70 results on '"YiJiang Shi"'
Search Results
2. Data from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Prior work indicates DEPTOR expression in multiple myeloma cells could be a therapeutic target. DEPTOR binds to mTOR via its PDZ domain and inhibits mTOR kinase activity. We previously identified a drug, which prevented mTOR–DEPTOR binding (NSC126405) and induced multiple myeloma cytotoxicity. We now report on a related therapeutic, drug 3g, which induces proteasomal degradation of DEPTOR. DEPTOR degradation followed drug 3g binding to its PDZ domain and was not due to caspase activation or enhanced mTOR phosphorylation of DEPTOR. Drug 3g enhanced mTOR activity, and engaged the IRS-1/PI3K/AKT feedback loop with reduced phosphorylation of AKT on T308. Activation of TORC1, in part, mediated multiple myeloma cytotoxicity. Drug 3g was more effective than NSC126405 in preventing binding of recombinant DEPTOR to mTOR, preventing binding of DEPTOR to mTOR inside multiple myeloma cells, in activating mTOR and inducing apoptosis in multiple myeloma cells. In vivo, drug 3g injected daily abrogated DEPTOR expression in xenograft tumors and induced an antitumor effect although modest weight loss was seen. Every-other-day treatment, however, was equally effective without weight loss. Drug 3g also reduced DEPTOR expression in normal tissues. Although no potential toxicity was identified in hematopoietic or hepatic function, moderate cardiac enlargement and glomerular mesangial hypertrophy was seen. DEPTOR protected multiple myeloma cells against bortezomib suggesting anti-DEPTOR drugs could synergize with proteasome inhibitors (PI). Indeed, combinations of drug NSC126405 + bortezomib were synergistic. In contrast, drug 3g was not and was even antagonistic. This antagonism was probably due to prevention of proteasomal DEPTOR degradation.
- Published
- 2023
3. figure S6 from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Suppl fig 6: Immunocompetent non-tumor bearing mice injected IP daily with increasing concentrations of drug 3g for 21 days followed by harvest of serum for assays of alkaline phosphatase, ALT, AST, bilirubin and BUN. Chemistry values of control mice injected daily with DMSO (3g dose=0) were arbitrarily made '1'. Data are means+/-SD, N=4 mice/group.
- Published
- 2023
4. Supplementary Figure 5 from Metabolomics Identifies Pyrimidine Starvation as the Mechanism of 5-Aminoimidazole-4-Carboxamide-1-β-Riboside-Induced Apoptosis in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Yijiang Shi, Bao Hoang, Patrick Frost, Sara Bassilian, Joseph R. Reeve, Sanjai Sharma, and Carolyne Bardeleben
- Abstract
PDF file - 82K, Cell cycle analysis of cells treated with AICAr in the presence or absence of uridine.
- Published
- 2023
5. figure S7 from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Suppl fig 7: A: Examples of IHC staining of normal tissues for DEPTOR expression following daily injection of DMSO or drug 3g (10mg/kg x 21 days); B) Immunoblot analysis of heart tissue for DEPTOR expression
- Published
- 2023
6. figure S3 from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Supplemental figure 3: Mice were challenged SQ with 8226 MM cells and, when tumors were 500mm3 mice were treated with DMSO(control) or drug 3g at 20mg/kg by daily IP injection (4 mice/group). Data are tumor volume at days 2, 3 or 4 after starting treatment, mean+/-SD.
- Published
- 2023
7. figure S8 from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Supplemental figure 8: Representative sections of liver obtained from control (DMSO) mice or mice injected IP daily with drug 3g at 20mg/kg. Magnification=100x
- Published
- 2023
8. supplemental figure 3 from SGK Kinase Activity in Multiple Myeloma Cells Protects against ER Stress Apoptosis via a SEK-Dependent Mechanism
- Author
-
Alan Lichtenstein, Carolyne Bardeleben, Veena Mysore, Patrick J. Frost, Yijiang Shi, and Bao Hoang
- Abstract
Suppl fig 3: A) MM1.S or 8226 cells lines transfected with empty vector (EV), wild type SGK (sgk) or phosphomimetic SGK (sgk422D) and cultured in serum free media for days 1-7. At designated days, apoptosis assay performed. Data are percent apoptosis, mean+/-SE, n=5; B) MM1.S or OPM-2 cell lines similarly transfected as in A were exposed to dexamethasone for 72 hrs followed by MTT assay. Data are mean cell survival+/-SE, n=4.
- Published
- 2023
9. figure S5 from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Suppl fig 5: A) Mice (4/group) bearing SQ 8226 xenografts were in- Jected IP with DMSO (control (C)) or drug 3g at 20 mg/kg and tumors were excised 24, 48 or 72 hrs later. Tumors were stained for DEPTOR expression. Data are number of DEPTOR-positive cells, mean+/-SD, N=4. *denotes significant decrease versus control, p
- Published
- 2023
10. supplemental figure 2 from SGK Kinase Activity in Multiple Myeloma Cells Protects against ER Stress Apoptosis via a SEK-Dependent Mechanism
- Author
-
Alan Lichtenstein, Carolyne Bardeleben, Veena Mysore, Patrick J. Frost, Yijiang Shi, and Bao Hoang
- Abstract
Suppl fig 2 ) MM1.S or OPM-2 MM cells stably transfected with empty vector (ev), wild type AKT (akt), phosphomimetic AKT, (akt473d), wild type SGK (sgk) or Phosphomimetic SGK (sgk422d), challenged with pp242 (left panels) or torin (right panels) followed by apoptosis assay. Data are percent apoptosis above control (no drug) mean+/-SE, n=5. Asterixes denote significantly decreased apoptosis (p
- Published
- 2023
11. Supplementary Figure 3 from Metabolomics Identifies Pyrimidine Starvation as the Mechanism of 5-Aminoimidazole-4-Carboxamide-1-β-Riboside-Induced Apoptosis in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Yijiang Shi, Bao Hoang, Patrick Frost, Sara Bassilian, Joseph R. Reeve, Sanjai Sharma, and Carolyne Bardeleben
- Abstract
PDF file - 2312K, Heat map and table of metabolites of significantly altered metabolites in AICAr-treated samples for rapamycin treated samples.
- Published
- 2023
12. supplemental figure 6 from SGK Kinase Activity in Multiple Myeloma Cells Protects against ER Stress Apoptosis via a SEK-Dependent Mechanism
- Author
-
Alan Lichtenstein, Carolyne Bardeleben, Veena Mysore, Patrick J. Frost, Yijiang Shi, and Bao Hoang
- Abstract
Suppl fig 6: Combination indices (CIs) and fraction affected in 8226 cells treated with either bortezomib +/- SGK inhibitor (in A) or Staurosporine +/- SGK inhibitor (in B). CIs determined from apoptosis data of 4 independent experiments using mean values.
- Published
- 2023
13. Supplementary Figure 1 from Metabolomics Identifies Pyrimidine Starvation as the Mechanism of 5-Aminoimidazole-4-Carboxamide-1-β-Riboside-Induced Apoptosis in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Yijiang Shi, Bao Hoang, Patrick Frost, Sara Bassilian, Joseph R. Reeve, Sanjai Sharma, and Carolyne Bardeleben
- Abstract
PDF file - 64K, Chemical structure of bortezomib, melphalan and metformin.
- Published
- 2023
14. supplemental figure 5 from SGK Kinase Activity in Multiple Myeloma Cells Protects against ER Stress Apoptosis via a SEK-Dependent Mechanism
- Author
-
Alan Lichtenstein, Carolyne Bardeleben, Veena Mysore, Patrick J. Frost, Yijiang Shi, and Bao Hoang
- Abstract
Suppl fig 5: SGK protein stability in 8226 cells treated +/- bortezomib (20nM); Data are Mean+/-SD, n=3.
