48 results on '"Frost, Patrick"'
Search Results
2. Dynamic cross-interaction between two adjacent rocking blocks
- Author
-
Frost, Patrick and Cacciola, Pierfrancesco
- Published
- 2024
- Full Text
- View/download PDF
3. Rocking of rigid blocks standing on a horizontally-moving compliant base
- Author
-
Frost, Patrick and Cacciola, Pierfrancesco
- Published
- 2023
- Full Text
- View/download PDF
4. AcidoCEST MRI Evaluates the Bone Microenvironment in Multiple Myeloma
- Author
-
Lombardi, Alecio F., Wong, Jonathan H., High, Rachel, Ma, Yajun, Jerban, Saeed, Tang, Qingbo, Du, Jiang, Frost, Patrick, Pagel, Mark D., and Chang, Eric Y.
- Published
- 2021
- Full Text
- View/download PDF
5. Acidification of intracellular pH in MM tumor cells overcomes resistance to hypoxia-mediated apoptosis in vitro and in vivo.
- Author
-
Gastelum, Gilberto, Kraut, Jeffry, Veena, Mysore, Baibussinov, Alisher, Lamb, Christopher, Lyons, Kylee, Chang, Eric Y., and Frost, Patrick
- Subjects
CARBONIC anhydrase inhibitors ,CANCER cells ,APOPTOSIS ,TUMOR growth ,PLASMACYTOMA ,BONE marrow ,BENZENESULFONAMIDES - Abstract
Introduction: Multiple myeloma (MM) is an incurable cancer of malignant plasma cells that engraft in the bone marrow (BM). It is more than likely that the poorly investigated physical parameters of hypoxia and pH in the tumor microenvironment (TME) is critical for MM survival. Here, we explore the effects of a hypoxic environment on pH regulation and its role in MM survival. Methods: We used in vitro models of MM, in which the culturing medium was modified to specific pH and pO2 levels and then measured the effects on cell survival that was correlated with changes in intracellular (pHi) and extracellular pH (pHe). In a MM xenograft model, we used PET/CT to study hypoxia-mediated effects on tumor growth. Results: Hypoxia-mediated apoptosis of MM cells is correlated with acidic intracellular pHi (less than < 6.6) that is dependent on HIF activity. Using a polyamide HIF responsive element binding compound, a carbonic anhydrase inhibitor (acetazolamide), and an NHE-1 inhibitor (amiloride) acidified the pHi and lead to cell death. In contrast, treatment of cells with an alkalization agent, Nalactate, rescued these cells by increasing the pHi (pH > 6.6). Finally, treatment of mice with acetazolamide decreased cell growth in the tumor nodules. Discussion: Targeting hypoxia and HIF have been proposed as an anti-tumor therapy but the clinical efficacy of such strategies are modest. We propose that targeting the pHi may be more effective at treating cancers within a hypoxic TME. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Targeting TORC2 in multiple myeloma with a new mTOR kinase inhibitor
- Author
-
Hoang, Bao, Frost, Patrick, Shi, Yijiang, Belanger, Eileen, Benavides, Angelica, Pezeshkpour, Gholam, Cappia, Susanna, Guglielmelli, Tommasina, Gera, Joseph, and Lichtenstein, Alan
- Published
- 2010
- Full Text
- View/download PDF
7. Interleukin-6 activates phosphoinositol-3′ kinase in multiple myeloma tumor cells by signaling through RAS-dependent and, separately, through p85-dependent pathways
- Author
-
Hsu, Jung-hsin, Shi, Yijiang, Frost, Patrick, Yan, Huajun, Hoang, Bao, Sharma, Sanjai, Gera, Joseph, and Lichtenstein, Alan
- Published
- 2004
- Full Text
- View/download PDF
8. Dietary β-glucan stimulate complement and C-reactive protein acute phase responses in common carp (Cyprinus carpio) during an Aeromonas salmonicida infection
- Author
-
Pionnier, Nicolas, Falco, Alberto, Miest, Joanna, Frost, Patrick, Irnazarow, Ilgiz, Shrive, Annette, and Hoole, Dave
- Published
- 2013
- Full Text
- View/download PDF
9. Reduced inflammatory response to Aeromonas salmonicida infection in common carp (Cyprinus carpio L.) fed with β-glucan supplements
- Author
-
Falco, Alberto, Frost, Patrick, Miest, Joanna, Pionnier, Nicolas, Irnazarow, Ilgiz, and Hoole, David
- Published
- 2012
- Full Text
- View/download PDF
10. Molecular cloning and expression of two β-defensin and two mucin genes in common carp (Cyprinus carpio L.) and their up-regulation after β-glucan feeding
- Author
-
van der Marel, Maria, Adamek, Mikołaj, Gonzalez, Santiago F., Frost, Patrick, Rombout, Jan H.W.M., Wiegertjes, Geert F., Savelkoul, Huub F.J., and Steinhagen, Dieter
- Published
- 2012
- Full Text
- View/download PDF
11. In vivo antitumor effects of the mTOR inhibitor CCI-779 against human multiple myeloma cells in a xenograft model
- Author
-
Frost, Patrick, Moatamed, Farhad, Hoang, Bao, Shi, Yijiang, Gera, Joseph, Yan, Huajun, Frost, Philip, Gibbons, Jay, and Lichtenstein, Alan
