus compared with those in Tsc1 cKO mice (Fig. 6B–D). We further investigated the impact the heterozygote deletion of Raptor on the development of TECs. The cellularity of TECs was partially rescued after heterozygote deletion of Raptor in Tsc1 cKO mice (Fig. 6E). Furthermore, the genetic knockdown of mTORC1 activity also resulted in partially but significant recovery of the percentage and cellularity of mTECs compared with those in Tsc1 cKO mice (Fig. 6F–H). In contrast, the absolute cell numbers of cTECs were comparable in WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice (Fig. 6J), although the frequency of cTECs displayed obvious change in different genotype mice (Fig. 6F, 6I). Importantly, the increased apoptosis of mTECs could also partially reverse in Tsc1 cKO Raptor cHE mice in contrast to Tsc1 cKO mice. These results clearly indicated that the impaired survival of mTECs in Tsc1 cKO mice is due to the hyperactivation of mTORC1.

FIGURE 6.

Constitutive mTORC1 overactivation results in apoptosis of mTECs. (A) Western blot analysis for the phosphorylation of pS6Ser240/244 in the sorted mTECs of 4-wk-old Tsc1fl/fl and Tsc1 cKO mice. Six to eight thymi were pooled together to acquire enough mTECs to perform Western blot. (B) Representative pictures of the thymus from 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. (C and D) Thymus weight (C) thymocyte cellularity (D) of 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. At least four mice were analyzed for each genotype. (E) TEC cellularity of 4-wk-old WT (n = 5), Tsc1 cKO (n = 7), and Tsc1 cKO Raptor cHE (n = 8) mice. (F) Flow cytometric profiles for the staining of mTECs and cTECs of 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. (G and H) Frequency (G) and cellularity (H) of mTECs from 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. At least five mice were analyzed for each genotype. (I and J) Frequency (I) and cellularity (J) of cTECs of 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. At least five mice were analyzed for each genotype. (K and L) Flow cytometric profiles (K) and statistical analysis (L) of active caspase-3+ mTECs from newborn WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. At least five mice were analyzed for each genotype. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 6.

Constitutive mTORC1 overactivation results in apoptosis of mTECs. (A) Western blot analysis for the phosphorylation of pS6Ser240/244 in the sorted mTECs of 4-wk-old Tsc1fl/fl and Tsc1 cKO mice. Six to eight thymi were pooled together to acquire enough mTECs to perform Western blot. (B) Representative pictures of the thymus from 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. (C and D) Thymus weight (C) thymocyte cellularity (D) of 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. At least four mice were analyzed for each genotype. (E) TEC cellularity of 4-wk-old WT (n = 5), Tsc1 cKO (n = 7), and Tsc1 cKO Raptor cHE (n = 8) mice. (F) Flow cytometric profiles for the staining of mTECs and cTECs of 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. (G and H) Frequency (G) and cellularity (H) of mTECs from 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. At least five mice were analyzed for each genotype. (I and J) Frequency (I) and cellularity (J) of cTECs of 4-wk-old WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. At least five mice were analyzed for each genotype. (K and L) Flow cytometric profiles (K) and statistical analysis (L) of active caspase-3+ mTECs from newborn WT, Tsc1 cKO, and Tsc1 cKO Raptor cHE mice. At least five mice were analyzed for each genotype. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

We found in this study that Tsc1 played a critical role for the survival of mTECs via preventing lysosomal-mediated apoptosis. Deficiency of Tsc1 in TECs reduced the mTEC compartment from embryonic to adult stages. Although Tsc1 deficiency reduced the cell numbers of mTECs dramatically, Tsc1 may be dispensable for the differentiation and functional maturation of mTECs, as evidenced by the intact frequencies of mature mTEC populations (CD40+, Aire+, and mTEChi cells) in Tsc1-deficient mice. It has been reported that mTECs derive from progenitor cells sharing a similarity with the cTEC lineage, including β5t, CD205, and IL-7 (4046). The similar expression of CD205 on WT and Tsc1 cKO TECs at E17.5 indicated the defect of mTECs may not be due to early stage of mTEC lineage commitment. Also, Tsc1 ablation exhibited little effect on the cell number of cTECs; the transcriptional program of cTECs may be altered as reflected by the reduction of cortex-dependent positive selection and the decrease of cortex-resident DP thymocytes. Recently, some studies discovered the unexpected contribution of several pathways to control TEC development. Similar to the role of Tsc1 in TECs, two studies demonstrated that Stat3 is also essential for the survival immature mTECs (21, 22). Another report revealed that Irf4 is imperative for tTreg homeostasis by regulating the expression of several chemokines and costimulatory molecules on TECs (69, 70). The well-known tumor suppressor p53 is also critical for the homeostasis of TECs, and TEC-specific ablation of p53 disrupts thymopoiesis and central immune tolerance (71). More studies need to be done to systematically uncover the molecular regulation of TEC development and homeostasis.

