LAMP3 induces apoptosis and autoantigen release in Sjögren's syndrome patients

Tsutomu Tanaka, Blake M Warner, Toshio Odani, Youngmi Ji, Ying-Qian Mo, Hiroyuki Nakamura, Shyh-Ing Jang, Hongen Yin, Drew G Michael, Noriyuki Hirata, Futoshi Suizu, Satoko Ishigaki, Fabiola Reis Oliveira, Ana Carolina F Motta, Alfredo Ribeiro-Silva, Eduardo M Rocha, Tatsuya Atsumi, Masayuki Noguchi, John A Chiorini, Tsutomu Tanaka, Blake M Warner, Toshio Odani, Youngmi Ji, Ying-Qian Mo, Hiroyuki Nakamura, Shyh-Ing Jang, Hongen Yin, Drew G Michael, Noriyuki Hirata, Futoshi Suizu, Satoko Ishigaki, Fabiola Reis Oliveira, Ana Carolina F Motta, Alfredo Ribeiro-Silva, Eduardo M Rocha, Tatsuya Atsumi, Masayuki Noguchi, John A Chiorini

Abstract

Primary Sjögren's syndrome (pSS) is a complex autoimmune disease characterized by dysfunction of secretory epithelia with only palliative therapy. Patients present with a constellation of symptoms, and the diversity of symptomatic presentation has made it difficult to understand the underlying disease mechanisms. In this study, aggregation of unbiased transcriptome profiling data sets of minor salivary gland biopsies from controls and Sjögren's syndrome patients identified increased expression of lysosome-associated membrane protein 3 (LAMP3/CD208/DC-LAMP) in a subset of Sjögren's syndrome cases. Stratification of patients based on their clinical characteristics suggested an association between increased LAMP3 expression and the presence of serum autoantibodies including anti-Ro/SSA, anti-La/SSB, anti-nuclear antibodies. In vitro studies demonstrated that LAMP3 expression induces epithelial cell dysfunction leading to apoptosis. Interestingly, LAMP3 expression resulted in the accumulation and release of intracellular TRIM21 (one component of SSA), La (SSB), and α-fodrin protein, common autoantigens in Sjögren's syndrome, via extracellular vesicles in an apoptosis-independent mechanism. This study defines a clear role for LAMP3 in the initiation of apoptosis and an independent pathway for the extracellular release of known autoantigens leading to the formation of autoantibodies associated with this disease.ClinicalTrials.gov Identifier: NCT00001196, NCT00001390, NCT02327884.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
LAMP3 expression is increased in Sjögren’s syndrome patients, and increased expression is associated with serum autoantibodies. (A) LAMP3 mRNA expression levels in controls (healthy volunteers; open circle), non-SS (black dot) and primary SS (red dot) patients (Mean ± SD). (B) LAMP3 mRNA expression levels in controls, non-SS patients and SS patients, with or without presence of serum anti-SSA antibody (Mean ± SD). (C) Representative confocal immunofluorescent (IF) images (×40 magnification) of LAMP3 protein expression in (left image) a patient who does not meet SS criteria (non-SS), (middle image) a patient who meets criteria for primary SS (pSS) with median LAMP3 expression, (right image) and a patient (pSS#) with a characteristic lymphocytic focus adjacent to normal salivary gland epithelial tissue (ducts and acini). LAMP3 positive aggregates are present in acinar (open triangle) and ductal epithelia (arrow), the luminal debri (asterisks), and the lymphocytic foci (dashed outline). (D) IF intensity for LAMP3 protein expression in MSG biopsies from non-SS and pSS patients (Mean ± SD). (E) Differential expression of LAMP3 in human primary salivary gland epithelial cells (pSGECs) established from 7 SS patients and 8 healthy volunteers was analyzed by using RT-qPCR (open circle: SSA and SSB negative, closed circle: SSA and/or SSB positive). *P < 0.05, ANoVA; ††P < 0.01, †††P < 0.001, unpaired Student’s t-test.