- Published
- 2004
- Full Text
- View/download PDF
12. Can Targeting Hypoxia-Mediated Acidification of the Bone Marrow Microenvironment Kill Myeloma Tumor Cells?
- Author
-
Gastelum, Gilberto, Veena, Mysore, Lyons, Kylee, Lamb, Christopher, Jacobs, Nicole, Yamada, Alexandra, Baibussinov, Alisher, Sarafyan, Martin, Shamis, Rebeka, Kraut, Jeffry, and Frost, Patrick
- Subjects
BONE marrow ,PLASMA cell diseases ,VASCULAR endothelial growth factors ,MULTIPLE myeloma ,CANCER cell physiology ,CANCER cells - Abstract
Multiple myeloma (MM) is an incurable cancer arising from malignant plasma cells that engraft in the bone marrow (BM). The physiology of these cancer cells within the BM microenvironment (TME) plays a critical role in MM development. These processes may be similar to what has been observed in the TME of other (non-hematological) solid tumors. It has been long reported that within the BM, vascular endothelial growth factor (VEGF), increased angiogenesis and microvessel density, and activation of hypoxia-induced transcription factors (HIF) are correlated with MM progression but despite a great deal of effort and some modest preclinical success the overall clinical efficacy of using anti-angiogenic and hypoxia-targeting strategies, has been limited. This review will explore the hypothesis that the TME of MM engrafted in the BM is distinctly different from non-hematological-derived solid tumors calling into question how effective these strategies may be against MM. We further identify other hypoxia-mediated effectors, such as hypoxia-mediated acidification of the TME, oxygen-dependent metabolic changes, and the generation of reactive oxygen species (ROS), that may prove to be more effective targets against MM. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
13. Restoration of the prolyl-hydroxylase domain protein-3 oxygen-sensing mechanism is responsible for regulation of HIF2α expression and induction of sensitivity of myeloma cells to hypoxia-mediated apoptosis.
- Author
-
Gastelum, Gilberto, Poteshkina, Aleksandra, Veena, Mysore, Artiga, Edgar, Weckstein, Geraldine, and Frost, Patrick
- Subjects
MULTIPLE myeloma diagnosis ,CANCER chemotherapy ,HYPOXIA-inducible factors ,APOPTOSIS inhibition ,DEOXYCYTIDINE - Abstract
Multiple myeloma (MM) is an incurable disease of malignant plasma B-cells that infiltrate the bone marrow (BM), resulting in bone destruction, anemia, renal impairment and infections. Physiologically, the BM microenvironment is hypoxic and this promotes MM progression and contributes to resistance to chemotherapy. Since aberrant hypoxic responses may result in the selection of more aggressive tumor phenotypes, we hypothesized that targeting the hypoxia-inducible factor (HIF) pathways will be an effective anti-MM therapeutic strategy. We demonstrated that MM cells are resistant to hypoxia-mediated apoptosis in vivo and in vitro, and that constitutive expression of HIF2α contributed to this resistance. Since epigenetic silencing of the prolyl-hydroxylase-domain-3 (PHD3) enzyme responsible for the O
2 -dependent regulation of HIF2α is frequently observed in MM tumors, we asked if PHD3 plays a role in regulating sensitivity to hypoxia. We found that restoring PHD3 expression using a lentivirus vector or overcoming PHD3 epigenetic silencing using a demethyltransferase inhibitor, 5-Aza-2’-deoxycytidine (5-Aza-dC), rescued O2 -dependent regulation of HIF2α and restored sensitivity of MM cells to hypoxia-mediated apoptosis. This provides a rationale for targeting the PHD3-mediated regulation of the adaptive cellular hypoxic response in MM and suggests that targeting the O2 -sensing pathway, alone or in combination with other anti-myeloma chemotherapeutics, may have clinical efficacy. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
14. IL-6-induced Enhancement of c-Myc Translation in Multiple Myeloma Cells: CRITICAL ROLE OF CYTOPLASMIC LOCALIZATION OF THE RNA-BINDING PROTEIN hnRNP A1*
- Author
-
Shi, Yijiang, Frost, Patrick, Hoang, Bao, Benavides, Angelica, Gera, Joseph, and Lichtenstein, Alan
- Subjects
Cell Nucleus ,Cytoplasm ,Interleukin-6 ,Heterogeneous Nuclear Ribonucleoprotein A1 ,Active Transport, Cell Nucleus ,Molecular Bases of Disease ,Peptide Fragments ,Proto-Oncogene Proteins c-myc ,HEK293 Cells ,Cell Line, Tumor ,Protein Biosynthesis ,Heterogeneous-Nuclear Ribonucleoprotein Group A-B ,Mutation ,Humans ,Phosphorylation ,Multiple Myeloma - 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
- 2010
15. MNK1-Induced eIF-4E Phosphorylation in Myeloma Cells: A Pathway Mediating IL-6-Induced Expansion and Expression of Genes Involved in Metabolic and Proteotoxic Responses.
- Author
-
Shi, Yijiang, Frost, Patrick, Hoang, Bao, Yang, Yonghui, Bardeleben, Carolyne, Gera, Joseph, and Lichtenstein, Alan
- Subjects
- *
CANCER risk factors , *CANCER cells , *PHOSPHORYLATION , *GENE expression , *INTERLEUKIN-6 , *ENDOPLASMIC reticulum - Abstract
Because multiple myeloma (MM) cells are at risk for endoplasmic reticulum (ER) stress, they require a carefully regulated mechanism to promote protein translation of selected transcripts when proliferation is stimulated. MAPK-interacting kinases (MNKs) may provide this mechanism by enhancing cap-dependent translation of a small number of critical transcripts. We, thus, tested whether MNKs played a role in MM responses to the myeloma growth factor interleukin-6 (IL-6). IL-6 activated MNK1 phosphorylation and induced phosphorylation of its substrate, eIF-4E, in MM lines and primary specimens. MNK paralysis, achieved pharmacologically or by shRNA, prevented MM expansion stimulated by IL-6. A phosphodefective eIF-4E mutant also prevented the IL-6 response, supporting the notion that MNK's role was via phosphorylation of eIF-4E. Both pharmacological MNK inhibition and expression of the phosphodefective eIF-4E mutant inhibited MM growth in mice. Although critical for IL-6-induced expansion, eIF-4E phosphorylation had no significant effect on global translation or Ig expression. Deep sequencing of ribosome-protected mRNAs revealed a repertoire of genes involved in metabolic processes and ER stress modulation whose translation was regulated by eIF-4E phosphorylation. These data indicate MM cells exploit the MNK/eIF-4E pathway for selective mRNA translation without enhancing global translation and risking ER stress. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
16. Mammalian Target of Rapamycin Inhibitors Induce Tumor Cell Apoptosis In Vivo Primarily by Inhibiting VEGF Expression and Angiogenesis.
