Introduction
Tumor necrosis factor (TNF)
2The abbreviations used are:
TNF
tumor necrosis factor
DD
death domain
MAPK
mitogen-activated protein kinase
DISC
death-inducing signaling complex
shRNA
short hairpin RNA
KD
knockdown
EV
empty vector
OE
overexpression
IKK
IκB kinase
JNK
c-Jun N-terminal kinase
ERK
extracellular signal-regulated kinase
Par
parental
PMA
phorbol 12-myristate 13-acetate
TBS
Tris-buffered saline
CHX
cycloheximide
HRP
horseradish peroxidase
SNA
Sambucus nigra agglutinin
TACE
tumor necrosis α–converting enzyme
TRAIL
TNF-related apoptosis-inducing ligand
FADD
Fas-associated death domain protein
TRADD
tumor necrosis factor receptor type 1-associated death domain protein.
is the prototypical member of the TNF superfamily of cytokines. Although it was first recognized for its anti-tumor activity, TNF has since been identified as a highly pleiotropic cytokine that mediates multiple cellular processes, including inflammation, cell differentiation, cell survival and proliferation, and apoptosis. TNF can bind and activate two receptors: tumor necrosis factor receptor 1 (TNFR1) and tumor necrosis factor receptor 2 (TNFR2), the latter of which is chiefly expressed in hematopoietic cells (
1- Ham B.
- Fernandez M.C.
- D'Costa Z.
- Brodt P.
The diverse roles of the TNF axis in cancer progression and metastasis.
). TNFR1, which is responsible for the majority of TNF-induced events (
2- Barbara J.A.
- Smith W.B.
- Gamble J.R.
- Van Ostade X.
- Vandenabeele P.
- Tavernier J.
- Fiers W.
- Vadas M.A.
- Lopez A.F.
Dissociation of TNF-α cytotoxic and proinflammatory activities by p55 receptor- and p75 receptor-selective TNF-α mutants.
,
3- Puimège L.
- Libert C.
- Van Hauwermeiren F.
Regulation and dysregulation of tumor necrosis factor receptor-1.
), is a ubiquitously expressed member of the death receptor subgroup of the TNF receptor superfamily. Death receptors, which also include Fas and the TRAIL receptors DR4 and DR5, are characterized by a cytoplasmic “death domain” (DD), a conserved sequence vital for apoptosis induction (
4- Locksley R.M.
- Killeen N.
- Lenardo M.J.
The TNF and TNF receptor superfamilies: integrating mammalian biology.
). TNFR2 lacks a DD and therefore cannot induce cell death.
When initially isolated in 1975, TNF was described as an “endotoxin” capable of inducing necrosis of tumors (
5- Carswell E.A.
- Old L.J.
- Kassel R.L.
- Green S.
- Fiore N.
- Williamson B.
An endotoxin-induced serum factor that causes necrosis of tumors.
). However, over the next two decades, the promise that TNF could serve as an antitumor therapy faded, as research revealed a contradictory role for TNF as an inducer of cell survival (
1- Ham B.
- Fernandez M.C.
- D'Costa Z.
- Brodt P.
The diverse roles of the TNF axis in cancer progression and metastasis.
). In recent years, intensive investigation has centered on defining the mechanisms by which the TNF/TNFR1 signaling axis pivots between cell survival and cell death. The TNFR1 receptor has a complex mechanism of regulation mediated by diverse processes, including receptor oligomerization, lipid raft recruitment, endocytosis, and TNFR1 ubiquitination and shedding (
6- Ali M.
- Fritsch J.
- Zigdon H.
- Pewzner-Jung Y.
- Schütze S.
- Futerman A.H.
Altering the sphingolipid acyl chain composition prevents LPS/GLN-mediated hepatic failure in mice by disrupting TNFR1 internalization.
7- Chhibber-Goel J.
- Coleman-Vaughan C.
- Agrawal V.
- Sawhney N.
- Hickey E.
- Powell J.C.
- McCarthy J.V.
γ-Secretase activity is required for regulated intramembrane proteolysis of tumor necrosis factor (TNF) receptor 1 and TNF-mediated pro-apoptotic signaling.
,
8- Fritsch J.
- Stephan M.
- Tchikov V.
- Winoto-Morbach S.
- Gubkina S.
- Kabelitz D.
- Schütze S.
Cell fate decisions regulated by K63 ubiquitination of tumor necrosis factor receptor 1.
,
9- Legler D.F.
- Micheau O.
- Doucey M.A.
- Tschopp J.
- Bron C.
Recruitment of TNF receptor 1 to lipid rafts is essential for TNFα-mediated NF-κB activation.
10- Schneider-Brachert W.
- Tchikov V.
- Neumeyer J.
- Jakob M.
- Winoto-Morbach S.
- Held-Feindt J.
- Heinrich M.
- Merkel O.