- Published
- 2023
15. supplemental figure 4 from SGK Kinase Activity in Multiple Myeloma Cells Protects against ER Stress Apoptosis via a SEK-Dependent Mechanism
- Author
-
Alan Lichtenstein, Carolyne Bardeleben, Veena Mysore, Patrick J. Frost, Yijiang Shi, and Bao Hoang
- Abstract
Suppl fig 4: Lysate harvested from tumors of mice challenged with EV- or SGK-transfected tumors and treated with 0, 0.5 or 1 mg/kg Bortezomib; Lysates are combined from 3 mice/group and harvested 1 week after initiating treatment (mice had received 2 treatments); IKB-alpha then immunoprecipitated from lysates and immunoblotted for IKB as well as ubiquitin
- Published
- 2023
16. Figure S1 from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Supplemental figure 1: A) Hematopoietic colony formation in marrow cells exposed to drug 3g for 14 days (assay as described in (3); Data represent % of control (0uM), mean+/-SD; B)8226 MM cells, PC3 prostate cancer cells, LLC Lewis lung cancer cells and JHH7 hepatocellular cancer cells were incubated for 48 hrs +/- drug 3g at 0 (DMSO), 1 or 2 uM in MTT assays. Data are mean+/-SD, n=3 for MTT cytotoxicity where DMSO controls are 100% survival
- Published
- 2023
17. Supplementary Figure from Critical Role for Cap-Independent c-MYC Translation in Progression of Multiple Myeloma
- Author
-
Alan Lichtenstein, Siegfried Janz, Joseph F. Gera, Binod Dhakal, Brent Holmes, Yan Cheng, Fumou Sun, and Yijiang Shi
- Abstract
Supplementary Figure from Critical Role for Cap-Independent c-MYC Translation in Progression of Multiple Myeloma
- Published
- 2023
18. figure S4 from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Suppl fig 4: A) Mice were challenged with 8226 cells (admixed with Matrigel) SC and, when tumor size was 300mm3, mice were randomized to receive drug 3g at 5 mg/kg IP daily (X) for 9 days or DMSO (o); Results shown are mean +/- SD tumor volume,n=8 mice/group. The tumor growth was significantly slowed (p
- Published
- 2023
19. Figure S2 from A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Alan Lichtenstein, Joseph F. Gera, Michael E. Jung, Jihye Lee, Rogelio Hernandez-Pando, Gabriela Antonio-Andres, Sara Huerta-Yepez, Patrick Frost, Yijiang Shi, and Mario I. Vega
- Abstract
Suppl fig 2:A) Co-IP between DEPTOR & mTOR in 8226 treated with NSC126405 or 3g; B,C&D) Immunoblot of 8226 cells after 6 hrs incubation with drugs. Relative ex- pression below gels are means of 3 separate experiments; E) Bead pull-down assay to quantify binding of recombinant DEPTOR to mTOR (see Materials & Methods); Lane 1 (furthest to the left) is a control showing anti-FLAG-containing beads (w/o conjugation to mTOR-FLAG) do not non-specifically bind to GST-DEPTOR while mTOR-FLAG-conjugated beads successfully bind GST-DEPTOR (lane 2); Below gel are results from 3 separate experiments showing mean +/-SD percent of control, n=3. DM=DMSO treatment; Drug J007 is a myc IRES inhibitor (6) of similar size to 3g and used as an additional negative control. *=significant decrease from control (DMSO), p
- Published
- 2023
20. Supplementary Figure 6 from Metabolomics Identifies Pyrimidine Starvation as the Mechanism of 5-Aminoimidazole-4-Carboxamide-1-β-Riboside-Induced Apoptosis in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Yijiang Shi, Bao Hoang, Patrick Frost, Sara Bassilian, Joseph R. Reeve, Sanjai Sharma, and Carolyne Bardeleben
- Abstract
PDF file - 76K, Cell cycle analysis of cells treated with PALA in the presence or absence of uridine.
- Published
- 2023
21. Supplementary Figure 2 from Metabolomics Identifies Pyrimidine Starvation as the Mechanism of 5-Aminoimidazole-4-Carboxamide-1-β-Riboside-Induced Apoptosis in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Yijiang Shi, Bao Hoang, Patrick Frost, Sara Bassilian, Joseph R. Reeve, Sanjai Sharma, and Carolyne Bardeleben
- Abstract
PDF file - 1005K, IGF does not protect against AICAr induced apoptosis.
- Published
- 2023
22. supplemental figure 2 from Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Joseph Gera, Michael E. Jung, Manuel L. Penichet, Carolyne Bardeleben, Richard S. Finn, Jihye Lee, Patrick Frost, Tracy R. Daniels-Wells, and Yijiang Shi
- Abstract
SUPPL FIG 2: A) 8226 cells treated with or w/o NSC 126405 for 6 hrs, followed by immunoprecipitation of mTOR (or control IgG). Immunoprecipitates immunoblotted for expression of mTOR and DEPTOR. Input shown for DEPTOR, mTOR and tubulin expression. B) 8226 cells treated with DMSO, pp242 or NSC 126405 and then tested for 4E-BP1 phosphorylation by flow cytometry. C) Summarized data from all 3 treatment groups
- Published
- 2023
23. supplemental figure legends from Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Joseph Gera, Michael E. Jung, Manuel L. Penichet, Carolyne Bardeleben, Richard S. Finn, Jihye Lee, Patrick Frost, Tracy R. Daniels-Wells, and Yijiang Shi
- Abstract
legends of supplemental figures
- Published
- 2023
24. supplemental figure 6 from Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Joseph Gera, Michael E. Jung, Manuel L. Penichet, Carolyne Bardeleben, Richard S. Finn, Jihye Lee, Patrick Frost, Tracy R. Daniels-Wells, and Yijiang Shi
- Abstract
SUPPL FIG 6: PBLs obtained from 3 normal individuals and incubated with increasing concentrations of NSC 126405 for 72 hrs. Surviving viable cells enumerated. Data are % of control (no drug B), mean+/-SD, n=3.
- Published
- 2023
25. supplemental figure 1 from Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Joseph Gera, Michael E. Jung, Manuel L. Penichet, Carolyne Bardeleben, Richard S. Finn, Jihye Lee, Patrick Frost, Tracy R. Daniels-Wells, and Yijiang Shi
- Abstract
SUPPL FIG 1: A) Only yeast transfected with both FAT domain/GAL4DBD +DEPTOR/GAL4 AD show growth on selective media. Yeast transfected w/ either construct alone can not grow. Two separate transformations shown. B) Four initial 'hits' from drug screen
- Published
- 2023
26. Supplementary Fig. S2 from Effect of autophagy on multiple myeloma cell viability
- Author
-
Alan Lichtenstein, Patrick Frost, Yijiang Shi, Angelica Benavides, and Bao Hoang
- Abstract
Supplementary Fig. S2 from Effect of autophagy on multiple myeloma cell viability
- Published
- 2023
27. Data from Effect of autophagy on multiple myeloma cell viability
- Author
-
Alan Lichtenstein, Patrick Frost, Yijiang Shi, Angelica Benavides, and Bao Hoang
- Abstract
Because accumulation of potentially toxic malfolded protein may be extensive in immunoglobulin-producing multiple myeloma (MM) cells, we investigated the phenomenon of autophagy in myeloma, a physiologic process that can protect against malfolded protein under some circumstances. Autophagy in MM cell lines that express and secrete immunoglobulin and primary specimens was significantly increased by treatment with the endoplasmic reticulum stress–inducing agent thapsigargin, the mammalian target of rapamycin inhibitor rapamycin, and the proteasome inhibitor bortezomib. Inhibition of basal autophagy in these cell lines and primary cells by use of the inhibitors 3-methyladenine and chloroquine resulted in a cytotoxic effect that was associated with enhanced apoptosis. Use of small interfering RNA to knock down expression of beclin-1, a key protein required for autophagy, also inhibited viable recovery of MM cells. Because the data suggested that autophagy protected MM cell viability, we predicted that autophagy inhibitors would synergize with bortezomib for enhanced antimyeloma effects. However, the combination of these drugs resulted in an antagonistic response. In contrast, the autophagy inhibitor 3-methyladenine did synergize with thapsigargin for an enhanced cytotoxic response. These data suggest that autophagy inhibitors have therapeutic potential in myeloma but caution against combining such drugs with bortezomib. [Mol Cancer Ther 2009;8(7):1974–84]
- Published
- 2023
28. supplemental figure 3 from Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Joseph Gera, Michael E. Jung, Manuel L. Penichet, Carolyne Bardeleben, Richard S. Finn, Jihye Lee, Patrick Frost, Tracy R. Daniels-Wells, and Yijiang Shi
- Abstract
SUPPL FIG 3: A) Relative DEPTOR expression in MMCLs and hepatocellular carcinoma (HCC) cell lines in a representative experiment (upper panel) and means+/-SD of 3 experiments (lower bar graph); B) Relative sensitivity of U266 and FR4 MMCLs vs DEPTOR-expressing JHH7, HUH-1 or HIH7 HCCs; data are means +/- SD (n=3); Asterix demonstrates significant (p
- Published
- 2023
29. supplemental figure 4 from Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Joseph Gera, Michael E. Jung, Manuel L. Penichet, Carolyne Bardeleben, Richard S. Finn, Jihye Lee, Patrick Frost, Tracy R. Daniels-Wells, and Yijiang Shi
- Abstract
SUPPL FIG 4: A) 8226 cells treated +/- NSC 126405 for 18 hrs followed by immunoblot assays. Fold BIM increase is determined by densitometry, mean of 3 experiments; B) Control (shSCR) or RAPTOR-silenced 8226 cells (shRAPTOR) treated +/- NSC 126405 for 18 hrs followed by immunoblot assays. Fold BIM increase is mean of 3 experiments.