- Author
-
Frost, Patrick, Eerlanger, Eileen, Mysore, Veena, Bao Hoang, YiJiang Shi, Gera, Joseph, and Lichtenstein, Alan
- Subjects
- *
RAPAMYCIN , *MTOR protein , *APOPTOSIS , *VASCULAR endothelial growth factors , *NEOVASCULARIZATION , *GENE expression , *B cell lymphoma , *IN vitro studies - Abstract
We found that rapalog mTOR inhibitors induce Gl arrest in the PTEN-null HS Sultan B-cell lymphoma line in vitro, but that administration of rapalogs in a HS Sultan xenograft model resulted in significant apoptosis, and that this correlated with induction of hypoxia and inhibition of neoangiogenesis and VEGF expression. Mechanistically, rapalogs prevent cap-dependent translation, but studies have shown that cap-independent, internal ribosome entry site (IRES)-mediated translation of genes, such as c-myc and cyclin D, can provide a fail-safe mechanism that regulates tumor survival. Therefore, we tested if IRES-dependent expression of VEGF could likewise regulate sensitivity of tumor cells in vivo. To achieve this, we developed isogenic HS Sultan cell lines that ectopically express the VEGF ORF fused to the p27 1RES, an 1RES sequence that is insensitive to AKT-mediated inhibition of 1RES activity and effective in PTEN-null tumors. Mice challenged with p27-VEGF transfected tumor cells were more resistant to the antiangiogenic and apoptotic effects of the rapalog, temsirolimus, and active site mTOR inhibitor, pp242. Our results confirm the critical role of VEGF expression in tumors during treatment with mTOR inhibitors and underscore the importance of 1RES activity as a resistance mechanism to such targeted therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
17. PAK1 Kinase Promotes Cell Motility and Invasiveness through CRK-II Serine Phosphorylation in Non-Small Cell Lung Cancer Cells.
- Author
-
Rettig, Matthew, Trinidad, Kenny, Pezeshkpour, G., Frost, Patrick, Sharma, Sherven, Moatamed, Farhad, Tamanoi, Fuyuhiko, and Mortazavi, Fariborz
- Subjects
CELL motility ,SMALL cell lung cancer ,CANCER cells ,CANCER invasiveness ,AMINO acids ,WOUND healing ,PHOSPHORYLATION ,GENE expression - Abstract
The role of c-Crk (CRK) in promoting metastasis is well described however the role of CRK phosphorylation and the corresponding signaling events are not well explained. We have observed CRK-II serine 41 phosphorylation is inversely correlated with p120-catenin and E-cadherin expressions in non-small cell lung cancer (NSCLC) cells. Therefore, we investigated the role of CRK-II serine 41 phosphorylation in the down-regulation of p120-catenin, cell motility and cell invasiveness in NSCLC cells. For this purpose, we expressed phosphomimetic and phosphodeficient CRK-II serine 41 mutants in NSCLC cells. NSCLC cells expressing phosphomimetic CRK-II seine 41 mutant showed lower p120-catenin level while CRK-II seine 41 phosphodeficient mutant expression resulted in higher p120-catenin. In addition, A549 cells expressing CRK-II serine 41 phosphomimetic mutant demonstrated more aggressive behavior in wound healing and invasion assays and, on the contrary, expression of phosphodeficient CRK-II serine 41 mutant in A549 cells resulted in reduced cell motility and invasiveness. We also provide evidence that PAK1 mediates CRK-II serine 41 phosphorylation. RNAi mediated silencing of PAK1 increased p120-catenin level in A549 and H157 cells. Furthermore, PAK1 silencing decreased cell motility and invasiveness in A549 cells. These effects were abrogated in A549 cells expressing phosphomimetic CRK-II serine 41. In summary, these data provide evidence for the role of PAK1 in the promotion of cell motility, cell invasiveness and the down regulation of p120-catenin through CRK serine 41 phosphorylation in NSCLC cells. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
18. IL -6-induced Enhancement of c-Myc Translation in Multiple Myeloma Cells: CRITICAL ROLE OF CYTOPLASMIC LOCALIZATION OF THE RNA-BINDING PROTEIN hnRNPA1.
- Author
-
Yijiang Shi, Frost, Patrick, Bao Hoang, Benavides, Angelica, Gera, Joseph, and Lichtenstein, Alan
- Subjects
- *
GENE expression , *GENETIC translation , *GENETIC transcription , *MULTIPLE myeloma , *PHOSPHORYLATION , *PROTEIN binding , *CELL lines - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
19. Little Wekiva River Restoration: A Watershed Management Strategy for an Urbanized River.
- Author
-
Frost, Patrick M.
- Published
- 1998
- Full Text
- View/download PDF
20. The Internal Ribosome Entry Site-Medaited Expression of VEGF Rescues HS Sultan Tumor Cells From Mammalian Target of Rapamycin (mTOR)-Inhibitors Induced Apoptosis in Vivo
- Author
-
Frost, Patrick, Gera, Joseph, and Lichtenstein, Alan K.
- Published
- 2012
- Full Text
- View/download PDF
21. Sensitization of immunoresistant prostate carcinoma cell lines to Fas/Fas ligand-mediated killing by cytotoxic lymphocytes: Independence of de novo protein synthesis.
- Author
-
Frost, Patrick J., Belldegrun, Arie, and Bonavida, Benjamin
- Published
- 1999
- Full Text
- View/download PDF
22. Steroid Receptor-Mediated Modulation of CD4+CD62L+ Cell Homing. Implications for Drug Abusersa.
- Author
-
CHIAPPELLI, FRANCESCO, MANFRINI, ERCOLANO, GWIRTSMAN, HARRY, GARCIA, CELSA, PHAM, LANCHI, LEE, PATTY, and FROST, PATRICK
- Published
- 1994
- Full Text
- View/download PDF
23. Immunotoxicity of Cocaethylene.
- Author
-
Chiappelli, Francesco, Kung, Michelle A., Villanueva, Pablo, Lee, Patricia, Frost, Patrick, and Prieto, Nerissa
- Published
- 1995
- Full Text
- View/download PDF
24. The Role of AKT and ERK in Regulating D-Cyclin Translation Inhibition and G1 Arrest in Multiple Myeloma Cells Treated with mTOR Inhibitors: Rationale for Combination Thereapy.