Figure 2
Figure 2
LAMP3 overexpression increases TRIM21 expression and number and size of nuclear aggregates. Representative confocal immunofluorescent (IF) images (60X objective) of (A) TRIM21/SSA and (C) SSB protein expression in control HSG cells (empty) and LAMP3-overexpressing (OE) HSG cells. Nuclear aggregate size, number of nuclear aggregates per nuclei, and total IF intensity for (B) TRIM21/SSA and (D) SSB in control and LAMP3 OE cells. TRIM21/SSA aggregate size: N = 144 cells (control) and 558 cells (LAMP3-OE); TRIM21/SSA aggregate nember per nuclei: N = 149 cells (control) and 84 cells (LAMP3-OE); TRIM21/SSA total IF intensity: N = 177 cells (control) and 146 cells (LAMP3-OE). SSB aggregate size: N = 359 cells (control) and 430 cells (LAMP3-OE); SSB aggregate nember per nuclei: N = 130 cells (control) and 117 cells (LAMP3-OE); SSB total IF intensity:N = 146 cells (control) and 154 cells (LAMP3-OE). ***P < 0.001, ANoVA. Values shown are mean ± SD.
Figure 3
Figure 3
LAMP3 inhibits cell growth and induces caspase-dependent apoptosis. (A) HSG and (B) A253 cells were transfected with 1.5 μg pME18S-empty or pME18S-LAMP3 plasmid, and then 2 × 105 cells of each cells were re-plated 24 h post-transfection. The number of cells was counted 96 h after re-plating with Countess Automated Cell Counter (N = 3 independent experiment and 2 technical replicates of each experiment). (C,D) HSG cells and (E,F) A253 cells were transfected with different concentrations of pME18S-LAMP3 plasmid or a pME18S-empty plasmid as control. Seventy-two hours post-transfection, apoptotic cells were counted by using flow cytometry with APC Annexin V/7-AAD. Difference from control cells is shown (N = 4). (G,H) Number of apoptotic cells in control and LAMP3-overexpressing (OE) HSG cell cultures 24, 48 and 72 h after transfection, assessed by flow cytometry using APC Annexin V/7-AAD. Difference from control cells is shown (N = 3). (I,J) Number of apoptotic cells in GFP+ control, LAMP3-OE and LAMP1-OE HSG cell cultures 48 h after transfection, assessed by flow cytometry using APC Annexin V/7-AAD. Difference from control cells is shown (N = 4). (K,L) Number of apoptotic cells in GFP+ control, LAMP3-OE and LAMP1-OE A253 cell cultures 48 h after transfection, assessed by flow cytometry using APC Annexin V/7-AAD. Difference from control cells is shown (N = 4). (M,N) Control and LAMP3-OE HSG cells were incubated with or without 20 μM Z-VAD-FMK (Z-VAD) for 20 h. Extent of apoptosis was determined by flow cytometry using APC Annexin V/7-AAD. Difference in rate of Annexin V+ cells in LAMP3-OE cell culture treated with or without 20 μM Z-VAD from that in control cell culture is shown (N = 4). *P < 0.05, **P < 0.01, ***P < 0.001, unpaired Student’s t-test. Values shown are mean ± SD.
Figure 3
Figure 3
LAMP3 inhibits cell growth and induces caspase-dependent apoptosis. (A) HSG and (B) A253 cells were transfected with 1.5 μg pME18S-empty or pME18S-LAMP3 plasmid, and then 2 × 105 cells of each cells were re-plated 24 h post-transfection. The number of cells was counted 96 h after re-plating with Countess Automated Cell Counter (N = 3 independent experiment and 2 technical replicates of each experiment). (C,D) HSG cells and (E,F) A253 cells were transfected with different concentrations of pME18S-LAMP3 plasmid or a pME18S-empty plasmid as control. Seventy-two hours post-transfection, apoptotic cells were counted by using flow cytometry with APC Annexin V/7-AAD. Difference from control cells is shown (N = 4). (G,H) Number of apoptotic cells in control and LAMP3-overexpressing (OE) HSG cell cultures 24, 48 and 72 h after transfection, assessed by flow cytometry using APC Annexin V/7-AAD. Difference from control cells is shown (N = 3). (I,J) Number of apoptotic cells in GFP+ control, LAMP3-OE and LAMP1-OE HSG cell cultures 48 h after transfection, assessed by flow cytometry using APC Annexin V/7-AAD. Difference from control cells is shown (N = 4). (K,L) Number of apoptotic cells in GFP+ control, LAMP3-OE and LAMP1-OE A253 cell cultures 48 h after transfection, assessed by flow cytometry using APC Annexin V/7-AAD. Difference from control cells is shown (N = 4). (M,N) Control and LAMP3-OE HSG cells were incubated with or without 20 μM Z-VAD-FMK (Z-VAD) for 20 h. Extent of apoptosis was determined by flow cytometry using APC Annexin V/7-AAD. Difference in rate of Annexin V+ cells in LAMP3-OE cell culture treated with or without 20 μM Z-VAD from that in control cell culture is shown (N = 4). *P < 0.05, **P < 0.01, ***P < 0.001, unpaired Student’s t-test. Values shown are mean ± SD.