Tsc1 deficiency resulted in excessive apoptosis of mTECs in vivo, and we further recapitulated this phenomenon using the in vitro TEC culture system. Many publications attributed the excessive apoptosis of Tsc1-deficient cells to downregulation of prosurvival member Bcl-2 and upregulation of proapoptotic member Bim (27, 28, 32, 72). Although, our results indicated that Tsc1 deficiency in mTECs did not alter the expression of Bim (data not shown). Although the expression of Bcl-2 showed a tendency to decrease in Tsc1-deficient mTECs (data not shown), a recent study clearly demonstrated that Bcl-2 is dispensable for TEC survival (23). Thus we ruled out the potential involvement of Bim and Bcl-2 in the defect survival capacity of Tsc1-deficient mTECs. Noticeably, by performing RNA-seq analysis and subsequent experiments, we proved that Tsc1-deificient mTECs undergo lysosomal-mediated apoptosis. Therefore, we revealed a previous unappreciated role of Tsc1 in the regulation of cell apoptosis in mTECs.

We further illustrated that Tsc1 deficiency impaired the survival of mTECs through hyperactivation of mTORC1 because genetic reduction of Raptor could partially ameliorate the increased apoptosis of mTECs. The tight control of mTOR activity is very important for the development of organisms (26). Tsc1-deficient TECs exhibited exorbitant mTORC1 activity, which leads to the increased apoptosis of mTECs. The increased apoptosis of mTECs could partially reverse by heterozygote deletion of Raptor in Tsc1 cKO TECs. These observations are consistent with another report in which the authors showed that the development defect of NK cells caused by Tsc1 ablation could partially rescue by genetic knockdown of Raptor (32). Combining with previous publications from our laboratory (19) and by Wang et al. (20), in which the authors showed that mTOR deficiency caused a poor mTEC development in mice, these results collectively documented the that the tightly control of mTORC1 activity is critical for mTEC development; that is, more or less mTOR activity is harmful to the mTEC development.

The present study also unveiled that Tsc1 ablation promoted the expression of cell metabolism and lysosomal biogenesis–related genes. It is widely acknowledged that mTORC1 promotes cell metabolism (26); however, the role of mTORC1 on lysosomal biogenesis is complicated. Some reports elucidated that mTORC1 inhibits lysosomal biogenesis (7375), whereas another group reported that mTORC1 activation caused by Tsc2 knockout enhances lysosomal biogenesis (76). Noticeably, two recent studies revealed that epidermal Tsc1 deletion upregulates lysosomal biogenesis (77), and Tsc2 deficiency also promotes lysosomal biogenesis (78), which is coincident with our present study.

It is well known that lysosome is closely related with the degradation of macromolecules, which is associated with autophagy, endocytosis, and phagocytosis (63). However, emerging evidence indicated that lysosome is also involved in cell death, but most of them are under the in vitro conditions (39, 79, 80). We revealed in this study that Tsc1-deficient mTECs underwent lysosomal-mediated apoptosis in vivo. We proved that Tsc1 ablation led to LMP of mTECs. In Tsc1-deficient mTECs, LMP may be induced by two factors: one is the overproduction of ROS (81), the other is the abnormal of lysosomal biogenesis because larger lysosome may breakdown more efficiently with stress (82). The results of LMP is the leakage of cathepsins from lysosome into cytosol and the cytosolic cathepsins cleaved inactive Bid into active tBid that induced mitochondria permeabilization and cell death of mTECs.

In summary, Tsc1 deficiency in TECs caused obvious poor survival of mTECs and disruption of thymus medulla, which resulted in the defect of thymocyte development. Tsc1 ablation in mTECs led to the hyperactivation of mTORC1, which resulted in the increment of oxidative phosphorylation and lysosomal biogenesis. The elevated oxidative phosphorylation produced excessive ROS that caused LMP. The LMP caused leakage of cathepsins into cytosol, and the cytoplasmic cathepsins subsequently cleaved Bid into active tBid. The tBid then translocated to the mitochondria and caused mitochondrial membrane depolarization, which at last initiated the apoptosis of mTECs. The schematic diagram of Tsc1 controlling lysosomal-mediated apoptosis in mTECs was shown in visual abstract.

We thank Dr. Peng Wang for critical reading the manuscript. We thank Dr. Yu Zhang for the gifts of Foxn1-Cre mice. We acknowledge Dr. Hongbing Zhang for the gifts of Tsc1flox/flox mice and Raptorflox/flox mice.

This work was supported by National Natural Science Foundation of China General and Key Program grants (31800754 [to Z.L.], 31930041 [to Y.Z.], and 82070774 [to X.X.]), the National Key Research and Development Program of China (2017YFA0105002 and 2017YFA0104402 [to Y.Z.]), and the Knowledge Innovation Program of Chinese Academy of Sciences (XDA16030301 [to Y.Z.]).