- Author
-
Frost, Patrick J., Shi, YiJiang, Bardalaban, Carolyne, Hoang, Bao, and Lichtenstein, Alan
- Published
- 2007
- Full Text
- View/download PDF
25. AKT Kinase Addiction in Multiple Myeloma.
- Author
-
Bardeleben, Carolyne, Yan, Huajun, Frost, Patrick, Hoang, Bao, Shi, Yijiang, Sharma, Sanjai, and Lichtenstein, Alan
- Published
- 2006
- Full Text
- View/download PDF
26. Mechanism by Which mTOR Inhibitors Sesitize Multiple Myeloma Cells to Dexamethasone-Induced Apoptosis.
- Author
-
Yan, Huajun, Frost, Patrick, Shi, Yijiang, Hoang, Bao, Sharma, Sanjai, Fisher, Myrna, Gera, Joseph, and Lichtenstein, Alan
- Published
- 2005
- Full Text
- View/download PDF
27. AKT Activity Regulates Sensitivity of Multiple Myeloma Cells to mTor Inhibitors.
- Author
-
Frost, Patrick, Hoang, Bao, Gera, Joseph, Sharma, Anushree, Shi, Yijiang, Yan, Huajun, and Lichtenstein, Alan
- Published
- 2004
- Full Text
- View/download PDF
28. Intestinal Helminth Parasites of the Bay Goby, Lepidogobius lepidus (Perciformes: Gobiidae), from Bodega Bay, California, U.S.A
- Author
-
Garcia, Danika, Resnikoff, Liat, Buth, Donald G., and Frost, Patrick
- Published
- 2004
- Full Text
- View/download PDF
29. Organic micropollutants in Swiss sewage sludge
- Author
-
Frost, Patrick, Camenzind, Rudolf, Mägert, André, Bonjour, René, and Karlaganis, Georg
- Published
- 1993
- Full Text
- View/download PDF
30. 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
-
Bao Hoang, Benavides, Angelica, Yijiang Shi, Yonghui Yang, Frost, Patrick, Gera, Joseph, and Lichtenstein, Alan
- Subjects
- *
MULTIPLE myeloma , *B cell lymphoma , *RAPAMYCIN , *IMMUNOSUPPRESSIVE agents , *DISEASE complications - 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 sitem TOR inhibitors overcome AKT activation often seen with rapalog therapy, feedback ERK activation is still aproblem 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. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
31. Gigaxonin Suppresses Epithelial-to-Mesenchymal Transition of Human Cancer Through Downregulation of Snail.
- Author
-
Veena MS, Gahng JJ, Alani M, Ko AY, Basak SK, Liu IY, Hwang KJ, Chatoff JR, Venkatesan N, Morselli M, Yan W, Ali I, Kaczor-Urbanowicz KE, Gowda BS, Frost P, Pellegrini M, Moatamed NA, Wilczynski SP, Bomont P, Wang MB, Shin DS, and Srivatsan ES
- Subjects
- Humans, Animals, Mice, Down-Regulation genetics, Cell Line, Tumor, Epithelial-Mesenchymal Transition genetics, Cadherins genetics, Head and Neck Neoplasms drug therapy
- Abstract
Gigaxonin is an E3 ubiquitin ligase that plays a role in cytoskeletal stability. Its role in cancer is not yet clearly understood. Our previous studies of head and neck cancer had identified gigaxonin interacting with p16 for NFκB ubiquitination. To explore its role in cancer cell growth suppression, we analyzed normal and tumor DNA from cervical and head and neck cancers. There was a higher frequency of exon 8 SNP (c.1293 C>T, rs2608555) in the tumor (46% vs. 25% normal, P = 0.011) pointing to a relationship to cancer. Comparison of primary tumor with recurrence and metastasis did not reveal a statistical significance. Two cervical cancer cell lines, ME180 and HT3 harboring exon 8 SNP and showing T allele expression correlated with higher gigaxonin expression, reduced in vitro cell growth and enhanced cisplatin sensitivity in comparison with C allele expressing cancer cell lines. Loss of gigaxonin expression in ME180 cells through CRISPR-Cas9 or siRNA led to aggressive cancer cell growth including increased migration and Matrigel invasion. The in vitro cell growth phenotypes were reversed with re-expression of gigaxonin. Suppression of cell growth correlated with reduced Snail and increased e-cadherin expression. Mouse tail vein injection studies showed increased lung metastasis of cells with low gigaxonin expression and reduced metastasis with reexpression of gigaxonin. We have found an association between C allele expression and RNA instability and absence of multimeric protein formation. From our results, we conclude that gigaxonin expression is associated with suppression of epithelial-mesenchymal transition through inhibition of Snail., Significance: Our results suggest that GAN gene exon 8 SNP T allele expression correlates with higher gigaxonin expression and suppression of aggressive cancer cell growth. There is downregulation of Snail and upregulation of e-cadherin through NFκB ubiquitination. We hypothesize that exon 8 T allele and gigaxonin expression could serve as diagnostic markers of suppression of aggressive growth of head and neck cancer., (© 2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
32. A Novel Therapeutic Induces DEPTOR Degradation in Multiple Myeloma Cells with Resulting Tumor Cytotoxicity.
- Author
-
Vega MI, Shi Y, Frost P, Huerta-Yepez S, Antonio-Andres G, Hernandez-Pando R, Lee J, Jung ME, Gera JF, and Lichtenstein A
- Subjects
- Animals, Antineoplastic Agents pharmacology, Bortezomib pharmacology, Bortezomib therapeutic use, Cell Line, Tumor, Humans, Mice, Inbred NOD, Mice, SCID, Multiple Myeloma metabolism, Treatment Outcome, Antineoplastic Agents therapeutic use, Apoptosis, Intracellular Signaling Peptides and Proteins metabolism, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Proteolysis drug effects
- 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., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
33. Multimodal Bioluminescent and Positronic-emission Tomography/Computational Tomography Imaging of Multiple Myeloma Bone Marrow Xenografts in NOG Mice.