- Published
- 2023
30. supplemental figure 5 from Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Joseph Gera, Michael E. Jung, Manuel L. Penichet, Carolyne Bardeleben, Richard S. Finn, Jihye Lee, Patrick Frost, Tracy R. Daniels-Wells, and Yijiang Shi
- Abstract
SUPPL fig 5: Growth of individual tumors from mice injected with vehicle (control) or drug at 10 or 20 mg/kg
- Published
- 2023
31. Data from Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Joseph Gera, Michael E. Jung, Manuel L. Penichet, Carolyne Bardeleben, Richard S. Finn, Jihye Lee, Patrick Frost, Tracy R. Daniels-Wells, and Yijiang Shi
- Abstract
DEPTOR is a 48 kDa protein that binds to mTOR and inhibits this kinase in TORC1 and TORC2 complexes. Overexpression of DEPTOR specifically occurs in a model of multiple myeloma. Its silencing in multiple myeloma cells is sufficient to induce cytotoxicity, suggesting that DEPTOR is a potential therapeutic target. mTORC1 paralysis protects multiple myeloma cells against DEPTOR silencing, implicating mTORC1 in the critical role of DEPTOR in multiple myeloma cell viability. Building on this foundation, we interrogated a small-molecule library for compounds that prevent DEPTOR binding to mTOR in a yeast-two-hybrid assay. One compound was identified that also prevented DEPTOR–mTOR binding in human myeloma cells, with subsequent activation of mTORC1 and mTORC2. In a surface plasmon resonance (SPR) assay, the compound bound to recombinant DEPTOR but not to mTOR. The drug also prevented binding of recombinant DEPTOR to mTOR in the SPR assay. Remarkably, although activating TORC1 and TORC2, the compound induced apoptosis and cell-cycle arrest in multiple myeloma cell lines and prevented outgrowth of human multiple myeloma cells in immunodeficient mice. In vitro cytotoxicity against multiple myeloma cell lines was directly correlated with DEPTOR protein expression and was mediated, in part, by the activation of TORC1 and induction of p21 expression. Additional cytotoxicity was seen against primary multiple myeloma cells, whereas normal hematopoietic colony formation was unaffected. These results further support DEPTOR as a viable therapeutic target in multiple myeloma and suggest an effective strategy of preventing binding of DEPTOR to mTOR. Cancer Res; 76(19); 5822–31. ©2016 AACR.
- Published
- 2023
32. Turnover of the mTOR inhibitor, DEPTOR, and downstream AKT phosphorylation in multiple myeloma cells, is dependent on ERK1-mediated phosphorylation
- Author
-
Mario Vega, Yu Chen, Yijiang Shi, Joseph Gera, and Alan Lichtenstein
- Subjects
Mitogen-Activated Protein Kinase 3 ,Mutation ,Intracellular Signaling Peptides and Proteins ,Serine ,Humans ,Cell Biology ,MTOR Inhibitors ,Phosphorylation ,Multiple Myeloma ,Molecular Biology ,Biochemistry ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
DEPTOR is a 48 kDa protein upregulated in multiple myeloma (MM) cells. DEPTOR inhibits mTOR and, by repressing a negative feedback loop, promotes AKT activation. We previously identified a compound that binds to DEPTOR in MM cells and induces its proteasomal degradation. To identify the mechanism of degradation, here, we screened for drug-induced posttranslational modifications and identified reduced phosphorylation of DEPTOR on serine 235 (S235). We show that an S235 phosphomimetic DEPTOR mutant was resistant to degradation, confirming the importance of this posttranslational modification. In addition, a DEPTOR mutant with a serine-to-alanine substitution at S235 could only be expressed upon concurrent proteasome inhibition. Thus, S235 phosphorylation regulates DEPTOR stability. Screening the DEPTOR interactome identified that the association of USP-7 deubiquitinase with DEPTOR was dependent upon S235 phosphorylation. Inhibition of USP-7 activity resulted in DEPTOR polyubiquitination and degradation. A scansite search suggested that ERK1 may be responsible for S235 phosphorylation, which was confirmed through the use of inhibitors, ERK1 knockdown, and an in vitro kinase assay. Inhibition of ERK1 also downregulated AKT phosphorylation. To test if DEPTOR phosphorylation mediated this crosstalk, MM cells were transfected with WT or phosphomimetic DEPTOR and exposed to ERK inhibitors. Although WT DEPTOR had no effect on the inhibition of AKT phosphorylation, the phosphomimetic DEPTOR prevented inhibition. These results indicate that ERK1 maintains AKT activity in MM cells via phosphorylation of DEPTOR. We propose that DEPTOR-dependent crosstalk provides MM cells with a viability-promoting signal (through AKT) when proliferation is stimulated (through ERK).
- Published
- 2021
33. A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity
- Author
-
Mario I. Vega, Gabriela Antonio-Andres, Michael E. Jung, Sara Huerta-Yepez, Yijiang Shi, Jihye Lee, Patrick Frost, Rogelio Hernández-Pando, Joseph Gera, and Alan Lichtenstein
- Subjects
0301 basic medicine ,Cancer Research ,Antineoplastic Agents ,Apoptosis ,Mice, SCID ,DEPTOR ,Article ,Bortezomib ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Kinase activity ,Cytotoxicity ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Chemistry ,Intracellular Signaling Peptides and Proteins ,Treatment Outcome ,030104 developmental biology ,Oncology ,Proteasome ,030220 oncology & carcinogenesis ,Proteolysis ,Cancer research ,Phosphorylation ,Multiple Myeloma ,medicine.drug - Abstract
Prior work indicates DEPTOR expression in multiple myeloma cells could be a therapeutic target. DEPTOR binds to mTOR via its PDZ domain and inhibits mTOR kinase activity. We previously identified a drug, which prevented mTOR–DEPTOR binding (NSC126405) and induced multiple myeloma cytotoxicity. We now report on a related therapeutic, drug 3g, which induces proteasomal degradation of DEPTOR. DEPTOR degradation followed drug 3g binding to its PDZ domain and was not due to caspase activation or enhanced mTOR phosphorylation of DEPTOR. Drug 3g enhanced mTOR activity, and engaged the IRS-1/PI3K/AKT feedback loop with reduced phosphorylation of AKT on T308. Activation of TORC1, in part, mediated multiple myeloma cytotoxicity. Drug 3g was more effective than NSC126405 in preventing binding of recombinant DEPTOR to mTOR, preventing binding of DEPTOR to mTOR inside multiple myeloma cells, in activating mTOR and inducing apoptosis in multiple myeloma cells. In vivo, drug 3g injected daily abrogated DEPTOR expression in xenograft tumors and induced an antitumor effect although modest weight loss was seen. Every-other-day treatment, however, was equally effective without weight loss. Drug 3g also reduced DEPTOR expression in normal tissues. Although no potential toxicity was identified in hematopoietic or hepatic function, moderate cardiac enlargement and glomerular mesangial hypertrophy was seen. DEPTOR protected multiple myeloma cells against bortezomib suggesting anti-DEPTOR drugs could synergize with proteasome inhibitors (PI). Indeed, combinations of drug NSC126405 + bortezomib were synergistic. In contrast, drug 3g was not and was even antagonistic. This antagonism was probably due to prevention of proteasomal DEPTOR degradation.