Z.L., Q.Z., Y.Z., and Liguang Sun designed the experiments; Z.L., Q.Z., Lina Sun, X.D., T.L., and L.T. conducted the experiments; Z.Z. performed the bioinformatics analysis; Z.L. and X.X. analyzed and/or interpreted the results and revised the manuscript; Z.L., Liguang Sun, and Y.Z. supervised the studies and wrote the manuscript.

The sequences presented in this article have been submitted to the National Center for Biotechnology Information Sequence Read Archive database (http://www.ncbi.nlm.nih.gov/sra/) under accession number PRJNA723503.

The online version of this article contains supplemental material.

Abbreviations used in this article

Aire

autoimmune regulator

CD4SP

CD4+CD8 single-positive

CD8SP

CD4CD8+ single-positive

CMJ

cortico-medullary junction

cTEC

cortical TEC

DN

double-negative

DP

double-positive

DSHB

Developmental Studies Hybridoma Bank

E17.5

embryonic day 17.5

K8

cytokeratin 8

KEGG

Kyoto Encyclopedia of Genes and Genomes

LMP

lysosomal membrane permeabilization

MHCII

MHC class II

mTEC

medullary TEC

RIPA

radioimmunoprecipitation assay

RNA-seq

RNA-sequencing

ROS

reactive oxygen species

tBid

truncated Bid

TEC

thymic epithelial cell

TRA

tissue-restricted Ag

Treg

regulatory T

TSC

tuberous sclerosis complex

Tsc1 cKO

Tsc1 knockout

tTreg

thymic Treg

UEA I

Ulex europaeus agglutinin I

WT

wild-type

1.
Abramson
J.
,
G.
Anderson
.
2017
.
Thymic epithelial cells.
Annu. Rev. Immunol.
35
:
85
118
.
2.
Nowell
C. S.
,
A. M.
Farley
,
C. C.
Blackburn
.
2007
.
Thymus organogenesis and development of the thymic stroma.
Methods Mol. Biol.
380
:
125
162
.
3.
Takada
K.
,
I.
Ohigashi
,
M.
Kasai
,
H.
Nakase
,
Y.
Takahama
.
2014
.
Development and function of cortical thymic epithelial cells.
Curr. Top. Microbiol. Immunol.
373
:
1
17
.
4.
Žuklys
S.
,
A.
Handel
,
S.
Zhanybekova
,
F.
Govani
,
M.
Keller
,
S.
Maio
,
C. E.
Mayer
,
H. Y.
Teh
,
K.
Hafen
,
G.
Gallone
, et al
2016
.
Foxn1 regulates key target genes essential for T cell development in postnatal thymic epithelial cells.
Nat. Immunol.
17
:
1206
1215
.
5.
Hozumi
K.
,
C.
Mailhos
,
N.
Negishi
,
K.
Hirano
,
T.
Yahata
,
K.
Ando
,
S.
Zuklys
,
G. A.
Holländer
,
D. T.
Shima
,
S.
Habu
.
2008
.
Delta-like 4 is indispensable in thymic environment specific for T cell development.
J. Exp. Med.
205
:
2507
2513
.
6.
Koch
U.
,
E.
Fiorini
,
R.
Benedito
,
V.
Besseyrias
,
K.
Schuster-Gossler
,
M.
Pierres
,
N. R.
Manley
,
A.
Duarte
,
H. R.
Macdonald
,
F.
Radtke
.
2008
.
Delta-like 4 is the essential, nonredundant ligand for Notch1 during thymic T cell lineage commitment.
J. Exp. Med.
205
:
2515
2523
.
7.
Malchow
S.
,
D. S.
Leventhal
,
V.
Lee
,
S.
Nishi
,
N. D.
Socci
,
P. A.
Savage
.
2016
.
Aire enforces immune tolerance by directing autoreactive T cells into the regulatory T cell lineage.
Immunity.
44
:
1102
1113
.
8.
Anderson
G.
,
S.
Baik
,
J. E.
Cowan
,
A. M.
Holland
,
N. I.
McCarthy
,
K.
Nakamura
,
S. M.
Parnell
,
A. J.
White
,
P. J.
Lane
,
E. J.
Jenkinson
,
W. E.
Jenkinson
.
2014
.
Mechanisms of thymus medulla development and function.
Curr. Top. Microbiol. Immunol.
373
:
19
47
.
9.
Bleul
C. C.
,
T.
Corbeaux
,
A.
Reuter
,
P.
Fisch
,
J. S.
Mönting
,
T.
Boehm
.
2006
.
Formation of a functional thymus initiated by a postnatal epithelial progenitor cell.