- Author
-
Gastelum G, Chang EY, Shackleford D, Bernthal N, Kraut J, Francis K, Smutko V, and Frost P
- Subjects
- Animals, Disease Models, Animal, Disease Progression, Humans, Mice, Multiple Myeloma pathology, Transfection, Xenograft Model Antitumor Assays, Bone Marrow pathology, Carrier Proteins genetics, Multiple Myeloma diagnostic imaging, Positron Emission Tomography Computed Tomography methods
- Abstract
Multiple myeloma (MM) tumors engraft in the bone marrow (BM) and their survival and progression are dependent upon complex molecular and cellular interactions that exist within this microenvironment. Yet the BM microenvironment cannot be easily replicated in vitro, which potentially limits the physiologic relevance of many in vitro and ex vivo experimental models. These issues can be overcome by utilizing a xenograft model in which luciferase (LUC)-transfected 8226 MM cells will specifically engraft in the mouse skeleton. When these mice are given the appropriate substrate, D-luciferin, the effects of therapy on tumor growth and survival can be analyzed by measuring changes in the bioluminescent images (BLI) produced by the tumors in vivo. This BLI data combined with positronic-emission tomography/computational tomography (PET/CT) analysis using the metabolic marker 2-deoxy-2-(
18 F)fluoro-D-glucose (18 F-FDG) is used to monitor changes in tumor metabolism over time. These imaging platforms allow for multiple noninvasive measurements within the tumor/BM microenvironment.- Published
- 2019
- Full Text
- View/download PDF
34. Cytotoxic Properties of a DEPTOR-mTOR Inhibitor in Multiple Myeloma Cells.
- Author
-
Shi Y, Daniels-Wells TR, Frost P, Lee J, Finn RS, Bardeleben C, Penichet ML, Jung ME, Gera J, and Lichtenstein A
- Subjects
- Animals, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular pathology, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p21 physiology, Humans, Liver Neoplasms drug therapy, Liver Neoplasms pathology, Mechanistic Target of Rapamycin Complex 1, Mice, Multiple Myeloma pathology, Multiprotein Complexes physiology, Proto-Oncogene Proteins c-bcl-2 physiology, TOR Serine-Threonine Kinases physiology, Intracellular Signaling Peptides and Proteins antagonists & inhibitors, Multiple Myeloma drug therapy, TOR Serine-Threonine Kinases antagonists & inhibitors
- 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., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
35. SGK Kinase Activity in Multiple Myeloma Cells Protects against ER Stress Apoptosis via a SEK-Dependent Mechanism.
- Author
-
Hoang B, Shi Y, Frost PJ, Mysore V, Bardeleben C, and Lichtenstein A
- Subjects
- Animals, Apoptosis, Bortezomib pharmacology, Cell Line, Tumor, Endoplasmic Reticulum Stress, Female, Gene Expression Regulation, Neoplastic, Humans, Mice, Multiple Myeloma genetics, Multiple Myeloma metabolism, Thapsigargin administration & dosage, Thapsigargin pharmacology, Tunicamycin administration & dosage, Tunicamycin pharmacology, Up-Regulation, Xenograft Model Antitumor Assays, Bortezomib administration & dosage, Drug Resistance, Neoplasm, Immediate-Early Proteins genetics, Immediate-Early Proteins metabolism, Multiple Myeloma drug therapy, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism
- Abstract
Unlabelled: 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., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
36. A DNA-binding Molecule Targeting the Adaptive Hypoxic Response in Multiple Myeloma Has Potent Antitumor Activity.
- Author
-
Mysore VS, Szablowski J, Dervan PB, and Frost PJ
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis, Azoles pharmacology, Cell Hypoxia drug effects, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Humans, Male, Mice, Multiple Myeloma metabolism, Nylons pharmacology, Protein Binding drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents administration & dosage, Azoles administration & dosage, Basic Helix-Loop-Helix Transcription Factors metabolism, DNA metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Multiple Myeloma drug therapy
- Abstract
Unlabelled: Multiple myeloma is incurable and invariably becomes resistant to chemotherapy. Although the mechanisms remain unclear, hypoxic conditions in the bone marrow have been implicated in contributing to multiple myeloma progression, angiogenesis, and resistance to chemotherapy. These effects occur via adaptive cellular responses mediated by hypoxia-inducible transcription factors (HIF), and targeting HIFs can have anticancer effects in both solid and hematologic malignancies. Here, it was found that in most myeloma cell lines tested, HIF1α, but not HIF2α expression was oxygen dependent, and this could be explained by the differential expression of the regulatory prolyl hydroxylase isoforms. The anti-multiple myeloma effects of a sequence-specific DNA-binding pyrrole-imidazole (Py-Im) polyamide (HIF-PA), which disrupts the HIF heterodimer from binding to its cognate DNA sequences, were also investigated. HIF-PA is cell permeable, localizes to the nuclei, and binds specific regions of DNA with an affinity comparable with that of HIFs. Most of the multiple myeloma cells were resistant to hypoxia-mediated apoptosis, and HIF-PA treatment could overcome this resistance in vitro. Using xenograft models, it was determined that HIF-PA significantly decreased tumor volume and increased hypoxic and apoptotic regions within solid tumor nodules and the growth of myeloma cells engrafted in the bone marrow. This provides a rationale for targeting the adaptive cellular hypoxic response of the O2-dependent activation of HIFα using polyamides., Implications: Py-Im polyamides target and disrupt the adaptive hypoxic responses in multiple myeloma cells that may have clinical significance as a therapeutic strategy to treat myeloma engrafted in the bone marrow microenvironment., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
37. DEPTOR is linked to a TORC1-p21 survival proliferation pathway in multiple myeloma cells.
- Author
-
Yang Y, Bardeleben C, Frost P, Hoang B, Shi Y, Finn R, Gera J, and Lichtenstein A
- 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
- Full Text
- View/download PDF
38. Metabolomics identifies pyrimidine starvation as the mechanism of 5-aminoimidazole-4-carboxamide-1-β-riboside-induced apoptosis in multiple myeloma cells.