- Published
- 2019
34. WITHDRAWN: Structure-activity relationship study of small molecule inhibitors of the DEPTOR-mTOR interaction
- Author
-
Yonghui Yang, Mario I. Vega, Michael E. Jung, Yijiang Shi, Jihye Lee, Joseph Gera, and Alan Lichtenstein
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,Kinase ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,mTORC1 ,DEPTOR ,Biochemistry ,mTORC2 ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Apoptosis ,Drug Discovery ,Molecular Medicine ,Cytotoxic T cell ,Molecular Biology ,PI3K/AKT/mTOR pathway - Abstract
DEPTOR is a 48 kDa protein that binds to mTOR and inhibits this kinase within mTORC1 and mTORC2 complexes. Over-expression of DEPTOR specifically occurs in the multiple myeloma (MM) tumor model and DEPTOR knockdown is cytotoxic to MM cells, suggesting it is a potential therapeutic target. Since mTORC1 paralysis protects MM cells against DEPTOR knockdown, it indicates that the protein-protein interaction between DEPTOR and mTOR is key to MM viability vs death. In a previous study, we used a yeast two-hybrid screen of a small inhibitor library to identify a compound that inhibited DEPTOR/mTOR binding in yeast. This therapeutic (compound B) also prevented DEPTOR/mTOR binding in MM cells and was selectively cytotoxic to MM cells. We now present a structure-activity relationship (SAR) study around this compound as a follow-up report of this previous work. This study has led to the discovery of five new leads – namely compounds 3g, 3k, 4d, 4e and 4g – all of which have anti-myeloma cytotoxic properties superior to compound B. Due to their targeting of DEPTOR, these compounds activate mTORC1 and selectively induce MM cell apoptosis and cell cycle arrest.
- Published
- 2017
35. Structure-activity relationship study of small molecule inhibitors of the DEPTOR-mTOR interaction
- Author
-
Yonghui Yang, Alan Lichtenstein, Joseph Gera, Mario I. Vega, Yijiang Shi, Jihye Lee, and Michael E. Jung
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,Clinical Biochemistry ,Pharmaceutical Science ,Apoptosis ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,DEPTOR ,Biochemistry ,mTORC2 ,Small Molecule Libraries ,03 medical and health sciences ,Inhibitory Concentration 50 ,Structure-Activity Relationship ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Humans ,Phosphorylation ,RNA, Small Interfering ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Kinase ,Chemistry ,TOR Serine-Threonine Kinases ,Organic Chemistry ,RPTOR ,Intracellular Signaling Peptides and Proteins ,Regulatory-Associated Protein of mTOR ,Cell biology ,030104 developmental biology ,Multiprotein Complexes ,Cancer research ,Molecular Medicine ,RNA Interference ,Protein Tyrosine Phosphatases ,Protein Binding - Abstract
DEPTOR is a 48 kDa protein that binds to mTOR and inhibits this kinase within mTORC1 and mTORC2 complexes. Over-expression of DEPTOR specifically occurs in the multiple myeloma (MM) tumor model and DEPTOR knockdown is cytotoxic to MM cells, suggesting it is a potential therapeutic target. Since mTORC1 paralysis protects MM cells against DEPTOR knockdown, it indicates that the protein–protein interaction between DEPTOR and mTOR is key to MM viability vs death. In a previous study, we used a yeast two-hybrid screen of a small inhibitor library to identify a compound that inhibited DEPTOR/mTOR binding in yeast. This therapeutic (compound B) also prevented DEPTOR/mTOR binding in MM cells and was selectively cytotoxic to MM cells. We now present a structure–activity relationship (SAR) study around this compound as a follow-up report of this previous work. This study has led to the discovery of five new leads – namely compounds 3g, 3k, 4d, 4e and 4g – all of which have anti-myeloma cytotoxic properties superior to compound B. Due to their targeting of DEPTOR, these compounds activate mTORC1 and selectively induce MM cell apoptosis and cell cycle arrest.
- Published
- 2017
36. DEPTOR is linked to a TORC1-p21 survival proliferation pathway in multiple myeloma cells
- Author
-
Bao Hoang, Joseph Gera, Yonghui Yang, Patrick Frost, Yijiang Shi, Richard A Finn, Alan Lichtenstein, and Carolyne Bardeleben
- Subjects
Cancer Research ,Gene knockdown ,Cell cycle checkpoint ,DEPTOR ,p21 ,AKT ,mTORC1 ,Biology ,Small hairpin RNA ,Downregulation and upregulation ,Genetics ,Cancer research ,Gene silencing ,Multiple Myeloma ,ER stress ,Protein kinase B ,Research Paper - Abstract
We investigated the mechanism by which gene silencing of the mTOR inhibitor, DEPTOR, induces cytoreductive effects on multiple myeloma (MM) cells. DEPTOR knockdown resulted in anti-MM effects in several MM cell lines. Using an inducible shRNA to silence DEPTOR, 8226 MM cells underwent TORC1 activation, downregulation of AKT/SGK activity, apoptosis, cell cycle arrest and senescence. These latter cytotoxic effects were prevented by TORC1 paralysis (Raptor knockdown) but not by over-expression of AKT activity. In addition, DEPTOR knockdown-induced MM death was not associated with activation of the unfolded protein response, suggesting that enhanced ER stress did not play a role. In contrast, DEPTOR knockdown in 8226 cells induced p21 expression, independent of p53, and p21 knockdown prevented all of the cytotoxic effects following DEPTOR silencing. DEPTOR silencing resulted in p21 upregulation in additional MM cell lines. Furthermore, DEPTOR silencing in a murine xenograft model resulted in anti-MM effects associated with p21 upregulation. DEPTOR knockdown also resulted in a decreased expression of p21-targeting miRNAs and transfection of miRNA mimics prevented p21 upregulation and apoptosis following DEPTOR silencing. Use of a shRNA-resistant DEPTOR construct ruled out off-target effects of the shRNA. These results indicate that DEPTOR regulates growth and survival of MM cells via a TORC1/p21 pathway and suggest an involvement of p21-targeted miRNAs.
- Published
- 2014
37. A Deptor Inhibitor Induces Its Degradation with Resulting Anti-Myeloma Cytotoxicity in Vitro and In Vivo
- Author
-
Sara Huerta-Yepez, Patrick Frost, Mario I. Vega, Yijiang Shi, and Alan Lichtenstein
- Subjects
biology ,Bortezomib ,Chemistry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,DEPTOR ,Biochemistry ,medicine.anatomical_structure ,In vivo ,Cell culture ,medicine ,biology.protein ,Cancer research ,Bone marrow ,Cytotoxicity ,Caspase ,Multiple myeloma ,medicine.drug - Abstract
Multiple myeloma (MM) is a hematological disorder characterized by a proliferation of malignant monoclonal plasma cells in the bone marrow (BM) and / or in extramedullary sites. Despite recent progress in OS rates, MM remains an incurable disease and most patients will relapse and require treatment. Deptor is a component of mTOR complexes and a constitutive inhibitor of their activities. It is known that the inhibition of Deptor results in the inhibition of the proliferation and induction of apoptosis in MM cells. In addition, high levels of Deptor are predictive of a poor response to conventional therapies, indicating that Deptor expression are important as a prognostic marker for patients with myeloma and is a possible therapeutic target. Our group previously identified a drug which prevents mTOR-Deptor binding (NSC126405) and induces cellular cytotoxicity in MM (Shi Y, et al 2016). In this study, we developed a new related chemical inhibitor (43 M) capable of inducing the inhibition of the mTOR / Deptor interaction and results in the negative regulation of Deptor that leads to the inhibition of proliferation and induces apoptosis in several MM cell lines. The cytotoxic effect of 43 M is not dependent of caspase activation and induces the activation of p70 and AKT (T308). This leads to the induction of apoptosis in MM cell lines and tumor cells derived from MM patients. The degradation of Deptor induced by 43 M is dependent on the proteasome complex since it was prevented in the presence of MG132. In vivo, 43 M prevents the expression of Deptor in a xenograft tumor, and delayed tumor growth and interestingly, induces the eradication of tumors in 40% of mice in a murine model of MM, without significant toxic implications. Recent studies show that Deptor expression protects MM cells against Bortezomib treatment, suggesting that anti-Deptor drugs can synergize with proteasome inhibitors (PIs). However, the combination of 43 M + Bortezomib was not synergistic, and was antagonistic in vitro. These results are probably due to the prevention of the proteasomal degradation of Deptor, suggesting a possible use of the 43 M inhibitor in MM in the absence of the current PIs. This study describes for the first time the possible role of Deptor as a therapeutic target using a chemical inhibitor capable of degrading and inducing a cytotoxic effect in MM cell lines. In addition, Deptor is reported as an important therapeutic target in an in vivo MM model. Shi Y, Daniels-Wells TR, Frost P, Lee J, Finn RS, Bardeleben C, Penichet ML, Jung ME, Gera J, Lichtenstein A. Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells. Cancer Res. 2016 Oct 1;76(19):5822-5831 Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
38. IL-6-induced Enhancement of c-Myc Translation in Multiple Myeloma Cells
- Author
-
Alan Lichtenstein, Bao Hoang, Yijiang Shi, Patrick Frost, Joseph Gera, and Angelica Benavides
- Subjects
Cell growth ,RNA-binding protein ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,Cell nucleus ,Internal ribosome entry site ,medicine.anatomical_structure ,Transcription (biology) ,Translational regulation ,medicine ,Protein biosynthesis ,Phosphorylation ,Molecular Biology - Abstract
Prior work indicates that IL-6 can stimulate c-Myc expression in multiple myeloma (MM) cells, which is independent of effects on transcription and due to enhanced translation mediated by an internal ribosome entry site in the 5′-UTR of the c-Myc RNA. The RNA-binding protein hnRNP A1 (A1) was also critical to IL-6-stimulated translation. Because A1 shuttles between nucleus and cytoplasm, we investigated whether the ability of IL-6 to enhance Myc translation was mediated by stimulation of A1 shuttling. In MM cell lines and primary specimens, IL-6 increased A1 cytoplasmic localization. In contrast, there was no effect on the total cellular levels of A1. Use of a dominant negative A1 construct, which prevents endogenous A1 from nucleus-to-cytoplasm transit, prevented the ability of IL-6 to enhance Myc internal ribosome entry site activity, Myc protein expression, and MM cell growth. IL-6-stimulated cytoplasmic localization was mediated by alterations in the C-terminal M9 peptide of A1, and this correlated with the ability of IL-6 to induce serine phosphorylation of this domain. A p38 kinase inhibitor prevented IL-6-induced A1 phosphorylation. Thus, IL-6 activates c-Myc translation in MM cells by inducing A1 phosphorylation and cytoplasmic localization in a p38-dependent fashion. These data suggest A1 as a potential therapeutic target in MM.