Figure 4
Figure 4
LAMP3-induced activation of caspase-3 is independent of endoplasmic reticulum stress. Caspase-3 (CASP3) mRNA expression levels were measured by qRT-PCR in control and LAMP3-overexpressing (OE) (A) HSG and (D) A253 cells. Western blot analysis was performed to measure (B,E) caspase-3 protein, (C,F) ATF4, spliced XBP1 (XBP1s) and unspliced XBP1 (XBP1u) protein levels in HSG and A253 control and LAMP3-OE cells, respectively. All ratios are relative value compared with control. Uncropped images are provided in Supplementary Figure S4.
Figure 5
Figure 5
LAMP3-induced accumulation of autoantigens and release via extracellular vesicles. (A) Images were collected by immunofluorescent microscopy at 100X magnification. LAMP3-overexpressing (OE) cells show a membranous and vesicular pattern of LAMP3 expression. Some of the vesicles appear just below or budding from the plasma membrane and colocalize with TRIM21/SSA and SSB (open triangles). TRIM21/SSA, SSB, α-fodrin and cleaved (cl.) α-fodrin protein levels in control and LAMP3-OE cells. Rows 2 and 4 are further enlarged images of specific cells shown in row 1 and 3. The specific cells in the merged image are boxed for clarity. Indicated protein expression in (B) HSG and (C) A253 cells, as determined by Western blotting. (D) Protein concentration in mixture of extracellular vesicles (EVs) isolated from control and LAMP3-OE HSG cells. Data are presented as relative change in expression compared with control. Western blotting analysis of TRIM21/SSA and SSB in EVs isolated from control and LAMP3-OE (E) HSG and (F) A253 cells. (G) Western blotting of α-fodrin and cl. α-fodrin in EVs from control and LAMP3-OE HSG cells. Protein levels were normalized to α-tubulin or flotillin-1 level. Uncropped images are provided in Supplementary Figure S5. ***p < 0.001, unpaired Student’s t-test.
Figure 5
Figure 5
LAMP3-induced accumulation of autoantigens and release via extracellular vesicles. (A) Images were collected by immunofluorescent microscopy at 100X magnification. LAMP3-overexpressing (OE) cells show a membranous and vesicular pattern of LAMP3 expression. Some of the vesicles appear just below or budding from the plasma membrane and colocalize with TRIM21/SSA and SSB (open triangles). TRIM21/SSA, SSB, α-fodrin and cleaved (cl.) α-fodrin protein levels in control and LAMP3-OE cells. Rows 2 and 4 are further enlarged images of specific cells shown in row 1 and 3. The specific cells in the merged image are boxed for clarity. Indicated protein expression in (B) HSG and (C) A253 cells, as determined by Western blotting. (D) Protein concentration in mixture of extracellular vesicles (EVs) isolated from control and LAMP3-OE HSG cells. Data are presented as relative change in expression compared with control. Western blotting analysis of TRIM21/SSA and SSB in EVs isolated from control and LAMP3-OE (E) HSG and (F) A253 cells. (G) Western blotting of α-fodrin and cl. α-fodrin in EVs from control and LAMP3-OE HSG cells. Protein levels were normalized to α-tubulin or flotillin-1 level. Uncropped images are provided in Supplementary Figure S5. ***p < 0.001, unpaired Student’s t-test.
Figure 6
Figure 6
LAMP3-induced autoantigens release was independent of apoptosis. (A) Protein concentration in mixture of extracellular vesicles (EVs) isolated from control and LAMP3-OE HSG cells treated with or without Z-VAD. Data are presented as relative change in expression compared with control cells. (B) Western blotting analysis of TRIM21/SSA, SSB and α-fodrin in EVs isolated from HSG control and LAMP3-OE cells treated with or withour Z-VAD. Protein levels of (C) TRIM21/SSA, (D) SSB and (E) α-fodrin were normalized to flotillin-1 level, and the value was plotted. Uncropped images are provided in Supplementary Figure S6. **p < 0.01, ***p < 0.001, unpaired Student’s t-test.