- Ehrenschwender M.
- Adam D.
- Mentlein R.
- Kabelitz D.
- Schütze S.
Compartmentalization of TNF receptor 1 signaling: internalized TNF receptosomes as death signaling vesicles.
). The events downstream of TNFR1 activation are directed by two distinct signaling complexes, one associated with survival (complex I) and the other with apoptosis (complex II). Complex I, composed of TRADD, TRAF2, RIP-1, and cIAP1/2, initiates and propagates survival signaling via activation of the NF-κB and MAPK pathways (
11Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes.
,
12- Schütze S.
- Tchikov V.
- Schneider-Brachert W.
Regulation of TNFR1 and CD95 signalling by receptor compartmentalization.
). Conversely, apoptotic signaling is initiated by TNFR1 internalization into endosomes, followed by formation of complex II, the death-inducing signaling complex (DISC) (
11Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes.
,
12- Schütze S.
- Tchikov V.
- Schneider-Brachert W.
Regulation of TNFR1 and CD95 signalling by receptor compartmentalization.
). The DISC consists of the DD adaptor proteins TRADD and FADD and the apoptosis initiator caspase-8, which is autoproteolytically cleaved into its active form upon recruitment to the DD. Activated caspase-8 then cleaves caspase-3, resulting in the activation of the effector arm of apoptosis.
Adding to the complexity of TNF/TNFR1 signaling, we previously reported that the glycosylation profile on the TNFR1 receptor significantly affects TNFR1 activity. TNFR1 was identified as a substrate for the ST6Gal-I sialyltransferase (
13- Liu Z.
- Swindall A.F.
- Kesterson R.A.
- Schoeb T.R.
- Bullard D.C.
- Bellis S.L.
ST6Gal-I regulates macrophage apoptosis via α2-6 sialylation of the TNFR1 death receptor.
), a Golgi enzyme that adds α2-6–linked sialic acids to
N-glycans on select receptors. The addition of α2-6 sialic acid to TNFR1 was shown to block TNF-induced apoptosis in human monocytic cells as well as monocytes from ST6Gal-I–overexpressing mice (
13- Liu Z.
- Swindall A.F.
- Kesterson R.A.
- Schoeb T.R.
- Bullard D.C.
- Bellis S.L.
ST6Gal-I regulates macrophage apoptosis via α2-6 sialylation of the TNFR1 death receptor.
). However, in this prior investigation, neither the mechanism by which sialylation regulates TNFR1 nor the specific signaling cascades downstream of TNFR1 sialylation were examined, thus limiting a fundamental understanding of the role of glycosylation in regulating TNFR1 function. Furthermore, the contribution of TNFR1 sialylation to cancer cell behavior has not been investigated previously, which is significant in that TNF was originally defined as an anti-tumor effector. Importantly, ST6Gal-I is up-regulated in many diverse cancer types (
14- Büll C.
- Stoel M.A.
- den Brok M.H.
- Adema G.J.
Sialic acids sweeten a tumor's life.
15- Dall'Olio F.
- Malagolini N.
- Trinchera M.
- Chiricolo M.
Sialosignaling: sialyltransferases as engines of self-fueling loops in cancer progression.
,
16Significance of β-galactoside α2,6 sialyltransferase 1 in cancers.
,
17- Harduin-Lepers A.
- Krzewinski-Recchi M.A.
- Colomb F.
- Foulquier F.
- Groux-Degroote S.
- Delannoy P.
Sialyltransferases functions in cancers.
18- Schultz M.J.
- Swindall A.F.
- Bellis S.L.
Regulation of the metastatic cell phenotype by sialylated glycans.
), implicating α2-6 sialylation of TNFR1 as a common feature of cancer cells.
In this study, we define a novel sialylation-dependent mechanism that inhibits TNF-induced TNFR1 internalization and shifts the balance of TNFR1 signaling to favor survival. Using ovarian and pancreatic cancer cell models with ST6Gal-I overexpression or knockdown, we find that ST6Gal-I–mediated TNFR1 sialylation blocks the apoptotic arm of TNFR1 signaling while leaving NF-κB- and Akt-mediated survival signaling intact. These findings highlight the importance of receptor glycosylation in the regulation of a key cell survival pathway critical to numerous physiologic and pathophysiologic processes.
Discussion
Dysregulation of the TNF/TNFR1 signaling axis contributes to a diversity of pathologies, including autoimmune diseases (
4- Locksley R.M.
- Killeen N.
- Lenardo M.J.
The TNF and TNF receptor superfamilies: integrating mammalian biology.
,
27TNF-mediated inflammatory disease.
) and cancer (
1- Ham B.
- Fernandez M.C.
- D'Costa Z.
- Brodt P.
The diverse roles of the TNF axis in cancer progression and metastasis.
,
28- Oshima H.
- Ishikawa T.