- Published
- 2011
39. Effect of autophagy on multiple myeloma cell viability
- Author
-
Patrick Frost, Bao Hoang, Alan Lichtenstein, Yijiang Shi, and Angelica Benavides
- Subjects
Cancer Research ,Small interfering RNA ,Antifungal Agents ,Thapsigargin ,Immunoblotting ,Antineoplastic Agents ,Apoptosis ,Biology ,Bortezomib ,Antimalarials ,chemistry.chemical_compound ,Autophagy ,Tumor Cells, Cultured ,medicine ,Humans ,Viability assay ,Enzyme Inhibitors ,RNA, Small Interfering ,Multiple myeloma ,Cell Proliferation ,Sirolimus ,Adenine ,Membrane Proteins ,Chloroquine ,medicine.disease ,Boronic Acids ,Cell biology ,Microscopy, Fluorescence ,Oncology ,chemistry ,Pyrazines ,Cancer research ,Proteasome inhibitor ,Beclin-1 ,Drug Therapy, Combination ,Apoptosis Regulatory Proteins ,Multiple Myeloma ,medicine.drug - Abstract
Because accumulation of potentially toxic malfolded protein may be extensive in immunoglobulin-producing multiple myeloma (MM) cells, we investigated the phenomenon of autophagy in myeloma, a physiologic process that can protect against malfolded protein under some circumstances. Autophagy in MM cell lines that express and secrete immunoglobulin and primary specimens was significantly increased by treatment with the endoplasmic reticulum stress–inducing agent thapsigargin, the mammalian target of rapamycin inhibitor rapamycin, and the proteasome inhibitor bortezomib. Inhibition of basal autophagy in these cell lines and primary cells by use of the inhibitors 3-methyladenine and chloroquine resulted in a cytotoxic effect that was associated with enhanced apoptosis. Use of small interfering RNA to knock down expression of beclin-1, a key protein required for autophagy, also inhibited viable recovery of MM cells. Because the data suggested that autophagy protected MM cell viability, we predicted that autophagy inhibitors would synergize with bortezomib for enhanced antimyeloma effects. However, the combination of these drugs resulted in an antagonistic response. In contrast, the autophagy inhibitor 3-methyladenine did synergize with thapsigargin for an enhanced cytotoxic response. These data suggest that autophagy inhibitors have therapeutic potential in myeloma but caution against combining such drugs with bortezomib. [Mol Cancer Ther 2009;8(7):1974–84]
- Published
- 2009
40. SGK Kinase Activity in Multiple Myeloma Cells Protects against ER Stress Apoptosis via a SEK-Dependent Mechanism
- Author
-
Bao Hoang, Alan Lichtenstein, Yijiang Shi, Veena Mysore, Patrick Frost, and Carolyne Bardeleben
- Subjects
0301 basic medicine ,Cancer Research ,MAP2K4 ,Apoptosis ,Protein Serine-Threonine Kinases ,Immediate early protein ,Article ,Immediate-Early Proteins ,Bortezomib ,03 medical and health sciences ,Mice ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Kinase activity ,Molecular Biology ,Multiple myeloma ,Chemistry ,Kinase ,Tunicamycin ,medicine.disease ,Endoplasmic Reticulum Stress ,Xenograft Model Antitumor Assays ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Unfolded protein response ,Thapsigargin ,Female ,Multiple Myeloma ,medicine.drug - Abstract
To assess the role of the serum and glucocorticoid-regulated kinase (SGK) kinase in multiple myeloma, we ectopically expressed wild type or a phosphomimetic version of SGK into multiple myeloma cell lines. These cells were specifically resistant to the ER stress inducers tunicamycin, thapsigargin, and bortezomib. In contrast, there was no alteration of sensitivity to dexamethasone, serum starvation, or mTORC inhibitors. Mining of genomic data from a public database indicated that low baseline SGK expression in multiple myeloma patients correlated with enhanced ability to undergo a complete response to subsequent bortezomib treatment and a longer time to progression and overall survival following treatment. SGK overexpressing multiple myeloma cells were also relatively resistant to bortezomib in a murine xenograft model. Parental/control multiple myeloma cells demonstrated a rapid upregulation of SGK expression and activity (phosphorylation of NDRG-1) during exposure to bortezomib and an SGK inhibitor significantly enhanced bortezomib-induced apoptosis in cell lines and primary multiple myeloma cells. In addition, a multiple myeloma cell line selected for bortezomib resistance demonstrated enhanced SGK expression and SGK activity. Mechanistically, SGK overexpression constrained an ER stress–induced JNK proapoptotic pathway and experiments with a SEK mutant supported the notion that SGK's protection against bortezomib was mediated via its phosphorylation of SEK (MAP2K4) which abated SEK/JNK signaling. These data support a role for SGK inhibitors in the clinical setting for myeloma patients receiving treatment with ER stress inducers like bortezomib. Implications: Enhanced SGK expression and activity in multiple myeloma cells contributes to resistance to ER stress, including bortezomib challenge. Mol Cancer Res; 14(4); 397–407. ©2016 AACR . This article is featured in Highlights of This Issue, [p. 313][1] [1]: /lookup/volpage/14/313?iss=4
- Published
- 2015
41. AKT activity regulates the ability of mTOR inhibitors to prevent angiogenesis and VEGF expression in multiple myeloma cells
- Author
-
Bao Hoang, Patrick Frost, Yijiang Shi, and Alan Lichtenstein
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,Angiogenesis ,Transplantation, Heterologous ,Antineoplastic Agents ,Mice, SCID ,Biology ,Neovascularization ,Mice ,chemistry.chemical_compound ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Sirolimus ,Neovascularization, Pathologic ,Cell growth ,TOR Serine-Threonine Kinases ,RPTOR ,Actins ,Gene Expression Regulation, Neoplastic ,Oncogene Protein v-akt ,Vascular endothelial growth factor ,Internal ribosome entry site ,chemistry ,Cancer research ,medicine.symptom ,Multiple Myeloma ,Protein Kinases - Abstract
We recently demonstrated that the mammalian target of rapamycin (mTOR) inhibitor, CCI-779, curtailed the growth of a subcutaneous challenge of multiple myeloma (MM) cells in immunodeficient mice. This antitumor effect was associated with prevention of cell proliferation, induction of apoptosis and inhibition of angiogenesis. Interestingly, myeloma tumors with heightened AKT activation were particularly sensitive to a CCI-779-induced antitumor response. To investigate whether part of the differential sensitivity was due to an AKT-regulated effect on angiogenesis, we compared the effects of mTOR inhibitors against isogenic MM cell lines that only differ by their degree of AKT activity. In this model, heightened AKT activity significantly sensitized MM cells to the following inhibitory effects of mTOR inhibition: angiogenesis in vivo, vascular endothelial growth factor (VEGF) expression in vitro and in vivo and VEGF translation (but not transcription). Assessment of p70S6 kinase activity indicated that rapamycin induced comparable mTOR inhibition in both cell lines suggesting that an adverse effect on VEGF cap-dependent translation would be comparable. Internal ribosome entry site (IRES)-mediated cap-independent translation is a salvage pathway for protein expression when mTOR is inhibited, so we analyzed a possible regulatory role of AKT on VEGF IRES activity. We found that elevated AKT activity inhibited VEGF IRES function. These results support a mechanism whereby AKT prevents VEGF IRES activity in myeloma cells during mTOR inhibition resulting in a more complete abrogation of VEGF translation, and ultimately, angiogenesis.