- Author
-
Bardeleben C, Sharma S, Reeve JR, Bassilian S, Frost P, Hoang B, Shi Y, and Lichtenstein A
- Subjects
- Apoptosis drug effects, Humans, Metabolomics methods, Multiple Myeloma genetics, Multiple Myeloma pathology, Pyrimidines biosynthesis, Aminoimidazole Carboxamide analogs & derivatives, Aminoimidazole Carboxamide pharmacology, Antineoplastic Agents pharmacology, Multiple Myeloma metabolism, Pyrimidines metabolism, Ribonucleosides pharmacology
- Abstract
To investigate the mechanism by which 5-aminoimidazole-4-carboxamide-1-β-riboside (AICAr) induces apoptosis in multiple myeloma cells, we conducted an unbiased metabolomics screen. AICAr had selective effects on nucleotide metabolism, resulting in an increase in purine metabolites and a decrease in pyrimidine metabolites. The most striking abnormality was a 26-fold increase in orotate associated with a decrease in uridine monophosphate (UMP) levels, indicating an inhibition of UMP synthetase (UMPS), the last enzyme in the de novo pyrimidine biosynthetic pathway, which produces UMP from orotate and 5-phosphoribosyl-α-pyrophosphate (PRPP). As all pyrimidine nucleotides can be synthesized from UMP, this suggested that the decrease in UMP would lead to pyrimidine starvation as a possible cause of AICAr-induced apoptosis. Exogenous pyrimidines uridine, cytidine, and thymidine, but not purines adenosine or guanosine, rescued multiple myeloma cells from AICAr-induced apoptosis, supporting this notion. In contrast, exogenous uridine had no protective effect on apoptosis resulting from bortezomib, melphalan, or metformin. Rescue resulting from thymidine add-back indicated apoptosis was induced by limiting DNA synthesis rather than RNA synthesis. DNA replicative stress was identified by associated H2A.X phosphorylation in AICAr-treated cells, which was also prevented by uridine add-back. Although phosphorylation of AICAr by adenosine kinase was required to induce multiple myeloma cell death, apoptosis was not associated with AMP-activated kinase activation or mTORC1 inhibition. A possible explanation for inhibition of UMP synthase activity by AICAr was a depression in cellular levels of PRPP, a substrate of UMP synthase. These data identify pyrimidine biosynthesis as a potential molecular target for future therapeutics in multiple myeloma cells.
- Published
- 2013
- Full Text
- View/download PDF
39. 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
-
Hoang B, Benavides A, Shi Y, Yang Y, Frost P, Gera J, and Lichtenstein A
- Subjects
- Apoptosis, Catalytic Domain, Cell Line, Tumor, Cell Survival, Enzyme Activation, Glutathione Transferase metabolism, Humans, Mechanistic Target of Rapamycin Complex 1, Multiple Myeloma drug therapy, Multiprotein Complexes, Phosphatidylinositol 3-Kinases metabolism, Proteins metabolism, Signal Transduction, Sirolimus pharmacology, Eukaryotic Initiation Factor-4E metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Indoles pharmacology, Multiple Myeloma metabolism, Purines pharmacology, TOR Serine-Threonine Kinases metabolism
- 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
- Full Text
- View/download PDF
40. Molecular cloning and expression of two β-defensin and two mucin genes in common carp (Cyprinus carpio L.) and their up-regulation after β-glucan feeding.
- Author
-
Marel Mv, Adamek M, Gonzalez SF, Frost P, Rombout JH, Wiegertjes GF, Savelkoul HF, and Steinhagen D
- Subjects
- Amino Acid Sequence, Animals, Carps immunology, Cloning, Molecular, Gene Expression Profiling, Immunologic Factors pharmacology, Molecular Sequence Data, Sequence Alignment, Carps genetics, Carps metabolism, Mucin-2 genetics, Mucin-2 metabolism, Mucin-5B genetics, Mucin-5B metabolism, Up-Regulation drug effects, beta-Defensins genetics, beta-Defensins metabolism, beta-Glucans pharmacology
- Abstract
In this study, we described the partial structure, mRNA tissue distribution and regulation of two carp mucin and two β-defensin genes. This is the first description of these genes in fish. The genes might provide relevant tools to monitor feed-related improvements of fish health under aquaculture conditions. Carp mucin 2 and mucin 5B genes show a high similarity to their mammalian and avian counterparts. The carp β-defensin 1 and β-defensin 2 genes cluster together well with their piscine family members. The influence of a β-glucan immunomodulant on the expression of these genes in mucosal tissues could be confirmed for the first time. Muc5B expression was significantly increased in the skin. For Muc2 no significant up- or down-regulation could be observed. Significantly higher expression levels of β-defensin 2 in gills and both β-defensin genes in skin were found. Thus, the mucosal system can be influenced by the addition of β-glucans to the food., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
41. IL-6-induced enhancement of c-Myc translation in multiple myeloma cells: critical role of cytoplasmic localization of the rna-binding protein hnRNP A1.