- Yoshida G.J.
- Naoi K.
- Maeda Y.
- Naka K.
- Ju X.
- Yamada Y.
- Minamoto T.
- Mukaida N.
- Saya H.
- Oshima M.
TNF-α/TNFR1 signaling promotes gastric tumorigenesis through induction of Noxo1 and Gna14 in tumor cells.
,
29- Charles K.A.
- Kulbe H.
- Soper R.
- Escorcio-Correia M.
- Lawrence T.
- Schultheis A.
- Chakravarty P.
- Thompson R.G.
- Kollias G.
- Smyth J.F.
- Balkwill F.R.
- Hagemann T.
The tumor-promoting actions of TNF-α involve TNFR1 and IL-17 in ovarian cancer in mice and humans.
). Although TNF was originally described as a tumoricidal molecule, it has since emerged that cancer cells are often resistant to TNF-induced cell death, and, in fact, TNF has potent tumor-promoting functions (
1- Ham B.
- Fernandez M.C.
- D'Costa Z.
- Brodt P.
The diverse roles of the TNF axis in cancer progression and metastasis.
,
30Tumor necrosis factor and cancer, buddies or foes?.
). The TNF/TNFR1 pathway fuels tumor growth via multiple conduits, including direct stimulation of tumor cell proliferation (
1- Ham B.
- Fernandez M.C.
- D'Costa Z.
- Brodt P.
The diverse roles of the TNF axis in cancer progression and metastasis.
) and induction of epithelial–mesenchymal transition (
31Tumor necrosis factor-α stimulates the epithelial-to-mesenchymal transition of human colonic organoids.
,
32- Wang H.
- Wang H.S.
- Zhou B.H.
- Li C.L.
- Zhang F.
- Wang X.F.
- Zhang G.
- Bu X.Z.
- Cai S.H.
- Du J.
Epithelial-mesenchymal transition (EMT) induced by TNF-α requires AKT/GSK-3β-mediated stabilization of snail in colorectal cancer.
). In addition, impaired apoptotic signaling by TNFR1 likely contributes to tumor evasion from immune surveillance, given that immune cells are the major source of TNF (
4- Locksley R.M.
- Killeen N.
- Lenardo M.J.
The TNF and TNF receptor superfamilies: integrating mammalian biology.
,
33- Guicciardi M.E.
- Gores G.J.
Life and death by death receptors.
). In view of these factors, a better understanding of the molecular mechanisms that subvert TNFR1 signaling to drive tumorigenesis is greatly needed.
In this investigation, we show that ST6Gal-I–mediated α2-6 sialylation of TNFR1 selectively blocks the apoptotic effects of TNF. Importantly, TNFR1 sialylation does not influence initial TNF-stimulated events such as IκBα destruction, NF-κB nuclear translocation, or rapid activation of Akt, JNK, and ERK. However, striking phenotypic and signaling differences in cells with variant ST6Gal-I expression become apparent upon prolonged TNF treatment (≥6 h). Under chronic TNF stimulation, cells with high ST6Gal-I levels exhibit an increase in survival characteristics, indicated by morphological evidence as well as sustained activation of the survival-associated molecules NF-κB and Akt. Correspondingly, ST6Gal-I–mediated α2-6 sialylation inhibits TNFR1-induced apoptosis, evidenced by diminished activation of caspases 8 and 3. Hence, ST6Gal-I activity shifts the overall balance of TNFR1 signaling toward survival.
We further investigated potential mechanisms by which α2-6 sialylation of TNFR1 might regulate intracellular signaling. Our combined studies point to a sialylation-dependent block in TNFR1 internalization as the principal driver of a signaling switch. This finding is in line with our prior studies of the Fas death receptor, which revealed that α2-6 sialylation of Fas prevents apoptosis by blocking Fas internalization (
34- Swindall A.F.
- Bellis S.L.
Sialylation of the Fas death receptor by ST6Gal-I provides protection against Fas-mediated apoptosis in colon carcinoma cells.
). Significantly, TRAIL-induced apoptosis is unaffected by ST6Gal-I activity, and the TRAIL receptor DR5 has no consensus sequences for
N-glycosylation (
34- Swindall A.F.
- Bellis S.L.
Sialylation of the Fas death receptor by ST6Gal-I provides protection against Fas-mediated apoptosis in colon carcinoma cells.
). The lack of effect on TRAIL-mediated apoptosis indicates that ST6Gal-I activity modulates the function of specific surface receptors and does not fundamentally alter the apoptotic machinery. It is well established that the localization of TNFR1 is critical for downstream signaling (
12- Schütze S.
- Tchikov V.
- Schneider-Brachert W.
Regulation of TNFR1 and CD95 signalling by receptor compartmentalization.