References

    1. Robert IF. Sjögren's syndrome. Lancet. 2005;366(9482):321–331. doi: 10.1016/S0140-6736(05)66990-5.
    1. Long S, Lakshmanan S. Autoantibodies, detection methods and panels for diagnosis of Sjögren's syndrome. Clin, Immunol. 2017;182:24–29. doi: 10.1016/j.clim.2017.03.017.
    1. Xiaomei L, et al. Clinical and laboratory profiles of primary Sjogren's syndrome in a Chinese population: A retrospective analysis of 315 patients. Int. J. Rheum Dis. 2015;18(4):439–446. doi: 10.1111/1756-185X.12583.
    1. Vitali C, et al. Classification criteria for Sjogren’s syndrome: A revised version of the European criteria proposed by the American–European Consensus Group. Ann. Rheum Dis. 2002;61:554–558. doi: 10.1136/ard.61.6.554.
    1. Theander E. Prediction of Sjögren's syndrome years before diagnosis and identification of patients with early onset and severe disease course by autoantibody profiling. Arthritis Rheumatol. 2015;67(9):2427–2436. doi: 10.1002/art.39214.
    1. Jonsson R, Theander E, Sjöström B, Brokstad K, Henriksson G. Autoantibodies present before symptom onset in primary Sjögren syndrome. JAMA. 2013;310(17):1854–1855. doi: 10.1001/jama.2013.278448.
    1. Ramos-Casals M, Font J. Primary Sjögren's syndrome: Current and emergent aetiopathogenic concepts. Rheumatol. (Oxford) 2005;44(11):1354–1367. doi: 10.1093/rheumatology/keh714.
    1. Manganelli P, Fietta P. Apoptosis and Sjögren syndrome. Semin Arthritis Rheum. 2003;33(1):49–65. doi: 10.1053/sarh.2003.50019.
    1. Baban B, Liu JY, Abdelsayed R, Mozaffari MS. Reciprocal relation between GADD153 and Del-1 in regulation of salivary gland inflammation in Sjögren syndrome. Exp. Mol. Pathol. 2013;95(3):288–297. doi: 10.1016/j.yexmp.2013.09.002.
    1. Brady OA, Martina JA, Puertollano R. Emerging roles for TFEB in the immune response and inflammation. Autophagy. 2018;14(2):181–189. doi: 10.1080/15548627.2017.1313943.
    1. Dash S, Aydin Y, Moroz K. Chaperone-mediated autophagy in the liver: Good or bad? Cells. 2019;8(11):1308. doi: 10.3390/cells8111308.
    1. Zhou Z, et al. Lysosome-associated membrane glycoprotein 3 is involved in influenza A virus replication in human lung epithelial (A549) cells. Virol J. 2011;8:384. doi: 10.1186/1743-422X-8-384.
    1. Salaun B, et al. CD208/dendritic cell-lysosomal associated membrane protein is a marker of normal and transformed type II pneumocytes. Am. J. Pathol. 2004;164(3):861–871. doi: 10.1016/S0002-9440(10)63174-4.
    1. Berger C, et al. Rapid generation of maturationally synchronized human dendritic cells: Contribution to the clinical efficacy of extracorporeal photochemotherapy. Blood. 2010;116(23):4838–4847. doi: 10.1182/blood-2009-11-256040.
    1. Barois N, de Saint-Vis B, Lebecque S, Geuze HJ, Kleijmeer MJ. MHC class II compartments in human dendritic cells undergo profound structural changes upon activation. Traffic. 2002;3(12):894–905. doi: 10.1034/j.1600-0854.2002.31205.x.
    1. Arruda LB, et al. Dendritic cell-lysosomal-associated membrane protein (LAMP) and LAMP-1-HIV-1 gag chimeras have distinct cellular trafficking pathways and prime T and B cell responses to a diverse repertoire of epitopes. J. Immunol. 2006;177(4):2265–2275. doi: 10.4049/jimmunol.177.4.2265.
    1. Dominguez-Bautista JA, et al. Loss of lysosome-associated membrane protein 3 (LAMP3) enhances cellular vulnerability against proteasomal inhibition. Eur J Cell Biol. 2015;94(3–4):148–161. doi: 10.1016/j.ejcb.2015.01.003.
    1. Nagelkerke A, et al. LAMP3 is involved in tamoxifen resistance in breast cancer cells through the modulation of autophagy. Endocr. Relat. Cancer. 2014;21(1):101–112. doi: 10.1530/ERC-13-0183.