- Author
-
Shi Y, Frost P, Hoang B, Benavides A, Gera J, and Lichtenstein A
- Subjects
- Active Transport, Cell Nucleus drug effects, Cell Line, Tumor, Cell Nucleus drug effects, Cell Nucleus metabolism, HEK293 Cells, Heterogeneous Nuclear Ribonucleoprotein A1, Heterogeneous-Nuclear Ribonucleoprotein Group A-B chemistry, Heterogeneous-Nuclear Ribonucleoprotein Group A-B genetics, Humans, Mutation, Peptide Fragments metabolism, Phosphorylation drug effects, Cytoplasm drug effects, Cytoplasm metabolism, Heterogeneous-Nuclear Ribonucleoprotein Group A-B metabolism, Interleukin-6 pharmacology, Multiple Myeloma pathology, Protein Biosynthesis drug effects, Proto-Oncogene Proteins c-myc biosynthesis
- 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
- Full Text
- View/download PDF
42. Effect of autophagy on multiple myeloma cell viability.
- Author
-
Hoang B, Benavides A, Shi Y, Frost P, and Lichtenstein A
- Subjects
- Adenine analogs & derivatives, Adenine pharmacology, Antifungal Agents pharmacology, Antimalarials pharmacology, Antineoplastic Agents pharmacology, Apoptosis Regulatory Proteins antagonists & inhibitors, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Beclin-1, Boronic Acids pharmacology, Bortezomib, Cell Proliferation drug effects, Chloroquine pharmacology, Drug Therapy, Combination, Enzyme Inhibitors pharmacology, Humans, Immunoblotting, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Membrane Proteins metabolism, Microscopy, Fluorescence, Multiple Myeloma drug therapy, Pyrazines pharmacology, RNA, Small Interfering pharmacology, Sirolimus pharmacology, Thapsigargin pharmacology, Tumor Cells, Cultured, Apoptosis drug effects, Autophagy drug effects, Multiple Myeloma pathology
- 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.
- Published
- 2009
- Full Text
- View/download PDF
43. Regulation of D-cyclin translation inhibition in myeloma cells treated with mammalian target of rapamycin inhibitors: rationale for combined treatment with extracellular signal-regulated kinase inhibitors and rapamycin.
- Author
-
Frost P, Shi Y, Hoang B, Gera J, and Lichtenstein A
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Extracellular Signal-Regulated MAP Kinases metabolism, Humans, Male, Mice, Multiple Myeloma genetics, Multiple Myeloma pathology, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt metabolism, Retinoblastoma metabolism, Ribosomes drug effects, Ribosomes metabolism, TOR Serine-Threonine Kinases, Cyclin D1 metabolism, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Multiple Myeloma metabolism, Protein Biosynthesis drug effects, Protein Kinase Inhibitors pharmacology, Protein Kinases metabolism, Sirolimus pharmacology
- Abstract
We have shown that heightened AKT activity sensitized multiple myeloma cells to the antitumor effects of the mammalian target of rapamycin inhibitor CCI-779. To test the mechanism of the AKT regulatory role, we stably transfected U266 multiple myeloma cell lines with an activated AKT allele or empty vector. The AKT-transfected cells were more sensitive to cytostasis induced in vitro by rapamycin or in vivo by its analogue, CCI-779, whereas cells with quiescent AKT were resistant. The ability of mammalian target of rapamycin inhibitors to down-regulate D-cyclin expression was significantly greater in AKT-transfected multiple myeloma cells due, in part, to the ability of AKT to curtail cap-independent translation and internal ribosome entry site (IRES) activity of D-cyclin transcripts. Similar AKT-dependent regulation of rapamycin responsiveness was shown in a second myeloma model: the PTEN-null OPM-2 cell line transfected with wild-type PTEN. Because extracellular signal-regulated kinase (ERK)/p38 activity facilitates IRES-mediated translation of some transcripts, we investigated ERK/p38 as regulators of AKT-dependent effects on rapamycin sensitivity. AKT-transfected U266 cells showed significantly decreased ERK and p38 activity. However, only an ERK inhibitor prevented D-cyclin IRES activity in resistant "low-AKT" myeloma cells. Furthermore, the ERK inhibitor successfully sensitized myeloma cells to rapamycin in terms of down-regulated D-cyclin protein expression and G1 arrest. However, ectopic overexpression of an activated MEK gene did not increase cap-independent translation of D-cyclin in "high-AKT" myeloma cells, indicating that mitogen-activated protein kinase/ERK kinase/ERK activity was required, but not sufficient, for activation of the IRES. These data support a scenario where heightened AKT activity down-regulates D-cyclin IRES function in multiple myeloma cells and ERK facilitates activity.
- Published
- 2009
- Full Text
- View/download PDF
44. IL-6-induced stimulation of c-myc translation in multiple myeloma cells is mediated by myc internal ribosome entry site function and the RNA-binding protein, hnRNP A1.
- Author
-
Shi Y, Frost PJ, Hoang BQ, Benavides A, Sharma S, Gera JF, and Lichtenstein AK
- Subjects
- Heterogeneous Nuclear Ribonucleoprotein A1, Heterogeneous-Nuclear Ribonucleoprotein Group A-B genetics, Humans, Multiple Myeloma drug therapy, Multiple Myeloma metabolism, Protein Biosynthesis drug effects, Proto-Oncogene Proteins c-myc genetics, Ribosomes genetics, Transfection, Genes, myc, Heterogeneous-Nuclear Ribonucleoprotein Group A-B metabolism, Interleukin-6 pharmacology, Multiple Myeloma genetics, Proto-Oncogene Proteins c-myc biosynthesis, Ribosomes metabolism
- Abstract
Prior work indicates that c-myc translation is up-regulated in multiple myeloma cells. To test a role for interleukin (IL)-6 in myc translation, we studied the IL-6-responsive ANBL-6 and IL-6-autocrine U266 cell lines as well as primary patient samples. IL-6 increased c-myc translation, which was resistant to rapamycin, indicating a mechanism independent of mammalian target of rapamycin (mTOR) and cap-dependent translation. In contrast, the cytokine enhanced cap-independent translation via a stimulatory effect on the myc internal ribosome entry site (IRES). As known IRES-trans-activating factors (ITAF) were unaffected by IL-6, we used a yeast-three-hybrid screen to identify novel ITAFs and identified hnRNP A1 (A1) as a mediator of the IL-6 effect. A1 specifically interacted with the myc IRES in filter binding assays as well as EMSAs. Treatment of myeloma cells with IL-6 induced serine phosphorylation of A1 and increased its binding to the myc IRES in vivo in myeloma cells. Primary patient samples also showed binding between A1 and the IRES. RNA interference to knock down hnRNP A1 prevented an IL-6 increase in myc protein expression, myc IRES activity, and cell growth. These data point to hnRNP A1 as a critical regulator of c-myc translation and a potential therapeutic target in multiple myeloma.