Nature.
441
:
992
996
.
10.
Gordon
J.
,
S. R.
Patel
,
Y.
Mishina
,
N. R.
Manley
.
2010
.
Evidence for an early role for BMP4 signaling in thymus and parathyroid morphogenesis.
Dev. Biol.
339
:
141
154
.
11.
Li
J.
,
J.
Gordon
,
E. L. Y.
Chen
,
S.
Xiao
,
L.
Wu
,
J. C.
Zúñiga-Pflücker
,
N. R.
Manley
.
2020
.
NOTCH1 signaling establishes the medullary thymic epithelial cell progenitor pool during mouse fetal development.
Development.
147
:
dev178988
.
12.
Liu
D.
,
A. I.
Kousa
,
K. E.
O’Neill
,
P.
Rouse
,
M.
Popis
,
A. M.
Farley
,
S. R.
Tomlinson
,
S.
Ulyanchenko
,
F.
Guillemot
,
P. A.
Seymour
, et al
2020
.
Canonical Notch signaling controls the early thymic epithelial progenitor cell state and emergence of the medullary epithelial lineage in fetal thymus development.
Development.
147
:
dev178582
.
13.
Sun
L.
,
H.
Li
,
H.
Luo
,
Y.
Zhao
.
2014
.
Thymic epithelial cell development and its dysfunction in human diseases.
BioMed Res. Int.
2014
:
206929
.
14.
Hikosaka
Y.
,
T.
Nitta
,
I.
Ohigashi
,
K.
Yano
,
N.
Ishimaru
,
Y.
Hayashi
,
M.
Matsumoto
,
K.
Matsuo
,
J. M.
Penninger
,
H.
Takayanagi
, et al
2008
.
The cytokine RANKL produced by positively selected thymocytes fosters medullary thymic epithelial cells that express autoimmune regulator.
Immunity.
29
:
438
450
.
15.
Luan
R.
,
Z.
Liang
,
Q.
Zhang
,
L.
Sun
,
Y.
Zhao
.
2019
.
Molecular regulatory networks of thymic epithelial cell differentiation.
Differentiation.
107
:
42
49
.
16.
Gray
D. H.
,
N.
Seach
,
T.
Ueno
,
M. K.
Milton
,
A.
Liston
,
A. M.
Lew
,
C. C.
Goodnow
,
R. L.
Boyd
.
2006
.
Developmental kinetics, turnover, and stimulatory capacity of thymic epithelial cells.
Blood.
108
:
3777
3785
.
17.
Heinonen
K. M.
,
J. R.
Vanegas
,
S.
Brochu
,
J.
Shan
,
S. J.
Vainio
,
C.
Perreault
.
2011
.
Wnt4 regulates thymic cellularity through the expansion of thymic epithelial cells and early thymic progenitors.
Blood.
118
:
5163
5173
.
18.
Sun
L.
,
H.
Luo
,
H.
Li
,
Y.
Zhao
.
2013
.
Thymic epithelial cell development and differentiation: cellular and molecular regulation.
Protein Cell.
4
:
342
355
.
19.
Liang
Z.
,
L.
Zhang
,
H.
Su
,
R.
Luan
,
N.
Na
,
L.
Sun
,
Y.
Zhao
,
X.
Zhang
,
Q.
Zhang
,
J.
Li
, et al
2018
.
MTOR signaling is essential for the development of thymic epithelial cells and the induction of central immune tolerance.
Autophagy.
14
:
505
517
.
20.
Wang
H. X.
,
J.
Shin
,
S.
Wang
,
B.
Gorentla
,
X.
Lin
,
J.
Gao
,
Y. R.
Qiu
,
X. P.
Zhong
.
2016
.
mTORC1 in Thymic epithelial cells is critical for thymopoiesis, T-cell generation, and temporal control of γδT17 development and TCRγ/δ recombination.
PLoS Biol.
14
:
e1002370
.
21.
Lomada
D.
,
M.
Jain
,
M.
Bolner
,
K. A.
Reeh
,
R.
Kang
,
M. C.
Reddy
,
J.
DiGiovanni
,
E. R.
Richie
.
2016
.
Stat3 signaling promotes survival and maintenance of medullary thymic epithelial cells.
PLoS Genet.
12
:
e1005777
.
22.
Satoh
R.
,
K.
Kakugawa
,
T.
Yasuda
,
H.
Yoshida
,
M.
Sibilia
,
Y.
Katsura
,
B.
Levi
,
J.
Abramson
,
Y.
Koseki
,
H.
Koseki
, et al
2016
.
Requirement of Stat3 signaling in the postnatal development of thymic medullary epithelial cells.
PLoS Genet.
12
:
e1005776
.
23.
Jain
R.
,
J. M.
Sheridan
,
A.
Policheni
,
M.
Heinlein
,
L. C.
Gandolfo
,
G.
Dewson
,
G. K.
Smyth
,
S. N.
Sansom
,
N. Y.
Fu
,
J. E.
Visvader
, et al
2017
.