- Published
- 2006
42. Oncogenic RAS mutations in myeloma cells selectively induce cox-2 expression, which participates in enhanced adhesion to fibronectin and chemoresistance
- Author
-
Alan Lichtenstein, Patrick Frost, Lee Goodglick, Sherven Sharma, Huajun Yan, Li Zhu, Sanjai Sharma, Steven M. Dubinett, Bao Hoang, and Yijiang Shi
- Subjects
Stromal cell ,medicine.medical_treatment ,Immunology ,medicine.disease_cause ,Biochemistry ,Dinoprostone ,Proto-Oncogene Proteins p21(ras) ,Cell Line, Tumor ,Cell Adhesion ,medicine ,Humans ,Cell adhesion ,Neoplasia ,biology ,Cell Biology ,Hematology ,Transfection ,Fibronectins ,Gene Expression Regulation, Neoplastic ,Fibronectin ,Genes, ras ,Cytokine ,Cyclooxygenase 2 ,Drug Resistance, Neoplasm ,Cell culture ,Mutation ,biology.protein ,Cancer research ,Stromal Cells ,Multiple Myeloma ,Carcinogenesis - Abstract
Oncogenic RAS expression occurs in up to 40% of multiple myeloma (MM) cases and correlates with aggressive disease. Since activated RAS induces cyclooxygenase-2 (cox-2) expression in other tumor models, we tested a role for cox-2 in mutant RAS–containing MM cells. We used the ANBL-6 isogenic MM cell lines in which the IL-6–dependent parental line becomes cytokine independent following transfection with mutated N-RAS or K-RAS. Both mutated N-RAS– and K-RAS–expressing ANBL-6 cells demonstrated a selective up-regulation of cox-2 expression and enhanced secretion of PGE2, a product of cox-2. Furthermore, in 3 primary marrow specimens, which contained MM cells expressing mutated RAS, 15% to 40% of tumor cells were positive for cox-2 expression by immunohistochemistry. We used cox-2 inhibitors, NS398 and celecoxib, and neutralizing anti-PGE2 antibody to test whether cox-2/PGE2 was involved in the aggressive phenotype of MM ANBL-6 cells containing mutated RAS. Although these interventions had no effect on IL-6–independent growth or adhesion to marrow stromal cells, they significantly inhibited the enhanced binding of mutant RAS– containing MM cells to fibronectin and the enhanced resistance to melphalan. These results indicate a selective induction of cox-2 in MM cells containing RAS mutations, which results in heightened binding to extracellular matrix protein and chemotherapeutic drug resistance.
- Published
- 2006
43. Mechanism by Which Mammalian Target of Rapamycin Inhibitors Sensitize Multiple Myeloma Cells to Dexamethasone-Induced Apoptosis
- Author
-
Sanjai Sharma, Joseph Gera, Myrna Fisher, Patrick Frost, Huajun Yan, Yijiang Shi, Alan Lichtenstein, and Bao Hoang
- Subjects
Cancer Research ,Programmed cell death ,MAP Kinase Kinase 4 ,Apoptosis ,Cell Cycle Proteins ,Mice, SCID ,Biology ,p38 Mitogen-Activated Protein Kinases ,Dexamethasone ,Mice ,Receptors, Glucocorticoid ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein kinase A ,Protein Kinase Inhibitors ,Protein kinase B ,Sensitization ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Sirolimus ,TOR Serine-Threonine Kinases ,RPTOR ,Ribosomal Protein S6 Kinases, 70-kDa ,Drug Synergism ,Phosphoproteins ,XIAP ,Focal Adhesion Kinase 2 ,medicine.anatomical_structure ,Oncology ,Cancer research ,bcl-Associated Death Protein ,Carrier Proteins ,Multiple Myeloma ,Protein Kinases - Abstract
Mammalian target of rapamycin (mTOR) inhibitors curtail cap-dependent translation. However, they can also induce post-translational modifications of proteins. We assessed both effects to understand the mechanism by which mTOR inhibitors like rapamycin sensitize multiple myeloma cells to dexamethasone-induced apoptosis. Sensitization was achieved in multiple myeloma cells irrespective of their PTEN or p53 status, enhanced by activation of AKT, and associated with stimulation of both intrinsic and extrinsic pathways of apoptosis. The sensitizing effect was not due to post-translational modifications of the RAFTK kinase, Jun kinase, p38 mitogen-activated protein kinase, or BAD. Sensitization was also not associated with a rapamycin-mediated increase in glucocorticoid receptor reporter expression. However, when cap-dependent translation was prevented by transfection with a mutant 4E-BP1 construct, which is resistant to mTOR-induced phosphorylation, cells responded to dexamethasone with enhanced apoptosis, mirroring the effect of coexposure to rapamycin. Thus, sensitization is mediated by inhibition of cap-dependent translation. A high-throughput screening for translational efficiency identified several antiapoptotic proteins whose translation was inhibited by rapamycin. Immunoblot assay confirmed rapamycin-induced down-regulated expressions of XIAP, CIAP1, HSP-27, and BAG-3, which may play a role in the sensitization to apoptosis. Studies in a xenograft model showed synergistic in vivo antimyeloma effects when dexamethasone was combined with the mTOR inhibitor CCI-779. Synergistic effects were associated with an enhanced multiple myeloma cell apoptosis in vivo. This study supports the strategy of combining dexamethasone with mTOR inhibitors in multiple myeloma and identifies a mechanism by which the synergistic effect is achieved. (Cancer Res 2006; 66(4): 2305-13)
- Published
- 2006
44. Mammalian target of rapamycin inhibitors activate the AKT kinase in multiple myeloma cells by up-regulating the insulin-like growth factor receptor/insulin receptor substrate-1/phosphatidylinositol 3-kinase cascade
- Author
-
Patrick Frost, Huajun Yan, Yijiang Shi, Alan Lichtenstein, and Joseph Gera
- Subjects
Cancer Research ,Insulin Receptor Substrate Proteins ,Apoptosis ,Mice, SCID ,Receptor, IGF Type 1 ,Bortezomib ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Insulin-Like Growth Factor I ,Protein Kinase Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Sirolimus ,Antibiotics, Antineoplastic ,biology ,Chemistry ,TOR Serine-Threonine Kinases ,Phosphoproteins ,Boronic Acids ,Xenograft Model Antitumor Assays ,IRS1 ,Enzyme Activation ,Insulin receptor ,Oncology ,Pyrazines ,biology.protein ,Cancer research ,Phosphorylation ,Multiple Myeloma ,Protein Kinases ,Proto-Oncogene Proteins c-akt ,medicine.drug - Abstract
Mammalian target of rapamycin (mTOR) inhibitors, such as rapamycin and CCI-779, have shown preclinical potential as therapy for multiple myeloma. By inhibiting expression of cell cycle proteins, these agents induce G1 arrest. However, by also inhibiting an mTOR-dependent serine phosphorylation of insulin receptor substrate-1 (IRS-1), they may enhance insulin-like growth factor-I (IGF-I) signaling and downstream phosphatidylinositol 3-kinase (PI3K)/AKT activation. This may be a particular problem in multiple myeloma where IGF-I-induced activation of AKT is an important antiapoptotic cascade. We, therefore, studied AKT activation in multiple myeloma cells treated with mTOR inhibitors. Rapamycin enhanced basal AKT activity, AKT phosphorylation, and PI3K activity in multiple myeloma cells and prolonged activation of AKT induced by exogenous IGF-I. CCI-779, used in a xenograft model, also resulted in multiple myeloma cell AKT activation in vivo. Blockade of IGF-I receptor function prevented rapamycin's activation of AKT. Furthermore, rapamycin prevented serine phosphorylation of IRS-1, enhanced IRS-1 association with IGF-I receptors, and prevented IRS-1 degradation. Although similarly blocking IRS-1 degradation, proteasome inhibitors did not activate AKT. Thus, mTOR inhibitors activate PI3-K/AKT in multiple myeloma cells; activation depends on basal IGF-R signaling; and enhanced IRS-1/IGF-I receptor interactions secondary to inhibited IRS-1 serine phosphorylation may play a role in activation of the cascade. In cotreatment experiments, rapamycin inhibited myeloma cell apoptosis induced by PS-341. These results provide a caveat for future use of mTOR inhibitors in myeloma patients if they are to be combined with apoptosis-inducing agents.