- Published
- 2008
- Full Text
- View/download PDF
45. Oncogenic RAS mutations in myeloma cells selectively induce cox-2 expression, which participates in enhanced adhesion to fibronectin and chemoresistance.
- Author
-
Hoang B, Zhu L, Shi Y, Frost P, Yan H, Sharma S, Sharma S, Goodglick L, Dubinett S, and Lichtenstein A
- Subjects
- Cell Line, Tumor, Cyclooxygenase 2 analysis, Dinoprostone analysis, Fibronectins metabolism, Genes, ras genetics, Humans, Multiple Myeloma genetics, Mutation, Stromal Cells cytology, Cell Adhesion genetics, Cyclooxygenase 2 genetics, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Neoplastic, Multiple Myeloma pathology, Proto-Oncogene Proteins p21(ras) genetics
- 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
- Full Text
- View/download PDF
46. Mechanism by which mammalian target of rapamycin inhibitors sensitize multiple myeloma cells to dexamethasone-induced apoptosis.
- Author
-
Yan H, Frost P, Shi Y, Hoang B, Sharma S, Fisher M, Gera J, and Lichtenstein A
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Carrier Proteins metabolism, Cell Cycle Proteins, Cell Line, Tumor, Drug Synergism, Focal Adhesion Kinase 2 metabolism, Humans, MAP Kinase Kinase 4 metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Multiple Myeloma drug therapy, Multiple Myeloma enzymology, Multiple Myeloma pathology, Phosphoproteins metabolism, Receptors, Glucocorticoid metabolism, Ribosomal Protein S6 Kinases, 70-kDa metabolism, Sirolimus analogs & derivatives, Sirolimus pharmacology, TOR Serine-Threonine Kinases, bcl-Associated Death Protein metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Apoptosis drug effects, Dexamethasone pharmacology, Protein Kinase Inhibitors pharmacology, Protein Kinases metabolism
- 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.
- Published
- 2006
- Full Text
- View/download PDF
47. 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
-
Shi Y, Yan H, Frost P, Gera J, and Lichtenstein A
- Subjects
- Animals, Antibiotics, Antineoplastic pharmacology, Apoptosis drug effects, Boronic Acids pharmacology, Bortezomib, Cell Line, Tumor, Enzyme Activation drug effects, Humans, Insulin Receptor Substrate Proteins, Mice, Mice, SCID, Multiple Myeloma enzymology, Multiple Myeloma metabolism, Phosphatidylinositol 3-Kinases metabolism, Pyrazines pharmacology, Receptor, IGF Type 1 antagonists & inhibitors, Sirolimus analogs & derivatives, Sirolimus pharmacology, TOR Serine-Threonine Kinases, Xenograft Model Antitumor Assays, Insulin-Like Growth Factor I metabolism, Multiple Myeloma drug therapy, Phosphoproteins metabolism, Protein Kinase Inhibitors pharmacology, Protein Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Receptor, IGF Type 1 metabolism
- 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
- Full Text
- View/download PDF
48. Immunosensitization of resistant human tumor cells to cytotoxicity by tumor infiltrating lymphocytes.
- Author
-
Frost P, Caliliw R, Belldegrun A, and Bonavida B
- Subjects
- Adenocarcinoma pathology, Apoptosis drug effects, Apoptosis physiology, Carcinoma pathology, Carcinoma, Renal Cell pathology, Combined Modality Therapy, Cytotoxicity, Immunologic drug effects, Egtazic Acid pharmacology, Fas Ligand Protein, Female, Humans, Ionomycin pharmacology, Kidney Neoplasms pathology, Lymphocytes, Tumor-Infiltrating drug effects, Magnesium Chloride pharmacology, Male, Membrane Glycoproteins physiology, Ovarian Neoplasms pathology, Perforin, Pore Forming Cytotoxic Proteins, Prostatic Neoplasms pathology, Recombinant Proteins pharmacology, Tetradecanoylphorbol Acetate pharmacology, Tumor Cells, Cultured drug effects, Tumor Cells, Cultured immunology, fas Receptor physiology, Adenocarcinoma immunology, Antineoplastic Agents pharmacology, Carcinoma immunology, Carcinoma, Renal Cell immunology, Cisplatin pharmacology, Drug Resistance, Neoplasm, Interleukin-2 pharmacology, Kidney Neoplasms immunology, Killer Cells, Lymphokine-Activated immunology, Lymphocytes, Tumor-Infiltrating immunology, Ovarian Neoplasms immunology, Prostatic Neoplasms immunology
- Abstract
Most anti-cancer therapies induce apoptotic cell death, but a major barrier to long-term cancer treatments is the generation of apoptosis-resistant tumor cells. Tumor cells that become resistant to one therapy are usually cross-resistant to subsequent therapies, including those with different cellular/molecular targets, suggesting that resistant tumor cells acquire modifications of the general apoptotic pathway. Most solid tumors are characterized by infiltration of lymphocytes (tumor infiltrating lymphocytes, TIL), which may serve as a basis for new strategies to generate tumor specific lymphocytes. However, TIL frequently are unable to kill autologous tumor cells suggesting that they are anergic/tolerant. It is possible that the TIL are functional but the tumor cells are resistant to TIL-mediated apoptotic pathways. Previous findings revealed that resistant tumor cells can be sensitized with cytokines or subtoxic concentrations of chemotherapeutic drugs and restore killing by cytotoxic lymphocytes. In this study, we examined whether TIL can kill autologous and allogeneic tumor cells following sensitization with chemotherapeutic drugs. Renal and prostate cancer-derived TIL were cytotoxic to chemosensitized resistant tumor cells. Killing by TIL was found to be perforin-dependent and perforin-independent. These findings demonstrate that combination drug and immunotherapy may be able to overcome tumor cell resistance to killing by TIL. Further, in vivo sensitization of drug-resistant tumor cells by subtoxic doses of sensitizing chemotherapeutic drugs may result in tumor regression by the host immune system.
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.