A critical epithelial survival axis regulated by MCL-1 maintains thymic function in mice.
Blood.
130
:
2504
2515
.
24.
Jain
R.
,
J. D.
Mintern
,
I.
Tan
,
G.
Dewson
,
A.
Strasser
,
D. H. D.
Gray
.
2018
.
How do thymic epithelial cells die?
Cell Death Differ.
25
:
1002
1004
.
25.
Henske
E. P.
,
S.
Jóźwiak
,
J. C.
Kingswood
,
J. R.
Sampson
,
E. A.
Thiele
.
2016
.
Tuberous sclerosis complex.
Nat. Rev. Dis. Primers
2
:
16035
.
26.
Saxton
R. A.
,
D. M.
Sabatini
.
2017
.
mTOR signaling in growth, metabolism, and disease. [Published erratum appears in 2017 Cell 169: 361–371.]
Cell
168
:
960
976
.
27.
Yang
K.
,
G.
Neale
,
D. R.
Green
,
W.
He
,
H.
Chi
.
2011
.
The tumor suppressor Tsc1 enforces quiescence of naive T cells to promote immune homeostasis and function.
Nat. Immunol.
12
:
888
897
.
28.
Zhang
L.
,
H.
Zhang
,
L.
Li
,
Y.
Xiao
,
E.
Rao
,
Z.
Miao
,
H.
Chen
,
L.
Sun
,
H.
Li
,
G.
Liu
,
Y.
Zhao
.
2012
.
TSC1/2 signaling complex is essential for peripheral naïve CD8+ T cell survival and homeostasis in mice.
PLoS One.
7
:
e30592
.
29.
Shrestha
S.
,
K.
Yang
,
J.
Wei
,
P. W.
Karmaus
,
G.
Neale
,
H.
Chi
.
2014
.
Tsc1 promotes the differentiation of memory CD8+ T cells via orchestrating the transcriptional and metabolic programs.
Proc. Natl. Acad. Sci. USA.
111
:
14858
14863
.
30.
Park
Y.
,
H. S.
Jin
,
J.
Lopez
,
C.
Elly
,
G.
Kim
,
M.
Murai
,
M.
Kronenberg
,
Y. C.
Liu
.
2013
.
TSC1 regulates the balance between effector and regulatory T cells.
J. Clin. Invest.
123
:
5165
5178
.
31.
Zhu
L.
,
T.
Yang
,
L.
Li
,
L.
Sun
,
Y.
Hou
,
X.
Hu
,
L.
Zhang
,
H.
Tian
,
Q.
Zhao
,
J.
Peng
, et al
2014
.
TSC1 controls macrophage polarization to prevent inflammatory disease.
Nat. Commun.
5
:
4696
.
32.
Yang
M.
,
S.
Chen
,
J.
Du
,
J.
He
,
Y.
Wang
,
Z.
Li
,
G.
Liu
,
W.
Peng
,
X.
Zeng
,
D.
Li
, et al
2016
.
NK cell development requires Tsc1-dependent negative regulation of IL-15-triggered mTORC1 activation.
Nat. Commun.
7
:
12730
.
33.
Wu
J.
,
J.
Yang
,
K.
Yang
,
H.
Wang
,
B.
Gorentla
,
J.
Shin
,
Y.
Qiu
,
L. G.
Que
,
W. M.
Foster
,
Z.
Xia
, et al
2014
.
iNKT cells require TSC1 for terminal maturation and effector lineage fate decisions. [Published errata appear in 2018 J. Clin. Invest. 128: 1707–1708 and 2017 J. Clin. Invest. 127: 4216.]
J. Clin. Invest.
124
:
1685
1698
.
34.
Soza-Ried
C.
,
C. C.
Bleul
,
M.
Schorpp
,
T.
Boehm
.
2008
.
Maintenance of thymic epithelial phenotype requires extrinsic signals in mouse and zebrafish.
J. Immunol.
181
:
5272
5277
.
35.
Sun
L.
,
H.
Li
,
H.
Luo
,
L.
Zhang
,
X.
Hu
,
T.
Yang
,
C.
Sun
,
H.
Chen
,
L.
Zhang
,
Y.
Zhao
.
2013
.
Phosphatase Wip1 is essential for the maturation and homeostasis of medullary thymic epithelial cells in mice.
J. Immunol.
191
:
3210
3220
.
36.
Chu
Z.
,
C.
Sun
,
L.
Sun
,
C.
Feng
,
F.
Yang
,
Y.
Xu
,
Y.
Zhao
.
2020
.
Primed macrophages directly and specifically reject allografts.
Cell. Mol. Immunol.
17
:
237
246
.
37.
Hou
Y.
,
L.
Zhu
,
H.
Tian
,
H. X.
Sun
,
R.
Wang
,
L.
Zhang
,
Y.
Zhao
.
2018
.
IL-23-induced macrophage polarization and its pathological roles in mice with imiquimod-induced psoriasis.
Protein Cell.
9
:
1027
1038
.
38.
Sun
L.
,
C.
Sun
,
Z.
Liang
,
H.
Li
,
L.
Chen
,
H.
Luo
,
H.
Zhang
,
P.
Ding
,
X.
Sun
,
Z.
Qin
,
Y.
Zhao
.
2015
.
FSP1(+) fibroblast subpopulation is essential for the maintenance and regeneration of medullary thymic epithelial cells.