    1. Serrano-Puebla A, Boya P. Lysosomal membrane permeabilization as a cell death mechanism in cancer cells. Biochem. Soc. Trans. 2018;46(2):207–215. doi: 10.1042/BST20170130.
    1. Lapointe J, et al. Gene expression profiling identifies clinically relevant subtypes of prostate cancer. Proc. Natl. Acad. Sci. USA. 2004;101(3):811–816. doi: 10.1073/pnas.0304146101.
    1. Leavey K, Bainbridge SA, Cox BJ. Large scale aggregate microarray analysis reveals three distinct molecular subclasses of human preeclampsia. PLoS ONE. 2015;10(2):e0116508. doi: 10.1371/journal.pone.0116508.
    1. Yin H, et al. Association of bone morphogenetic protein 6 with exocrine gland dysfunction in patients with Sjögren’s syndrome and in mice. Arthritis Rheum. 2013;65(12):3228–3238. doi: 10.1002/art.38123.
    1. Greenwell-Wild T, et al. Chitinases in the salivary glands and circulation of patients with Sjögren's syndrome: Macrophage harbingers of disease severity. Arthritis Rheum. 2011;63(10):3103–3115. doi: 10.1002/art.30465.
    1. Tsuboi H, et al. DNA microarray analysis of labial salivary glands in IgG4-related disease: Comparison with Sjögren's syndrome. Arthritis Rheumatol. 2014;66(10):2892–2899. doi: 10.1002/art.38748.
    1. Takahashi H, et al. cDNA microarray analysis identifies NR4A2 as a novel molecule involved in the pathogenesis of Sjögren's syndrome. Clin. Exp. Immunol. 2017;190(1):96–109. doi: 10.1111/cei.13000.
    1. Burbelo PD, et al. Sensitive and robust luminescent profiling of anti-La and other autoantibodies in Sjogren's syndrome. Autoimmunity. 2009;42(6):515–524. doi: 10.1080/08916930902911738.
    1. Kyriakidis NC, et al. Toll-like receptor 3 stimulation promotes Ro52/TRIM21 synthesis and nuclear redistribution in salivary gland epithelial cells, partially via type I interferon pathway. Clin. Exp. Immunol. 2014;178(3):548–560. doi: 10.1111/cei.12432.
    1. Clarke P, Tyler KL. Apoptosis in animal models of virus-induced disease. Nat. Rev. Microbiol. 2009;7(2):144–155. doi: 10.1038/nrmicro2071.
    1. Li C, et al. Follistatin-like protein 5 inhibits hepatocellular carcinoma progression by inducing caspase-dependent apoptosis and regulating Bcl-2 family proteins. J. Cell. Mol. Med. 2018;22(12):6190–6201. doi: 10.1111/jcmm.13906.
    1. Slee EA, et al. Benzyloxycarbonyl-Val-Ala-Asp (OMe) fluoromethylketone (Z-VAD.FMK) inhibits apoptosis by blocking the processing of CPP32. Biochem. J. 1996;315(Pt 1):21–24. doi: 10.1042/bj3150021.
    1. Egger L, Madden DT, Rhême C, Rao RV, Bredesen DE. Endoplasmic reticulum stress-induced cell death mediated by the proteasome. Cell Death Differ. 2007;14(6):1172–1180. doi: 10.1038/sj.cdd.4402125.
    1. Martín-Pérez R, et al. Activated ERBB2/HER2 licenses sensitivity to apoptosis upon endoplasmic reticulum stress through a PERK-dependent pathway. Cancer Res. 2014;74(6):1766–1777. doi: 10.1158/0008-5472.CAN-13-1747.
    1. Kapsogeorgou EK, Abu-Helu RF, Moutsopoulos HM, Manoussakis MN. Salivary gland epithelial cell exosomes: A source of autoantigenic ribonucleoproteins. Arthritis Rheum. 2005;52(5):1517–1521. doi: 10.1002/art.21005.
    1. Pisetsky DS, Gauley J, Ullal AJ. Microparticles as a source of extracellular DNA. Immunol. Res. 2011;49(1–3):227–234. doi: 10.1007/s12026-010-8184-8.
    1. Cloutier N, et al. The exposure of autoantigens by microparticles underlies the formation of potent inflammatory components: The microparticle-associated immune complexes. EMBO Mol. Med. 2013;5(2):235–249. doi: 10.1002/emmm.201201846.
    1. Hasilo CP, et al. Presence of diabetes autoantigens in extracellular vesicles derived from human islets. Sci. Rep. 2017;7(1):5000. doi: 10.1038/s41598-017-04977-y.
