Добірка наукової літератури з теми "P73, multiple myeloma"

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся зі списками актуальних статей, книг, дисертацій, тез та інших наукових джерел на тему "P73, multiple myeloma".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Статті в журналах з теми "P73, multiple myeloma"

1

Hao, Mu, Yu Qin, Meirong Zang, Yan Xu, Gang An, Changhong Li, Ye Yang, Zhimin Gu, Fenghuang Zhan, and Lugui Qiu. "Hypermethylation of TAp73 Suppresses ABL1-Involved DNA Damage Response in Multiple Myeloma." Blood 124, no. 21 (December 6, 2014): 3374. http://dx.doi.org/10.1182/blood.v124.21.3374.3374.

Повний текст джерела
Анотація:
Abstract Background: More recently, multiple myeloma (MM) cells evade apoptosis despite pervasive DNA damage was demonstrated. However, the relevance of ongoing DNA damage and the mechanisms by which apoptosis is suppressed remain to be fully elucidated. p53 deletion and mutations do not appear to be a pivotal event in the evolution from pre-malignancy toward malignancy in MM. The protooncogene ABL1 was an alternative pathway to p53 down stream of ATM/ATR, which is commonly translocated in Chronic Myelogenous Leukemia (CML). ABL1 forms a complex with the tumor suppressor gene TP73, which belongs to the p53 family. P73 is expressed as multiple isoforms due to the usage of two different promoters, P1 promoter of TAp73 inducing apoptosis and P2 promoter of Δ Np73 promoting survival. In this study, we explored the role of ABL1/p73 axis in MM cells evading ongoing DNA damage induced apoptosis. Materials and methods: Real-time-PCR was used to detect the ABL1 and miR-203a expression in MM primary samples and MM cell lines. Immunofluency staining was performed to detect the ɣ-H2A.X level in MM cells. Flow cytometry was performed to detect the apoptosis in MM cells. Bisulfite Pyrosequencing and Methylation Specific-PCR were used to detect the p73 promoter methylation. Results: Our results revealed that ABL1 level was up-regulated both in primary MM samples and MM cell lines (-1.25±0.28 vs. 0.06±0.24, p=0.02). MiR-203 which suppresses ABL1 expression was down-regulated (0.01±0.01 vs. 0.97±0.08, p=0.01). MM cell lines and primary cells showed high ɣ-H2A.X staining. Immunofluency staining showed that ABL1 relocalized in the nucleus of MM cells after treated with doxorubicin. The apoptosis of MM cells was significantly up-regulated (7.8±2.1)% vs. (25.4±4.5)%, p<0.05. Doxorubicin treatment combined with ABL1 inhibitor (STI571) suppressed the apoptosis significantly, (25.4±4.5)% vs. (12.2±3.4)%, p=0.03. Bisulfite Pyrosequencing and MS-PCR of 42 newly diagnosed MM patient sample revealed that the P1 promoter of p73 was hypermethylated compared with normal plasma cells (86% ±7% vs. 58%±4%, p=0.032). RT-qPCR and western blotting showed that Δ Np73 levels were significantly higher than TAp73 (148.6±19.3 vs. 6.8±2.4, p=0.021) in plasma cells of those patients and MM cell lines. Conclusion: Hypermethylation of p73 promoter suppresses TAp73 expression. The deregulated of ABL1-p73 pathway in MM cells resulted in marked reduction of apoptosis induced by ongoing DNA damage. Disclosures No relevant conflicts of interest to declare.
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Schultheis, B., A. Krämer, A. Willer, U. Hegenbart, H. Goldschmidt, and R. Hehlmann. "Analysis of p73 and p53 gene deletions in multiple myeloma." Leukemia 13, no. 12 (December 1999): 2099–103. http://dx.doi.org/10.1038/sj.leu.2401609.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Lunghi, Paolo, Nicola Giuliani, Laura Mazzera, Francesca Morandi, Luigi Salvatore, Marcellina Mangoni, Vittorio Rizzoli, and Antonio Bonati. "Targeting MEK/MAPK Signal Transduction Module Potentiates Arsenic Trioxide (ATO)-Induced Apoptosis in Multiple Myeloma Cells through Multiple Signaling Pathways." Blood 110, no. 11 (November 16, 2007): 1517. http://dx.doi.org/10.1182/blood.v110.11.1517.1517.