- Published
- 2005
45. Cyclin D1 and c-myc Internal Ribosome Entry Site (IRES)-dependent Translation Is Regulated by AKT Activity and Enhanced by Rapamycin through a p38 MAPK- and ERK-dependent Pathway
- Author
-
Hong Wu, Joseph Gera, Anushree Sharma, Yijiang Shi, and Alan Lichtenstein
- Subjects
MAPK/ERK pathway ,Genes, myc ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Biology ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,Cell Line ,Cyclin D1 ,Proto-Oncogene Proteins ,Protein biosynthesis ,Humans ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Sirolimus ,Kinase ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,fungi ,PTEN Phosphohydrolase ,Translation (biology) ,Cell Biology ,Phosphoric Monoester Hydrolases ,Cell biology ,Internal ribosome entry site ,Protein Biosynthesis ,5' Untranslated Regions ,Protein Kinases ,Proto-Oncogene Proteins c-akt ,Ribosomes ,Cyclin-Dependent Kinase Inhibitor p27 ,Signal Transduction - Abstract
The macrolide antibiotic rapamycin inhibits the mammalian target of rapamycin protein (mTOR) kinase resulting in the global inhibition of cap-dependent protein synthesis, a blockade in ribosome component biosynthesis, and G1 cell cycle arrest. G1 arrest may occur by inhibiting the protein synthesis of critical factors required for cell cycle progression. Hypersensitivity to mTOR inhibitors has been demonstrated in cells having elevated levels of AKT kinase activity, whereas cells containing quiescent AKT activity are relatively resistant. Our previous data suggest that low AKT activity induces resistance by allowing continued cap-independent protein synthesis of cyclin D1 and c-Myc proteins. In support of this notion, the current study demonstrates that the human cyclin D1 mRNA 5' untranslated region contains an internal ribosome entry site (IRES) and that both this IRES and the c-myc IRES are negatively regulated by AKT activity. Furthermore, we show that cyclin D1 and c-myc IRES function is enhanced following exposure to rapamycin and requires both p38 MAPK and RAF/MEK/ERK signaling, as specific inhibitors of these pathways reduce IRES-mediated translation and protein levels under conditions of quiescent AKT activity. Thus, continued IRES-mediated translation initiation may permit cell cycle progression upon mTOR inactivation in cells in which AKT kinase activity is relatively low.
- Published
- 2005
46. Fructose feeding and intermittent hypoxia affect ventilatory responsiveness to hypoxia and hypercapnia in rats
- Author
-
Curtis K. Kost, Evelyn H. Schlenker, Douglas S. Martin, Joni Wipf, and Yijiang Shi
- Subjects
Male ,medicine.medical_specialty ,Physiology ,medicine.medical_treatment ,Administration, Oral ,Hemodynamics ,Blood Pressure ,Fructose ,Severity of Illness Index ,Hypercapnia ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Heart Rate ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Hypoxia ,Pancreatic hormone ,Pulmonary Gas Exchange ,business.industry ,Insulin ,Intermittent hypoxia ,Hypoxia (medical) ,Adaptation, Physiological ,Rats ,Endocrinology ,Blood pressure ,chemistry ,medicine.symptom ,Pulmonary Ventilation ,business - Abstract
We hypothesized that, in male rats, 10% fructose in drinking water would depress ventilatory responsiveness to acute hypoxia (10% O2 in N2) and hypercapnia (5% CO2 in O2) that would be depressed further by exposure to intermittent hypoxia. Minute ventilation (V̇e) in air and in response to acute hypoxia and hypercapnia was evaluated in 10 rats before fructose feeding (FF), during 6 wk of FF, and after FF was removed for 2 wk. During FF, five rats were exposed to intermittent air and five to intermittent hypoxia for 13 days. Six rats given tap water acted as control and were exposed to intermittent air and subsequently intermittent hypoxia. In FF rats, plasma insulin levels increased threefold in the rats exposed to intermittent hypoxia and during washout returned to levels observed in rats exposed to intermittent air. During FF, ventilatory responsiveness to acute hypoxia was depressed because of decreased tidal volume (Vt) responsiveness. During washout, V̇e decreased as a result of decreased Vt and frequency of breathing, and the ventilatory responsiveness to hypoxia in intermittent hypoxia rats did not recover. In all rats, the ventilatory responses to hypercapnia were decreased during FF and recovered after washout because of an increased Vt responsiveness. In the control group, hypoxic responsiveness was not depressed after intermittent hypoxia and was augmented after washout. Thus FF attenuated the ventilatory responsiveness of conscious rats to hypoxia and hypercapnia. Intermittent hypoxia interacted with FF to increase insulin levels and depress ventilatory responses to acute hypoxia that remained depressed during washout.
- Published
- 2004
47. AKT Activity Determines Sensitivity to Mammalian Target of Rapamycin (mTOR) Inhibitors by Regulating Cyclin D1 and c-myc Expression
- Author
-
Charles L. Sawyers, Joseph Gera, Yijiang Shi, Ingo K. Mellinghoff, Jung-hsin Hsu, Alan Lichtenstein, Matthew Rettig, and Chris Tran
- Subjects
Translational efficiency ,Genes, myc ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,Cell Line ,Proto-Oncogene Proteins c-myc ,Cyclin D1 ,Proto-Oncogene Proteins ,Polysome ,Humans ,Protein Kinase Inhibitors ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Sirolimus ,TOR Serine-Threonine Kinases ,RPTOR ,G1 Phase ,Cell Biology ,Transfection ,Molecular biology ,Cell biology ,Gene Expression Regulation ,Protein Kinases ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Prior work demonstrates that AKT activity regulates sensitivity of cells to G(1) arrest induced by mammalian target of rapamycin (mTOR) inhibitors such as rapamycin and CCI-779. To investigate this, a novel high-throughput microarray polysome analysis was performed to identify genes whose mRNA translational efficiency was differentially affected following mTOR inhibition. The analysis also allowed the assessment of steady-state transcript levels. We identified two transcripts, cyclin D1 and c-myc, which exhibited differential expression in an AKT-dependent manner: High levels of activated AKT resulted in rapamycin-induced down-regulation of expression, whereas low levels resulted in up-regulation of expression. To ectopically express these proteins we exploited the finding that the p27(kip1) mRNA was efficiently translated in the face of mTOR inhibition irrespective of AKT activity. Thus, the p27(kip1) 5'-untranslated region was fused to the cyclin D1 and c-myc coding regions and these constructs were expressed in cells. In transfected cells, expression of cyclin D1 or c-myc was not decreased by rapamycin. Most importantly, this completely converted sensitive cells to a phenotype resistant to G(1) arrest. Furthermore, the AKT-dependent differential expression patterns of these two genes was also observed in a mouse xenograft model following in vivo treatment with CCI-779. These results identify two critical downstream molecular targets whose expression is regulated by AKT activity and whose down-regulation is required for rapamycin/CCI-779 sensitivity.