). Many studies have shown that the activation of TNFR1 by TNF at the cell surface initiates NF-κB-mediated survival signaling, whereas TNFR1 internalization into endosomes is essential for robust formation of the DISC and apoptosis induction (
10- Schneider-Brachert W.
- Tchikov V.
- Neumeyer J.
- Jakob M.
- Winoto-Morbach S.
- Held-Feindt J.
- Heinrich M.
- Merkel O.
- Ehrenschwender M.
- Adam D.
- Mentlein R.
- Kabelitz D.
- Schütze S.
Compartmentalization of TNF receptor 1 signaling: internalized TNF receptosomes as death signaling vesicles.
,
35- Schütze S.
- Machleidt T.
- Adam D.
- Schwandner R.
- Wiegmann K.
- Kruse M.L.
- Heinrich M.
- Wickel M.
- Krönke M.
Inhibition of receptor internalization by monodansylcadaverine selectively blocks p55 tumor necrosis factor receptor death domain signaling.
,
36- Schneider-Brachert W.
- Tchikov V.
- Merkel O.
- Jakob M.
- Hallas C.
- Kruse M.L.
- Groitl P.
- Lehn A.
- Hildt E.
- Held-Feindt J.
- Dobner T.
- Kabelitz D.
- Krönke M.
- Schütze S.
Inhibition of TNF receptor 1 internalization by adenovirus 14.7K as a novel immune escape mechanism.
). As examples, preventing TNFR1 internalization through pharmacologic intervention or by deleting the TNFR1 internalization domain obstructs caspase activation while simultaneously preserving NF-κB survival signaling (
10- Schneider-Brachert W.
- Tchikov V.
- Neumeyer J.
- Jakob M.
- Winoto-Morbach S.
- Held-Feindt J.
- Heinrich M.
- Merkel O.
- Ehrenschwender M.
- Adam D.
- Mentlein R.
- Kabelitz D.
- Schütze S.
Compartmentalization of TNF receptor 1 signaling: internalized TNF receptosomes as death signaling vesicles.
,
35- Schütze S.
- Machleidt T.
- Adam D.
- Schwandner R.
- Wiegmann K.
- Kruse M.L.
- Heinrich M.
- Wickel M.
- Krönke M.
Inhibition of receptor internalization by monodansylcadaverine selectively blocks p55 tumor necrosis factor receptor death domain signaling.
,
36- Schneider-Brachert W.
- Tchikov V.
- Merkel O.
- Jakob M.
- Hallas C.
- Kruse M.L.
- Groitl P.
- Lehn A.
- Hildt E.
- Held-Feindt J.
- Dobner T.
- Kabelitz D.
- Krönke M.
- Schütze S.
Inhibition of TNF receptor 1 internalization by adenovirus 14.7K as a novel immune escape mechanism.
). However, the biological mechanisms that regulate the partitioning of TNFR1 between these compartments remain elusive. As ST6Gal-I expression is dynamically regulated in many cell types, fluctuating levels of TNFR1 α2-6 sialylation are physiologically relevant and offer a novel mechanism for dictating translocation of TNFR1 between the cell surface and endosomes.
Elevated receptor α2-6 sialylation is prevalent in tumor cells because of the up-regulation of ST6Gal-I in diverse human malignancies (
16Significance of β-galactoside α2,6 sialyltransferase 1 in cancers.
,
18- Schultz M.J.
- Swindall A.F.
- Bellis S.L.
Regulation of the metastatic cell phenotype by sialylated glycans.
,
37- Dall'Olio F.
- Chiricolo M.
Sialyltransferases in cancer.
). Immunohistochemical studies show that ST6Gal-I is highly expressed in pancreatic, ovarian, and colon cancer, whereas ST6Gal-I levels are low in the normal epithelium of these organs (
21- Swindall A.F.
- Londoño-Joshi A.I.
- Schultz M.J.
- Fineberg N.
- Buchsbaum D.J.
- Bellis S.L.
ST6Gal-I protein expression is upregulated in human epithelial tumors and correlates with stem cell markers in normal tissues and colon cancer cell lines.
,
22- Schultz M.J.
- Holdbrooks A.T.
- Chakraborty A.
- Grizzle W.E.
- Landen C.N.
- Buchsbaum D.J.
- Conner M.G.
- Arend R.C.
- Yoon K.J.
- Klug C.A.
- Bullard D.C.
- Kesterson R.A.
- Oliver P.G.
- O'Connor A.K.
- Yoder B.K.
- Bellis S.L.
The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a cancer stem cell phenotype.
). High ST6Gal-I expression is correlated with poor patient prognosis in ovarian, colon, breast, and pancreatic cancer (
22- Schultz M.J.
- Holdbrooks A.T.
- Chakraborty A.
- Grizzle W.E.
- Landen C.N.
- Buchsbaum D.J.
- Conner M.G.
- Arend R.C.
- Yoon K.J.
- Klug C.A.
- Bullard D.C.