Повний текст джерела
Анотація:
Abstract Multiple Myeloma (MM) cells are extremely resistant to apoptosis and currently new potential drug combinations are under investigation. We have shown that the combined treatment with the MEK1/2 inhibitor PD184352 (PD) and Arsenic Trioxide (ATO) resulted in the synergistic (Combination Index &lt;1.0) induction of apoptosis in 7 human myeloma cell lines (HMCLs: XG1, XG6, OPM2, JJN3, RPMI, H929, Sultan) analyzed, irrespective of their p53 status. The combined treatment was also a highly potent inducer of apoptosis and mitochondrial damage in the majority of the primary multiple myeloma (MM) cell samples ex vivo analyzed at different disease stage (9 out of 12). Growth factors, IL-6 or insulin-like growth factor 1 (IGF-1), or a co-culture system with bone marrow stromal cells (BMSCs) failed to confer resistance to this combination regimen. The combination PD/ATO had a minimal effect on normal B cells in vitro. By investigating the molecular mechanisms involved in MM cells PD/ATO-induced apoptosis, we found that co-treatment with PD strikingly elevated the (DR4+DR5)/(DcR1+DcR2) tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors ratio, caspase-8 activation, Bid fragmentation, mitochondrial depolarization and caspase-9 activation of ATO-treated HMCLs that do not have a functional p53 pathway. In HMCLs carrying a functional p53 pathway, the treatment with PD greatly enhanced the ATO-induced p53 accumulation (two fold increase) and p73, a p53 paralogue, cooperated with p53 in the pro-apoptotic p53/p73 target genes up regulation, caspase-9, -3 activation and apoptosis induction; in these HMCLs the selective down-regulation of p53 or p73 demonstrated that both have a biological relevance in PD/ATO-induced caspase-3 activation, PARP fragmentation and apoptosis. In HMCLs carrying a functional p53 the extrinsic caspase-8 mediated pathway was partially activated by PD/ATO treatment. We also demonstrated that, in MM cells carrying or not a functional p53 pathway, the combined treatment PD/ATO increased the level of the pro-apoptotic Bim (PD-mediated) and decreased its neutralizing anti-apoptotic protein Mcl-1 (ATO-mediated). The selective down-regulation of Bim significantly diminished caspase-8/-9/-3 cleavage/activation, PARP fragmentation and apoptosis of PD/ATO-treated MM cells, thereby indicating that Bim can play an important role not only in the intrinsic mitochondrial programmed cell death but also in the extrinsic caspase-8 mediated pathway. Accordingly, a physical interaction between Bim and DR4/DR5 TRAIL receptors in PD/ATO-treated MM cells carrying a non functional p53 was found by coimmunoprecipitation and Western blot studies. Our experiments have enlightened some relevant mechanisms that explain the apoptotic response of myeloma cells to ATO plus MEK inhibitor combination.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Raab, Marc S., Klaus Podar, Jing Zhang, Giovanni Tonon, Johannes H. Fruehauf, Iris Breitkreutz, Boris K. Lin, Teru Hideshima, Dharminder Chauhan, and Kenneth C. Anderson. "Targeting Proteinkinase C Alters ER-Stress and b-Catenin Signaling in Multiple Myeloma: Therapeutic Implications." Blood 110, no. 11 (November 16, 2007): 258. http://dx.doi.org/10.1182/blood.v110.11.258.258.