    1. McEwan WA, et al. Intracellular antibody-bound pathogens stimulate immune signaling via the Fc receptor TRIM21. Nat. Immunol. 2013;14(4):327–336. doi: 10.1038/ni.2548.
    1. Rakebrandt N, Lentes S, Neumann H, James LC, Neumann-Staubitz P. Antibody- and TRIM21-dependent intracellular restriction of Salmonella enterica. Pathog. Dis. 2014;72(2):131–137.
    1. Miranda-Carús ME, et al. Induction of antibodies reactive with SSA/Ro-SSB/La and development of congenital heart block in a murine model. J. Immunol. 1998;161(11):5886–5892.
    1. Szczerba BM, et al. Interaction between innate immunity and Ro52-induced antibody causes Sjögren's syndrome-like disorder in mice. Ann. Rheum. Dis. 2016;75(3):617–622. doi: 10.1136/annrheumdis-2014-206297.
    1. Haneji N, et al. Identification of alpha-fodrin as a candidate autoantigen in primary Sjögren's syndrome. Science. 1997;276(5312):604–607. doi: 10.1126/science.276.5312.604.
    1. Sroka M, Bagavant H, Biswas I, Ballard A, Deshmukh US. Immune response against the coiled coil domain of Sjögren's syndrome associated autoantigen Ro52 induces salivary gland dysfunction. Clin. Exp. Rheumatol. 2018;36 Suppl 112(3):41–46.
    1. Reed JH, Jackson MW, Gordon TP. B cell apotopes of the 60-kDa Ro/SSA and La/SSB autoantigens. J. Autoimmun. 2008;31(3):263–267. doi: 10.1016/j.jaut.2008.04.008.
    1. Duhlin A, et al. Selective memory to apoptotic cell-derived self-antigens with implications for systemic lupus erythematosus development. J. Immunol. 2016;197(7):2618–2626. doi: 10.4049/jimmunol.1401129.
    1. Navid F, Colbert RA. Causes and consequences of endoplasmic reticulum stress in rheumatic disease. Nat. Rev. Rheumatol. 2017;13(1):25–40. doi: 10.1038/nrrheum.2016.192.
    1. Smith JA. Regulation of cytokine production by the unfolded protein response; implications for infection and autoimmunity. Front. Immunol. 2018;9:422. doi: 10.3389/fimmu.2018.00422.
    1. Barrera MJ, et al. Pro-inflammatory cytokines enhance ERAD and ATF6α pathway activity in salivary glands of Sjögren's syndrome patients. J. Autoimmun. 2016;75:68–81. doi: 10.1016/j.jaut.2016.07.006.
    1. Mujcic H, et al. Hypoxic activation of the unfolded protein response (UPR) induces expression of the metastasis-associated gene LAMP3. Radiother. Oncol. 2009;92(3):450–459. doi: 10.1016/j.radonc.2009.08.017.
    1. Kroemer G, et al. Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009;16(1):3–11. doi: 10.1038/cdd.2008.150.
    1. Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy-inflammation-cell death axis in organismal aging. Science. 2011;333(6046):1109–1112. doi: 10.1126/science.1201940.
    1. Noguchi M, Hirata N, Suizu F. The links between AKT and two intracellular proteolytic cascades: Ubiquitination and autophagy. Biochim. Biophys. Acta. 2014;1846(2):342–352.
    1. Barrett T, et al. NCBI GEO: Archive for functional genomics data sets—Update. Nucleic Acids Res. 2013;41:D991–D995. doi: 10.1093/nar/gks1193.
    1. Azuma M, Tamatani T, Kasai Y, Sato M. Immortalization of normal human salivary gland cells with duct-, myoepithelial-, acinar-, or squamous phenotype by transfection with SV40 ori-mutant deoxyribonucleic acid. Lab. Invest. 1993;69(1):24–42.
    1. Laine J, Künstle G, Obata T, Sha M, Noguchi M. The protooncogene TCL1 is an Akt kinase coactivator. Mol. Cell. 2000;6(2):395–407. doi: 10.1016/S1097-2765(00)00039-3.
    1. Jang SI, Ong HL, Gallo A, Liu X, Illei G, Alevizos I. Establishment of functional acinar-like cultures from human salivary glands. J. Dent Res. 2015;94(2):304–311. doi: 10.1177/0022034514559251.

Source: PubMed

3
Abonneren