- Kesterson R.A.
- Oliver P.G.
- O'Connor A.K.
- Yoder B.K.
- Bellis S.L.
The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a cancer stem cell phenotype.
,
38- Lise M.
- Belluco C.
- Perera S.P.
- Patel R.
- Thomas P.
- Ganguly A.
Clinical correlations of α2,6-sialyltransferase expression in colorectal cancer patients.
,
39- Recchi M.A.
- Hebbar M.
- Hornez L.
- Harduin-Lepers A.
- Peyrat J.P.
- Delannoy P.
Multiplex reverse transcription polymerase chain reaction assessment of sialyltransferase expression in human breast cancer.
40- Hsieh C.C.
- Shyr Y.M.
- Liao W.Y.
- Chen T.H.
- Wang S.E.
- Lu P.C.
- Lin P.Y.
- Chen Y.B.
- Mao W.Y.
- Han H.Y.
- Hsiao M.
- Yang W.B.
- Li W.S.
- Sher Y.P.
- Shen C.N.
Elevation of β-galactoside α2,6-sialyltransferase 1 in a fructose-responsive manner promotes pancreatic cancer metastasis.
), and animal models support a tumor-promoting function for ST6Gal-I (
22- Schultz M.J.
- Holdbrooks A.T.
- Chakraborty A.
- Grizzle W.E.
- Landen C.N.
- Buchsbaum D.J.
- Conner M.G.
- Arend R.C.
- Yoon K.J.
- Klug C.A.
- Bullard D.C.
- Kesterson R.A.
- Oliver P.G.
- O'Connor A.K.
- Yoder B.K.
- Bellis S.L.
The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a cancer stem cell phenotype.
,
40- Hsieh C.C.
- Shyr Y.M.
- Liao W.Y.
- Chen T.H.
- Wang S.E.
- Lu P.C.
- Lin P.Y.
- Chen Y.B.
- Mao W.Y.
- Han H.Y.
- Hsiao M.
- Yang W.B.
- Li W.S.
- Sher Y.P.
- Shen C.N.
Elevation of β-galactoside α2,6-sialyltransferase 1 in a fructose-responsive manner promotes pancreatic cancer metastasis.
). In the azoxymethane/dextran sulfate sodium (AOM/DSS) inflammation-associated colon tumorigenesis model, for which the TNF/TNFR1 axis plays a major role (
41- Popivanova B.K.
- Kitamura K.
- Wu Y.
- Kondo T.
- Kagaya T.
- Kaneko S.
- Oshima M.
- Fujii C.
- Mukaida N.
Blocking TNF-α in mice reduces colorectal carcinogenesis associated with chronic colitis.
), mice with forced ST6Gal-I overexpression have an increased incidence of tumor development and overall greater tumor burden (
22- Schultz M.J.
- Holdbrooks A.T.
- Chakraborty A.
- Grizzle W.E.
- Landen C.N.
- Buchsbaum D.J.
- Conner M.G.
- Arend R.C.
- Yoon K.J.
- Klug C.A.
- Bullard D.C.
- Kesterson R.A.
- Oliver P.G.
- O'Connor A.K.
- Yoder B.K.
- Bellis S.L.
The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a cancer stem cell phenotype.
). As another example, ST6Gal-I overexpression propels pancreatic cancer metastasis in an orthotopic transplant model (
40- Hsieh C.C.
- Shyr Y.M.
- Liao W.Y.
- Chen T.H.
- Wang S.E.
- Lu P.C.
- Lin P.Y.
- Chen Y.B.
- Mao W.Y.
- Han H.Y.
- Hsiao M.
- Yang W.B.
- Li W.S.
- Sher Y.P.
- Shen C.N.
Elevation of β-galactoside α2,6-sialyltransferase 1 in a fructose-responsive manner promotes pancreatic cancer metastasis.
). ST6Gal-I activity likely contributes to malignant progression through imparting metastatic cellular behaviors such as invasiveness (
40- Hsieh C.C.
- Shyr Y.M.
- Liao W.Y.
- Chen T.H.
- Wang S.E.
- Lu P.C.
- Lin P.Y.
- Chen Y.B.
- Mao W.Y.
- Han H.Y.
- Hsiao M.
- Yang W.B.
- Li W.S.
- Sher Y.P.
- Shen C.N.
Elevation of β-galactoside α2,6-sialyltransferase 1 in a fructose-responsive manner promotes pancreatic cancer metastasis.
,
42- Lin S.
- Kemmner W.
- Grigull S.
- Schlag P.M.
Cell surface α2,6 sialylation affects adhesion of breast carcinoma cells.
43- Christie D.R.
- Shaikh F.M.
- Lucas 4th, J.A.
- Lucas 3rd, J.A.
- Bellis S.L.
ST6Gal-I expression in ovarian cancer cells promotes an invasive phenotype by altering integrin glycosylation and function.