Sci. Rep.
5
:
14871
.
39.
Huai
J.
,
F. N.
Vögtle
,
L.
Jöckel
,
Y.
Li
,
T.
Kiefer
,
J. E.
Ricci
,
C.
Borner
.
2013
.
TNFα-induced lysosomal membrane permeability is downstream of MOMP and triggered by caspase-mediated NDUFS1 cleavage and ROS formation.
J. Cell Sci.
126
:
4015
4025
.
40.
Anderson
G.
,
Y.
Takahama
.
2012
.
Thymic epithelial cells: working class heroes for T cell development and repertoire selection.
Trends Immunol.
33
:
256
263
.
41.
Mayer
C. E.
,
S.
Žuklys
,
S.
Zhanybekova
,
I.
Ohigashi
,
H. Y.
Teh
,
S. N.
Sansom
,
N.
Shikama-Dorn
,
K.
Hafen
,
I. C.
Macaulay
,
M. E.
Deadman
, et al
2016
.
Dynamic spatio-temporal contribution of single β5t+ cortical epithelial precursors to the thymus medulla.
Eur. J. Immunol.
46
:
846
856
.
42.
Ohigashi
I.
,
S.
Zuklys
,
M.
Sakata
,
C. E.
Mayer
,
Y.
Hamazaki
,
N.
Minato
,
G. A.
Hollander
,
Y.
Takahama
.
2015
.
Adult thymic medullary epithelium is maintained and regenerated by lineage-restricted cells rather than bipotent progenitors.
Cell Rep.
13
:
1432
1443
.
43.
Ohigashi
I.
,
S.
Zuklys
,
M.
Sakata
,
C. E.
Mayer
,
S.
Zhanybekova
,
S.
Murata
,
K.
Tanaka
,
G. A.
Holländer
,
Y.
Takahama
.
2013
.
Aire-expressing thymic medullary epithelial cells originate from β5t-expressing progenitor cells.
Proc. Natl. Acad. Sci. USA.
110
:
9885
9890
.
44.
Ribeiro
A. R.
,
C.
Meireles
,
P. M.
Rodrigues
,
N. L.
Alves
.
2014
.
Intermediate expression of CCRL1 reveals novel subpopulations of medullary thymic epithelial cells that emerge in the postnatal thymus.
Eur. J. Immunol.
44
:
2918
2924
.
45.
Ribeiro
A. R.
,
P. M.
Rodrigues
,
C.
Meireles
,
J. P.
Di Santo
,
N. L.
Alves
.
2013
.
Thymocyte selection regulates the homeostasis of IL-7-expressing thymic cortical epithelial cells in vivo.
J. Immunol.
191
:
1200
1209
.
46.
Baik
S.
,
E. J.
Jenkinson
,
P. J. L.
Lane
,
G.
Anderson
,
W. E.
Jenkinson
.
2013
.
Generation of both cortical and Aire(+) medullary thymic epithelial compartments from CD205(+) progenitors.
Eur. J. Immunol.
43
:
589
594
.
47.
Takahama
Y.
,
I.
Ohigashi
,
S.
Baik
,
G.
Anderson
.
2017
.
Generation of diversity in thymic epithelial cells.
Nat. Rev. Immunol.
17
:
295
305
.
48.
Hsu
F. C.
,
A. G.
Pajerowski
,
M.
Nelson-Holte
,
R.
Sundsbak
,
V. S.
Shapiro
.
2011
.
NKAP is required for T cell maturation and acquisition of functional competency.
J. Exp. Med.
208
:
1291
1304
.
49.
Li
J.
,
Y.
Li
,
J. Y.
Yao
,
R.
Jin
,
M. Z.
Zhu
,
X. P.
Qian
,
J.
Zhang
,
Y. X.
Fu
,
L.
Wu
,
Y.
Zhang
,
W. F.
Chen
.
2007
.
Developmental pathway of CD4+CD8- medullary thymocytes during mouse ontogeny and its defect in Aire-/- mice.
Proc. Natl. Acad. Sci. USA.
104
:
18175
18180
.
50.
Stephen
T. L.
,
B. S.
Wilson
,
T. M.
Laufer
.
2012
.
Subcellular distribution of Lck during CD4 T-cell maturation in the thymic medulla regulates the T-cell activation threshold.
Proc. Natl. Acad. Sci. USA.
109
:
7415
7420
.
51.
Hinterberger
M.
,
G.
Wirnsberger
,
L.
Klein
.
2011
.
B7/CD28 in central tolerance: costimulation promotes maturation of regulatory T cell precursors and prevents their clonal deletion.
Front. Immunol.
2
:
30
.
52.
Klein
L.
,
B.
Kyewski
,
P. M.
Allen
,
K. A.
Hogquist
.
2014
.
Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see).
Nat. Rev. Immunol.
14
:
377
391