- Published
- 2004
48. Signal Pathways Involved in Activation of p70S6K and Phosphorylation of 4E-BP1 following Exposure of Multiple Myeloma Tumor Cells to Interleukin-6
- Author
-
Joseph Gera, Jung-hsin Hsu, Yijiang Shi, Alan Lichtenstein, and Liping Hu
- Subjects
MAPK/ERK pathway ,MAP Kinase Signaling System ,Cell Cycle Proteins ,P70-S6 Kinase 1 ,mTORC1 ,Protein Serine-Threonine Kinases ,Biochemistry ,Wortmannin ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Proto-Oncogene Proteins ,Tumor Cells, Cultured ,Humans ,Enzyme Inhibitors ,Insulin-Like Growth Factor I ,Phosphorylation ,Kinase activity ,Molecular Biology ,Protein kinase B ,Adaptor Proteins, Signal Transducing ,Flavonoids ,Sirolimus ,Interleukin-6 ,Chemistry ,Ribosomal Protein S6 Kinases ,Cell Biology ,Phosphoproteins ,Recombinant Proteins ,Cell biology ,Androstadienes ,Enzyme Activation ,Repressor Proteins ,Kinetics ,Cancer research ,Signal transduction ,Carrier Proteins ,Multiple Myeloma ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Interleukin-6 (IL-6) is a prominent tumor growth factor for malignant multiple myeloma cells. In addition to its known activation of the Janus tyrosine kinase-STAT and RAS-MEK-ERK pathways, recent work suggests that IL-6 can also activate the phosphatidylinositol 3-kinase (PI3-K)/AKT kinase pathway in myeloma cells. Because activation of the PI3-K/AKT as well as RAS-MEK-ERK pathways may result in downstream stimulation of the p70(S6K) (p70) and phosphorylation of the 4E-BP1 translational repressor, we assessed these potential molecular targets in IL-6-treated myeloma cells. IL-6 rapidly activated p70 kinase activity and p70 phosphorylation. Activation was inhibited by wortmannin, rapamycin, and the ERK inhibitors PD98059 and UO126, as well as by a dominant negative mutant of AKT. The concurrent requirements for both ERK and PI3-K/AKT appeared to be a result of their ability to phosphorylate p70 on different residues. In contrast, IL-6-induced phosphorylation of 4E-BP1 was inhibited by rapamycin, wortmannin, and dominant negative AKT but ERK inhibitors had no effect, indicating ERK function was dispensable. In keeping with these data, a dominant active AKT mutant was sufficient to induce 4E-BP1 phosphorylation but could not by itself activate p70 kinase activity. Prevention of IL-6-induced p70 activation and 4E-BP1 phosphorylation by the mammalian target of rapamycin inhibitors rapamycin and CCI-779 resulted in inhibition of IL-6-induced myeloma cell growth. These results indicate that both ERK and PI3-K/AKT pathways are required for optimal IL-6-induced p70 activity, but PI3-K/AKT is sufficient for 4E-BP1 phosphorylation. Both effects are mediated via mammalian target of rapamycin function, and, furthermore, these effects are critical for IL-6-induced tumor cell growth.
- Published
- 2002
49. Brief co-incubation of sperm and oocytes for in vitro fertilization techniques
- Author
-
Jun Li, Shangwei Li, Yijiang Shi, Zhongying Huang, Jing Yan, and Li Wang
- Subjects
Male ,medicine.medical_specialty ,Time Factors ,Pregnancy Rate ,medicine.medical_treatment ,Fertilization in Vitro ,Cochrane Library ,Insemination ,law.invention ,Miscarriage ,Randomized controlled trial ,law ,Pregnancy ,Medicine ,Humans ,Pharmacology (medical) ,Randomized Controlled Trials as Topic ,Gynecology ,Sperm-Ovum Interactions ,In vitro fertilisation ,business.industry ,medicine.disease ,Coculture Techniques ,Culture Media ,Abortion, Spontaneous ,Pregnancy rate ,Female ,business ,Reactive Oxygen Species ,Embryo quality - Abstract
Background The in vitro fertilization (IVF) technique is commonly used and is the only treatment option for a proportion of infertile couples. To obtain better outcomes of IVF, it is important to enhance embryo quality by optimizing IVF techniques. In IVF procedures, oocytes and sperm are routinely co-incubated overnight, which may expose oocytes and zygotes to suboptimal culture conditions with increased reactive oxygen species (ROS) produced by sperm in this long term culture. As an attempt to avoid possible detrimental effects on the oocytes from long exposure to sperm, the brief co-incubation insemination protocol was developed. However, despite a number of studies in this area, it is unclear whether brief co-incubation improves the IVF outcomes compared with the standard overnight insemination protocol. Objectives This Cochrane review aimed to determine whether brief co-incubation of sperm and oocytes improves outcomes compared with the standard overnight insemination protocol for women undergoing IVF. Search methods We searched the Cochrane Menstrual Disorders and Subfertility Group Register (14 June 2012), Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2012, 1st quarter), MEDLINE (1948 to 14 June 2012), EMBASE (1989 to 14 June 2012), PsycINFO (1806 to 14 June 2012) and CINAHL (1980 to 26 July 2012). In addition, we searched trials registers, reference lists of articles, conference proceedings (American Society for Reproductive Medicine (ASRM), European Society of Human Reproduction and Embryology (ESHRE)) and contacted experts in the field. Selection criteria We included randomized controlled trials (RCTs) comparing brief co-incubation of gametes with the standard overnight insemination protocol. Data collection and analysis Two review authors independently assessed studies for inclusion and trial quality, and extracted data. Disagreements were resolved by discussion with a third author. Statistical analysis was performed using RevMan software. Main results Eight RCTs with 733 women in total that compared brief co-incubation and the standard insemination protocol were included. Live birth was not reported in the included studies. For ongoing pregnancy rate, there were 127 ongoing pregnancies in two trials including 426 women. The low quality evidence showed that brief co-incubation was associated with an increased ongoing pregnancy rate compared to the standard insemination protocol (pooled odds ratio (OR) 2.42, 95% confidence interval (CI) 1.55 to 3.77; P < 0.0001, I2 = 0%). Measuring clinical pregnancy rate, there were 93 clinical pregnancies in three trials including 372 women. The low quality evidence showed that brief co-incubation was associated with a significantly higher clinical pregnancy rate than the overnight insemination protocol (pooled OR 2.36, 95% CI 1.45 to 3.85; P = 0.0006, I2 = 0%). For the miscarriage rate, there were six miscarriages in one trial including 167 women. This low quality evidence suggested no significant difference in the odds of miscarriage between brief co-incubation and standard insemination (OR 1.98, 95% CI 0.35 to 11.09; P = 0.44). Authors' conclusions This review has provided evidence that brief co-incubation of sperm and oocytes may improve the ongoing pregnancy and clinical pregnancy rates for infertile women undergoing IVF cycles. More RCTs are required to assess whether brief co-incubation would contribute to a higher live birth rate and a lower miscarriage rate compared to the standard overnight insemination protocol.
- Published
- 2013
50. The PP242 mammalian target of rapamycin (mTOR) inhibitor activates extracellular signal-regulated kinase (ERK) in multiple myeloma cells via a target of rapamycin complex 1 (TORC1)/eukaryotic translation initiation factor 4E (eIF-4E)/RAF pathway and activation is a mechanism of resistance
- Author
-
Yonghui Yang, Patrick Frost, Joseph Gera, Alan Lichtenstein, Angelica Benavides, Bao Hoang, and Yijiang Shi
- Subjects
MAPK/ERK pathway ,Indoles ,Cell Survival ,Apoptosis ,mTORC1 ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,Biochemistry ,Phosphatidylinositol 3-Kinases ,Catalytic Domain ,Cell Line, Tumor ,medicine ,Humans ,Extracellular Signal-Regulated MAP Kinases ,Molecular Biology ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Glutathione Transferase ,Sirolimus ,TOR Serine-Threonine Kinases ,Proteins ,Cell Biology ,Cell biology ,Enzyme Activation ,Eukaryotic Initiation Factor-4E ,Purines ,Multiprotein Complexes ,Phosphorylation ,Signal transduction ,Multiple Myeloma ,medicine.drug ,Signal Transduction - Abstract
Activation of PI3-K-AKT and ERK pathways is a complication of mTOR inhibitor therapy. Newer mTOR inhibitors (like pp242) can overcome feedback activation of AKT in multiple myeloma (MM) cells. We, thus, studied if feedback activation of ERK is still a complication of therapy with such drugs in this tumor model. PP242 induced ERK activation in MM cell lines as well as primary cells. Surprisingly, equimolar concentrations of rapamycin were relatively ineffective at ERK activation. Activation was not correlated with P70S6kinase inhibition nor was it prevented by PI3-kinase inhibition. ERK activation was prevented by MEK inhibitors and was associated with concurrent stimulation of RAF kinase activity but not RAS activation. RAF activation correlated with decreased phosphorylation of RAF at Ser-289, Ser-296, and Ser-301 inhibitory residues. Knockdown studies confirmed TORC1 inhibition was the key proximal event that resulted in ERK activation. Furthermore, ectopic expression of eIF-4E blunted pp242-induced ERK phosphorylation. Since pp242 was more potent than rapamycin in causing sequestering of eIF-4E, a TORC1/4E-BP1/eIF-4E-mediated mechanism of ERK activation could explain the greater effectiveness of pp242. Use of MEK inhibitors confirmed ERK activation served as a mechanism of resistance to the lethal effects of pp242. Thus, although active site mTOR inhibitors overcome AKT activation often seen with rapalog therapy, feedback ERK activation is still a problem of resistance, is more severe than that seen with use of first generation rapalogs and is mediated by a TORC1- and eIF-4E-dependent mechanism ultimately signaling to RAF.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.