,
44- Zhu Y.
- Srivatana U.
- Ullah A.
- Gagneja H.
- Berenson C.S.
- Lance P.
Suppression of a sialyltransferase by antisense DNA reduces invasiveness of human colon cancer cells in vitro.
45- Shaikh F.M.
- Seales E.C.
- Clem W.C.
- Hennessy K.M.
- Zhuo Y.
- Bellis S.L.
Tumor cell migration and invasion are regulated by expression of variant integrin glycoforms.
), epithelial–mesenchymal transition (
46- Lu J.
- Isaji T.
- Im S.
- Fukuda T.
- Hashii N.
- Takakura D.
- Kawasaki N.
- Gu J.
β-Galactoside α2,6-sialyltranferase 1 promotes transforming growth factor-β-mediated epithelial-mesenchymal transition.
), and hallmark cancer stem cell characteristics, including anchorage-independent tumor spheroid growth and tumor-initiating potential (
22- Schultz M.J.
- Holdbrooks A.T.
- Chakraborty A.
- Grizzle W.E.
- Landen C.N.
- Buchsbaum D.J.
- Conner M.G.
- Arend R.C.
- Yoon K.J.
- Klug C.A.
- Bullard D.C.
- Kesterson R.A.
- Oliver P.G.
- O'Connor A.K.
- Yoder B.K.
- Bellis S.L.
The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a cancer stem cell phenotype.
).
Accumulating evidence suggests that one of the main functions of ST6Gal-I may be to protect cancer cells against a variety of microenvironmental assaults. In addition to inhibiting Fas (
34- Swindall A.F.
- Bellis S.L.
Sialylation of the Fas death receptor by ST6Gal-I provides protection against Fas-mediated apoptosis in colon carcinoma cells.
) and TNFR1-dependent cell death, α2-6 sialylation of select receptors inhibits apoptosis induced by extracellular galectins (
47- Zhuo Y.
- Chammas R.
- Bellis S.L.
Sialylation of β1 integrins blocks cell adhesion to galectin-3 and protects cells against galectin-3-induced apoptosis.
,
48- Fukumori T.
- Takenaka Y.
- Yoshii T.
- Kim H.R.
- Hogan V.
- Inohara H.
- Kagawa S.
- Raz A.
CD29 and CD7 mediate galectin-3-induced type II T-cell apoptosis.
49- Toscano M.A.
- Bianco G.A.
- Ilarregui J.M.
- Croci D.O.
- Correale J.
- Hernandez J.D.
- Zwirner N.W.
- Poirier F.
- Riley E.M.
- Baum L.G.
- Rabinovich G.A.
Differential glycosylation of TH1, TH2 and TH-17 effector cells selectively regulates susceptibility to cell death.
), a family of galactose-binding lectins. ST6Gal-I also protects cells from cytotoxicity induced by serum deprivation (
19- Britain C.M.
- Dorsett K.A.
- Bellis S.L.
The glycosyltransferase ST6Gal-I protects tumor cells against serum growth factor withdrawal by enhancing survival signaling and proliferative potential.
), radiation (
50- Lee M.
- Park J.J.
- Lee Y.S.
Adhesion of ST6Gal I-mediated human colon cancer cells to fibronectin contributes to cell survival by integrin β1-mediated paxillin and AKT activation.
), and chemotherapy drugs (
20- Schultz M.J.
- Swindall A.F.
- Wright J.W.
- Sztul E.S.
- Landen C.N.
- Bellis S.L.
ST6Gal-I sialyltransferase confers cisplatin resistance in ovarian tumor cells.
,
22- Schultz M.J.
- Holdbrooks A.T.
- Chakraborty A.
- Grizzle W.E.
- Landen C.N.
- Buchsbaum D.J.
- Conner M.G.
- Arend R.C.
- Yoon K.J.
- Klug C.A.
- Bullard D.C.
- Kesterson R.A.
- Oliver P.G.
- O'Connor A.K.
- Yoder B.K.
- Bellis S.L.
The tumor-associated glycosyltransferase ST6Gal-I regulates stem cell transcription factors and confers a cancer stem cell phenotype.
,
51- Chen X.
- Wang L.
- Zhao Y.
- Yuan S.
- Wu Q.
- Zhu X.
- Niang B.
- Wang S.
- Zhang J.
ST6Gal-I modulates docetaxel sensitivity in human hepatocarcinoma cells via the p38 MAPK/caspase pathway.
). Our results add to this body of literature by elucidating a novel pathway by which ST6Gal-I may promote tumor cell survival within TNF-rich inflammatory tumor microenvironments. Furthermore, this study addresses one of the major knowledge gaps in TNF signaling; namely, the mechanisms that dictate whether TNF induces cell survival or cell death.