Повний текст джерела
Анотація:
Abstract We have previously shown that the novel orally available small molecule inhibitor of PKC enzastaurin (Eli Lilly and Company) inhibits MM cell growth, survival and angiogenesis both in vitro and in vivo. To date, however, the downstream effects contributing to growth inhibition and cell death remain to be determined. Here, we performed global gene expression profiling on enzastaurin treated MM cells and identified 200 Genes to be differentially regulated with a &gt; 2-fold cut off. Strikingly, two major groups of up-regulated probe sets were associated with either of two pathways - endoplasmatic reticulum (ER)-stress response or WNT-signaling. Importantly, MM cells, producing high levels of paraprotein, are highly susceptible to perturbation of ER function and protein folding. Moreover, PKC isoforms have been reported to directly regulate the canonical WNT pathway via phosphorylation of b-catenin (CAT), leading to its ubiquination and proteasomal degradation. Specifically, we fist evaluated the role of enzastaurin in mediating ER-stress in MM cells. The transcriptional up-regulation of genes involved in ER-stress (GADD153/CHOP, GADD34, ATF3), triggered by enzastaurin at 3h, was confirmed by western blot analysis, accompanied by induction of the molecular ER chaperone BiP/grp78, phosphorylation of eIF2a consistent with PERK activation, and up-regulation of p21. These events were preceded by an early (1h) increase of intracellular calcium levels, a hallmark of ER-stress, assessed by FLUO4 staining. These data suggest an important role of ER-stress response in the early growth inhibition of MM cells caused by enzastaurin. Second, we delineated effects of enzastaurin on WNT pathway in MM and other tumor cell lines. Upon enzastaurin treatment, CAT was dephosphorylated at Ser33, 37, 41 in a dose- and time-dependent manner in all cell lines tested (10 MM, 3 colon cancer, HeLa, as well as human embryonic kidney 293 cells). Consequently, accumulation of CAT occurred in both cytosolic and nuclear fractions of treated MM cells, associated with activated TOPflash LUC-reporter system, confirming nuclear transactivating activity. Specific inhibition of CAT by siRNA partially rescued HeLa, HEK 293, and MM cells from cell death induced by enzastaurin. Analysis of downstream target molecules revealed a CAT-dependent up-regulation of c-Jun, but not of c-Myc or Cyclin D1. c-Jun has been reported to stabilize p73, a pro-apoptotic p53-family member; CAT induction by enzastaurin led to p73 (but not p53) activation and was also abrogated by CAT-specific siRNA. In turn, specific knockdown of p73 by siRNA rescued cells from enzastaurin-induced apoptosis. Finally, ectopic overexpression of CAT in HeLa and MM cells induced c-Jun expression and p73 activation, followed by apoptotic cell death. Our studies therefore indicate that ER-stress response contributes to the immediate inhibition of proliferation by enzastaurin, followed by CAT accumulation leading to p73 activation, contributing to enzastaurin-mediated cell death. These findings provide a novel link between CAT and p53-family members. Moreover p73, which is only rarely mutated in human cancers, represents a novel therapeutic target in MM.
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Lunghi, Paolo, Nicola Giuliani, Laura Mazzera, Guerino Lombardi, Micaela Ricca, Attilio Corradi, Anna Maria Cantoni, et al. "Targeting MEK/MAPK signal transduction module potentiates ATO-induced apoptosis in multiple myeloma cells through multiple signaling pathways." Blood 112, no. 6 (September 15, 2008): 2450–62. http://dx.doi.org/10.1182/blood-2007-10-114348.

Повний текст джерела
Анотація:
Abstract We demonstrate that blockade of the MEK/ERK signaling module, using the small-molecule inhibitors PD184352 or PD325901 (PD), strikingly enhances arsenic trioxide (ATO)–induced cytotoxicity in human myeloma cell lines (HMCLs) and in tumor cells from patients with multiple myeloma (MM) through a caspase-dependent mechanism. In HMCLs retaining a functional p53, PD treatment greatly enhances the ATO-induced p53 accumulation and p73, a p53 paralog, cooperates with p53 in caspase activation and apoptosis induction. In HMCLs carrying a nonfunctional p53, cotreatment with PD strikingly elevates the (DR4 + DR5)/(DcR1 + DcR2) tumor necrosis factor (TNF)–related apoptosis-inducing ligand (TRAIL) receptors ratio and caspase-8 activation of ATO-treated cells. In MM cells, irrespective of p53 status, the combined PD/ATO treatment increases the level of the proapoptotic protein Bim (PD-mediated) and decreases antiapoptotic protein Mcl-1 (ATO-mediated). Moreover, Bim physically interacts with both DR4 and DR5 TRAIL receptors in PD/ATO-treated cells, and loss of Bim interferes with the activation of both extrinsic and intrinsic apoptotic pathways in response to PD/ATO. Finally, PD/ATO treatment induces tumor regression, prolongs survival, and is well tolerated in vivo in a human plasmacytoma xenograft model. These preclinical studies provide the framework for testing PD325901 and ATO combination therapy in clinical trials aimed to improve patient outcome in MM.
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Shammas, Masood A., Paola Neri, Hemanta Koley, Ramesh B. Batchu, Robert C. Bertheau, Vidit Munshi, Rao Prabhala, et al. "Specific killing of multiple myeloma cells by (-)-epigallocatechin-3-gallate extracted from green tea: biologic activity and therapeutic implications." Blood 108, no. 8 (October 15, 2006): 2804–10. http://dx.doi.org/10.1182/blood-2006-05-022814.