.
53.
Boursalian
T. E.
,
J.
Golob
,
D. M.
Soper
,
C. J.
Cooper
,
P. J.
Fink
.
2004
.
Continued maturation of thymic emigrants in the periphery.
Nat. Immunol.
5
:
418
425
.
54.
Sansom
S. N.
,
N.
Shikama-Dorn
,
S.
Zhanybekova
,
G.
Nusspaumer
,
I. C.
Macaulay
,
M. E.
Deadman
,
A.
Heger
,
C. P.
Ponting
,
G. A.
Holländer
.
2014
.
Population and single-cell genomics reveal the Aire dependency, relief from Polycomb silencing, and distribution of self-antigen expression in thymic epithelia.
Genome Res.
24
:
1918
1931
.
55.
Ferreirinha
P.
,
C.
Ribeiro
,
J.
Morimoto
,
J. J. M.
Landry
,
M.
Matsumoto
,
C.
Meireles
,
A. J.
White
,
I.
Ohigashi
,
L.
Araújo
,
V.
Benes
, et al
2021
.
A novel method to identify Post-Aire stages of medullary thymic epithelial cell differentiation.
Eur. J. Immunol.
51
:
311
318
.
56.
Bornstein
C.
,
S.
Nevo
,
A.
Giladi
,
N.
Kadouri
,
M.
Pouzolles
,
F.
Gerbe
,
E.
David
,
A.
Machado
,
A.
Chuprin
,
B.
Tóth
, et al
2018
.
Single-cell mapping of the thymic stroma identifies IL-25-producing tuft epithelial cells.
Nature.
559
:
622
626
.
57.
Dumont-Lagacé
M.
,
S.
Brochu
,
C.
St-Pierre
,
C.
Perreault
.
2014
.
Adult thymic epithelium contains nonsenescent label-retaining cells.
J. Immunol.
192
:
2219
2226
.
58.
St-Pierre
C.
,
S.
Brochu
,
J. R.
Vanegas
,
M.
Dumont-Lagacé
,
S.
Lemieux
,
C.
Perreault
.
2013
.
Transcriptome sequencing of neonatal thymic epithelial cells.
Sci. Rep.
3
:
1860
.
59.
Zhang
Q.
,
Z.
Liang
,
J.
Zhang
,
T.
Lei
,
X.
Dong
,
H.
Su
,
Y.
Chen
,
Z.
Zhang
,
L.
Tan
,
Y.
Zhao
.
2021
.
Sirt6 regulates the development of medullary thymic epithelial cells and contributes to the establishment of central immune tolerance.
Front. Cell Dev. Biol.
9
:
655552
.
60.
Liang
Z.
,
Q.
Zhang
,
X.
Dong
,
Z.
Zhang
,
H.
Wang
,
J.
Zhang
,
Y.
Zhao
.
2021
.
mTORC2 negatively controls the maturation process of medullary thymic epithelial cells by inhibiting the LTβR/RANK-NF-κB axis.
J. Cell. Physiol.
236
:
4725
4737
.
61.
Guicciardi
M. E.
,
M.
Leist
,
G. J.
Gores
.
2004
.
Lysosomes in cell death.
Oncogene.
23
:
2881
2890
.
62.
Boya
P.
,
K.
Andreau
,
D.
Poncet
,
N.
Zamzami
,
J. L.
Perfettini
,
D.
Metivier
,
D. M.
Ojcius
,
M.
Jäättelä
,
G.
Kroemer
.
2003
.
Lysosomal membrane permeabilization induces cell death in a mitochondrion-dependent fashion.
J. Exp. Med.
197
:
1323
1334
.
63.
Appelqvist
H.
,
P.
Wäster
,
K.
Kågedal
,
K.
Öllinger
.
2013
.
The lysosome: from waste bag to potential therapeutic target.
J. Mol. Cell Biol.
5
:
214
226
.
64.
Rossi
A.
,
Q.
Deveraux
,
B.
Turk
,
A.
Sali
.
2004
.
Comprehensive search for cysteine cathepsins in the human genome.
Biol. Chem.
385
:
363
372
.
65.
Appelqvist
H.
,
A. C.
Johansson
,
E.
Linderoth
,
U.
Johansson
,
B.
Antonsson
,
R.
Steinfeld
,
K.
Kågedal
,
K.
Ollinger
.
2012
.
Lysosome-mediated apoptosis is associated with cathepsin D-specific processing of bid at Phe24, Trp48, and Phe183.
Ann. Clin. Lab. Sci.
42
:
231
242
.
66.
Chwieralski
C. E.
,
T.
Welte
,
F.
Bühling
.
2006
.
Cathepsin-regulated apoptosis.
Apoptosis.
11
:
143
149
.
67.
Vander Heiden
M. G.
,
N. S.
Chandel
,
E. K.
Williamson
,
P. T.
Schumacker
,
C. B.
Thompson
.
1997
.
Bcl-xL regulates the membrane potential and volume homeostasis of mitochondria.
Cell
91
:
627
637
.
68.
Li
H.
,
H.
Zhu
,
C. J.
Xu
,
J.
Yuan
.
1998
.
Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis.
Cell
94
:
491
501
.
69.
Haljasorg
U.
,
J.
Dooley
,
M.
Laan
,
K.
Kisand
,
R.
Bichele
,
A.
Liston
,
P.
Peterson
.
2017
.
Irf4 expression in thymic epithelium is critical for thymic regulatory T cell homeostasis.
J. Immunol.
198
:
1952
1960
.
70.
Rodrigues
P. M.
,
P.
Peterson
,
N. L.
Alves
.
2018
.
Setting up the perimeter of tolerance: insights into mTEC physiology.
Trends Immunol.
39
:
2
5
.
71.
Rodrigues
P. M.
,
A. R.
Ribeiro
,
C.
Perrod
,
J. J. M.
Landry
,
L.
Araújo
,
I.
Pereira-Castro
,
V.
Benes
,
A.
Moreira
,
H.
Xavier-Ferreira
,
C.
Meireles
,
N. L.
Alves
.
2017
.
Thymic epithelial cells require p53 to support their long-term function in thymopoiesis in mice.
Blood.
130
:
478
488
.
72.
Wang
Y.
,
G.
Huang
,
H.
Zeng
,
K.
Yang
,
R. F.
Lamb
,
H.
Chi
.
2013
.
Tuberous sclerosis 1 (Tsc1)-dependent metabolic checkpoint controls development of dendritic cells.
Proc. Natl. Acad. Sci. USA.
110
:
E4894
E4903
.
73.
Settembre
C.
,
R.
Zoncu
,
D. L.
Medina
,
F.
Vetrini
,
S.
Erdin
,
S.
Erdin
,
T.
Huynh
,
M.
Ferron
,
G.
Karsenty
,
M. C.
Vellard
, et al
2012
.
A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB.
EMBO J.
31
:
1095
1108
.
74.
Martina
J. A.
,
Y.
Chen
,
M.
Gucek
,
R.
Puertollano
.
2012
.
MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB.
Autophagy.
8
:
903
914
.
75.
Roczniak-Ferguson
A.
,
C. S.
Petit
,
F.
Froehlich
,
S.
Qian
,
J.
Ky
,
B.
Angarola
,
T. C.
Walther
,
S. M.
Ferguson
.
2012
.
The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis.
Sci. Signal.
5
:
ra42
.
76.
Peña-Llopis
S.
,
S.
Vega-Rubin-de-Celis
,
J. C.
Schwartz
,
N. C.
Wolff
,
T. A.
Tran
,
L.
Zou
,
X. J.
Xie
,
D. R.
Corey
,
J.
Brugarolas
.
2011
.
Regulation of TFEB and V-ATPases by mTORC1.
EMBO J.
30
:
3242
3258
.
77.
Asrani
K.
,
S.
Murali
,
B.
Lam
,
C. H.
Na
,
P.
Phatak
,
A.
Sood
,
H.
Kaur
,
Z.
Khan
,
M.
Noë
,
R. K.
Anchoori
, et al
2019
.
mTORC1 feedback to AKT modulates lysosomal biogenesis through MiT/TFE regulation.
J. Clin. Invest.
129
:
5584
5599
.
78.
Alesi
N.
,
E. W.
Akl
,
D.
Khabibullin
,
H. J.
Liu
,
A. S.
Nidhiry
,
E. R.
Garner
,
H.
Filippakis
,
H. C.
Lam
,
W.
Shi
,
S. R.
Viswanathan
, et al
2021
.
TSC2 regulates lysosome biogenesis via a non-canonical RAGC and TFEB-dependent mechanism.
Nat. Commun.
12
:
4245
.
79.
Cirman
T.
,
K.
Oresić
,
G. D.
Mazovec
,
V.
Turk
,
J. C.
Reed
,
R. M.
Myers
,
G. S.
Salvesen
,
B.
Turk
.
2004
.
Selective disruption of lysosomes in HeLa cells triggers apoptosis mediated by cleavage of Bid by multiple papain-like lysosomal cathepsins.
J. Biol. Chem.
279
:
3578
3587
.
80.
Kreuzaler
P. A.
,
A. D.
Staniszewska
,
W.
Li
,
N.
Omidvar
,
B.
Kedjouar
,
J.
Turkson
,
V.
Poli
,
R. A.
Flavell
,
R. W.
Clarkson
,
C. J.
Watson
.
2011
.
Stat3 controls lysosomal-mediated cell death in vivo.
Nat. Cell Biol.
13
:
303
309
.
81.
Persson
H. L.
,
Z.
Yu
,
O.
Tirosh
,
J. W.
Eaton
,
U. T.
Brunk
.
2003
.
Prevention of oxidant-induced cell death by lysosomotropic iron chelators.
Free Radic. Biol. Med.
34
:
1295
1305
.
82.
Ono
K.
,
S. O.
Kim
,
J.
Han
.
2003
.
Susceptibility of lysosomes to rupture is a determinant for plasma membrane disruption in tumor necrosis factor alpha-induced cell death.
Mol. Cell. Biol.
23
:
665
676
.

The authors have no financial conflicts of interest.

Supplementary data