Experimental procedures
Cell culture
MiaPaCa-2 cells were purchased from the ATCC, and OV4 cells were obtained from Dr. Timothy Eberlein at Harvard University (Boston, MA). Cells were grown in Dulbecco’s modified Eagle’s medium/F12 (OV4) or Dulbecco’s modified Eagle’s medium (MiaPaCa-2) containing 10% fetal bovine serum and 1% antibiotic/antimycotic supplements (GE Healthcare Hyclone). Stable polyclonal cell lines were created by transducing cells with an empty vector lentivirus (Sigma) or a lentivirus encoding either the ST6Gal-I gene (Genecopoeia) or shRNA against ST6Gal-I (Sigma, TN00000035432, sequence CCGGCGTGTGCTACTACTACCAGAACTCGAGTTCTGGTAGTAGTAGCACACGTTTTTG), followed by selection with 10 μg/ml of puromycin (Sigma). Puromycin was removed from the medium at least 2 days prior to all experiments.
ST6Gal-I expression and activity confirmation assays
ST6Gal-I overexpression or knockdown was verified by immunoblotting. Briefly, cells were lysed in radioimmune precipitation assay buffer supplemented with 1× protease and phosphatase inhibitors (Pierce). Total protein concentration was measured by BCA (Pierce). Samples were resolved by SDS-PAGE and transferred to polyvinylidene difluoride membranes. Membranes were incubated with 5% nonfat dry milk in TBS buffer containing 0.1% Tween 20 (TBS-T) and then probed with a goat polyclonal antibody against ST6Gal-I (R&D Systems, AF5924, lot CDSF0114101). Membranes were incubated with anti-goat secondary antibody and developed using ECL detection methods. In addition to immunoblotting, ST6Gal-I activity was monitored by using the SNA lectin to detect surface α2-6 sialic acids. MiaPaCa-2 or OV4 cells were stained with a 1:100 or 1:500 dilution, respectively, of FITC-conjugated SNA lectin (Vector, B-1305) for 30 min at 4 °C. SNA lectin binding was quantified via flow cytometry. To confirm that TNFR1 is a substrate for ST6Gal-I and that α2-6 sialylation of TNFR1 is altered by ST6Gal-I manipulation, cell lysates (500 μg for MiPaCa-2 and 200 μg for OV4) were incubated with 50 μl of SNA-conjugated agarose beads (Vector, AL-1303) overnight at 4 °C. α2-6–sialylated proteins bound to the beads were then precipitated by centrifugation, washed twice with PBS, and resuspended in 1× sample buffer (Bio-Rad) plus 10% 2-mercaptoethanol (Sigma). Proteins were resolved by SDS-PAGE and immunoblotted against TNFR1 (Cell Signaling Technology, 3736, lot 2).
TNF treatments
All studies involving TNF treatments were done in medium containing 1% serum (2 h preincubation in 1% serum-containing medium, followed by administration of TNF in 1% serum). For studies evaluating cellular morphological changes and ST6Gal-I enrichment, MiaPaCa-2 cells, which are highly sensitive to TNF-induced cell death, were cultured with 100 ng/ml recombinant human TNF (R&D Systems, 210-TA) for 24 h, whereas OV4 cells were cultured with a combination of 100 ng/ml TNF plus 5 μm cycloheximide (CHX, Sigma) to amplify the cytotoxicity of TNF. To examine TNF-induced signaling, cells were cultured with 10 ng/ml TNF for the indicated times, and the only exception to this method was the evaluation of caspase-3/8 activation in OV4 cells, in which 5 μm CHX was used in addition to 10 ng/ml TNF. To examine the contribution of TNFR1 to basal signaling differences in OV4 cells, OV4 EV or OE cells were incubated with anti-TNFR1 neutralizing antibody (R&D Systems, MAB225, lot IP0914031) in 1% serum-containing medium, and lysates were collected 24 h later and immunoblotted.
Immunoblotting
Cells were treated with TNF for the indicated times and then lysed in radioimmune precipitation assay buffer as described previously. Following SDS-PAGE and transfer, membranes were blocked in 5% nonfat dry milk in TBS-T. Immunoblots were probed with antibodies against ST6Gal-I (R&D Systems, AF5924, lot CDSF0114101), cIAP2 (Cell Signaling Technology, 3130, lot 6), pAkt (Ser-473, Cell Signaling Technology, 4060, lot 19), total Akt (Cell Signaling Technology, 4691, lot 20), pNF-κB–p65 (Ser-536, Cell Signaling Technology, 3033, lot 14), total NF-κB–p65 (Cell Signaling Technology, 8242, lot 4), pJNK (Thr-183/Tyr-185, Cell Signaling Technology, 9255, lot 32), total JNK (Cell Signaling Technology, 9258, lot 17), IκBα (Cell Signaling Technology, 4812, lot 9), pErk1/2 (Thr-202/Tyr-204, Cell Signaling Technology, 4370, lot 15), total Erk1/2 (Cell Signaling Technology, 9102, lot 26), cleaved caspase-3 (Asp-175, Cell Signaling Technology, 9661, lot 43), cleaved caspase-8 (Asp-391, Cell Signaling Technology, 9496, lot 7), and TNFR1 (Cell Signaling Technology, 3736, lot 2). Protein loading was verified using horseradish peroxidase (HRP)–conjugated anti-actin (Abcam, ab20272, lot GR201277) or HRP-conjugated anti-tubulin (Abcam, ab21058, lot GR284232). Membranes were incubated with appropriate HRP-coupled secondary antibodies (anti-rabbit and anti-mouse IgG, Cell Signaling Technology; anti-goat IgG, Santa Cruz Biotechnology), and protein was detected by ECL (Pierce), Clarity (Bio-Rad), or SuperSignal West Femto substrate (Pierce).