Повний текст джерела
Анотація:
AbstractEpigallocatechin-3-gallate (EGCG), a polyphenol extracted from green tea, is an antioxidant with chemopreventive and chemotherapeutic actions. Based on its ability to modulate growth factor-mediated cell proliferation, we evaluated its efficacy in multiple myeloma (MM). EGCG induced both dose- and time-dependent growth arrest and subsequent apoptotic cell death in MM cell lines including IL-6-dependent cells and primary patient cells, without significant effect on the growth of peripheral blood mononuclear cells (PBMCs) and normal fibroblasts. Treatment with EGCG also led to significant apoptosis in human myeloma cells grown as tumors in SCID mice. EGCG interacts with the 67-kDa laminin receptor 1 (LR1), which is significantly elevated in myeloma cell lines and patient samples relative to normal PBMCs. RNAi-mediated inhibition of LR1 resulted in abrogation of EGCG-induced apoptosis in myeloma cells, indicating that LR1 plays an important role in mediating EGCG activity in MM while sparing PBMCs. Evaluation of changes in gene expression profile indicates that EGCG treatment activates distinct pathways of growth arrest and apoptosis in MM cells by inducing the expression of death-associated protein kinase 2, the initiators and mediators of death receptor-dependent apoptosis (Fas ligand, Fas, and caspase 4), p53-like proteins (p73, p63), positive regulators of apoptosis and NF-κB activation (CARD10, CARD14), and cyclin-dependent kinase inhibitors (p16 and p18). Expression of related genes at the protein level were also confirmed by Western blot analysis. These data demonstrate potent and specific antimyeloma activity of EGCG and provide the rationale for its clinical evaluation.
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Shammas, Masood A., Ramesh B. Batchu, Hemanta Koley, Robert C. Bertheau, Paola Neri, Raj Goyal, Kenneth C. Anderson, and Nikhil C. Munshi. "A Green Tea Polyphenol, Epigallocatechin-3-Gallate, Induces Selective Apoptosis in Multiple Myeloma Cells: Mechanism of Action and Therapeutic Potential." Blood 106, no. 11 (November 16, 2005): 1590. http://dx.doi.org/10.1182/blood.v106.11.1590.1590.

Повний текст джерела
Анотація:
Abstract Epigallocatechin-3-gallate (EGCG), a polyphenol extracted from green tea, induces dose and time dependent cell death in both IL-6-dependent and independent multiple myeloma cell lines and primary patient cells, with minimal or no effect on the growth of normal cells. The cell death is apoptotic as determined by annexin V staining and is not inhibited by IL-6. Evaluation of molecular mechanism of action by gene expression profiling indicated that EGCG had a profound effect on transcription of major regulatory genes involved in distinct pathways controlling cell growth arrest and apoptosis: Exposure of myeloma cells to EGCG induced the expression of: 1) Fas ligand, Fas, and caspase 4, the initiators and mediators of death receptor dependent apoptosis; 2) death-associated protein kinase 2, a multifunctional pro-apoptotic protein kinase; 3) P53-like proteins, p73, p63; 4) CARD10 and CARD14, positive regulators of apoptosis and NF-kappaB activation; and 5) Cyclin-dependent kinase inhibitors, p16 and p18. In a subset of these selected genes, the expression data is also confirmed with western blot analyses. We have also demonstrated that the transcript and protein levels of a metastasis associated laminin receptor 1 are significantly elevated in myeloma cell lines and patient samples compared to normal cells. RNAi mediated inhibition of laminin receptor 1, abrogated EGCG-induced apoptosis in myeloma cells thus indicating that the profound anti-cancer effect of this compound is probably mediated through this receptor. The selective expression of this receptor explains the selective activity of EGCG in multiple myeloma cells without adversely affecting normal cells. Taken together these data confirm significant and selective anti-cancer activity of EGCG, a natural product, in MM and provides the basis for its clinical evaluation.
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Cottini, Francesca, Teru Hideshima, Martin Sattler, Federico Caligaris-Cappio, Kenneth C. Anderson, and Giovanni Tonon. "The Role of the ABL1/YAP1/P73 Axis in Prevention of DNA Damage-Mediated Apoptosis in Multiple Myeloma." Blood 120, no. 21 (November 16, 2012): 725. http://dx.doi.org/10.1182/blood.v120.21.725.725.