Immunofluorescence assays
Cells of the same background were prelabeled with 25 μm of their own distinct red or green CellTracker dyes (Thermo Scientific) (Thermo Scientific) and incubated at 37 °C for 30 min. Cells were then washed three times with 1× PBS and seeded at equal densities into the same tissue culture–treated plate. After allowing the cells to adhere for 4 h, the cells were then treated with 100 ng/ml TNF (OV4, TNF plus 5 μm CHX). After 24-h treatment, the cells were imaged with an EVOS fluorescence microscope. To visualize NF-κB subcellular localization, cells were seeded onto lysine-coated chamber slides and treated with 10 ng/ml TNF for 15 min. The cells were immediately washed in ice-cold PBS to block TNF signaling and fixed with 4% paraformaldehyde for 10 min at room temperature. Cells were then permeabilized using 0.1% Triton X-100 (Fisher) for 10 min. The cells were blocked using 10% newborn calf serum (Atlanta Biologicals) and then incubated with total NF-κB-p65 antibody overnight at 4 °C. The primary antibody was washed off, followed by incubation with a donkey anti-rabbit IgG secondary antibody conjugated with Alexa Fluor 488 (Life Technologies). Coverslips were then mounted, and the slides were imaged with a fluorescence microscope (Nikon) fitted with a Nikon CoolSNAP camera.
Caspase-3/7 luminescence assays
Cells were seeded at equal densities into a 96-well tissue culture plate and allowed to adhere overnight. Prior to TNF treatment, cells were incubated in 1% serum-containing medium for 2 h and then treated with 100 ng/ml TNF (OV4, TNF plus 5 μm CHX) for the indicated times. Reconstituted Caspase-Glo 3/7 assay reagent (Promega) was then added to each well, mixed via an orbital shaker, and incubated at room temperature for 45 min. Luminescence was quantified with a BioTek Synergy H1 instrument. In the same experiment, with a separate 96-well plate of the same treatment conditions, the CellTiter-Glo (ATP) assay system (Promega) was utilized as a representation of overall cell number. The values represented are the ratios of values for Caspase-Glo relative to CellTiter-Glo.
TNFR1 shedding and internalization assays
To evaluate TNFR1 shedding, U937 EV or OE cells were treated with either TNF (100 ng/ml) or PMA (200 ng/ml) for 24 h. The supernatant was then collected and analyzed via an ELISA kit specific for soluble TNFR1 (R&D Systems). To examine surface TNFR1 levels, OV4 EV or OE cells were incubated in serum-free medium with TNF-biotin (R&D Systems) at 4 °C for 1 h. Unbound TNF-biotin was washed off with ice-cold PBS. Cells were then incubated with prewarmed 37 °C serum-free medium for 10 min. Control cells were maintained at 4 °C. The cells were fixed with 2% paraformaldehyde for 20 min at room temperature. Following fixation, the cells were incubated with 5 μg/ml streptavidin–FITC (Invitrogen) for 30 min on ice. Cells were then washed twice with PBS, and residual surface-bound TNF-biotin–TNFR1 complexes were quantified via flow cytometry. To analyze the effect of internalization and shedding inhibition, OV4 EV or OE cells were preincubated for 1 h with 5 μm Dyngo-4a or 50 μm TAPI-1 (both from Selleckchem) and then treated with TNF (100 ng/ml) plus CHX (1 μm). Images were obtained following 24 h of treatment.
Article info
Publication history
Published online: December 12, 2017
Received in revised form:
November 5,
2017
Received:
June 9,
2017
Edited by Alex Toker
Footnotes
This work was supported by National Institutes of Health Grants R01 GM111093 and R21 CA192629 (to S. L. B.), a T32 HL007918 cardiovascular pathophysiology training grant fellowship (to A. T. H.), and an American Heart Association Predoctoral Fellowship (to A. T. H.). The authors declare that they have no conflicts of interest with the contents of this article. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Copyright
© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.