Повний текст джерела
Анотація:
Abstract Abstract 725 Background: Genome integrity plays a crucial role in the development of normal plasma cells to eliminate aberrant ones. Multiple myeloma (MM) is a plasma cell malignancy characterized by complex heterogeneous cytogenetic abnormalities. MM cells show constitutive DNA Damage Response (DDR) and activate compensatory mechanisms to prevent DNA-damage mediated apoptosis. Here we define the molecular mechanisms of these protective effects. Methods: A panel of 15 MM cell lines was used. Blood and BM samples from healthy volunteers and MM patients were obtained after informed consent and subjected to Ficoll-Paque density sedimentation to get mononuclear cells (MNCs). Patient MM cells were isolated from BM MNCs by CD138-positive selection. Lentiviral delivery system was used for expression and knock-down of YAP1 in KMS-18, KMS-20, MM.1S and UTMC-2 MM cell lines. The biologic impact of YAP1 phenotype was evaluated using cell growth, viability and apoptosis assays. Results: We confirmed that a wide range of MM patient cells and MM cell lines have markers of constitutive DDR, including phosphorylation of H2A.X, ATM, ATR, Chk2 and Chk1, assessed by western blot analysis and immunofluorescence. However, these MM cells do not show basal level of apoptosis. Specifically, cleaved forms of caspase 3 and PARP are lacking in non-treated cells, and the absence of co-expression of cleaved caspase 3 with phospho-H2A.X by immunofluorescence confirms that phospho-H2A.X positive cells are viable cells. Since DDR is present in both p53-wild-type (wt) and p53-mutated cell lines, we examined whether ABL1/YAP1/p73 axis represents an alternative and crucial pathway to avoid DNA-damage mediated apoptosis. Indeed, ABL1 is up-regulated and predominantly localizes in the nucleus, a potential apoptotic stimulus, in MM cells, as assessed by western blot and immunofluorescence. To define if ABL1 nuclear localization was triggered by DDR, we treated MM cells with a specific ATM inhibitor (Ku55933) and a DNA damaging agent, doxorubicin. Inhibition of DDR in both p53 wt cell lines (MM.1S and H929) and p53 mutated cell lines (UTMC-2, JJN-3 and KMS-20) causes ABL1 cytoplasmic retention, while doxorubicin increases ABL1 nuclear translocation. Co-treatment with doxorubicin and ABL1 inhibitor STI-571 rescues MM cells from doxorubicin-mediated cell death. In particular, apoptotic cells decrease from 47.2% to 21.5% in U266, from 55.3% to 12.4% in MM.1S, and from 57.9% to 19.1% in UTMC-2 cells in response to combination treatment. To delineate the molecular mechanisms whereby MM cells repress ABL1 pro-apoptotic function, we focused on YAP1, a downstream target of the Hippo pathway involved in ABL1 cascade. We explored a large dataset of aCGH data on MM patients and discovered that YAP1 genomic locus (chr. 11q22) is deleted, associated with BIRC2 and BIRC3 in 11% of patients. This genetic abnormality was also found in KMS-18 and KMS-20 cell lines. Although YAP1 expression was normal in peripheral blood MNCs (PBMCs), its expression was decreased in the majority of patient MM cells and MM cell lines regardless of the presence of focal deletion. Importantly, low expression of YAP1 is associated with poor prognosis of MM patients. To further delineate the biologic significance of YAP1 in MM cells, we re-expressed pLENTI4-YAP1-EGFP in MM cell lines with either YAP1 deletion (KMS-18, KMS-20) or YAP1 low expression (MM.1S). We also silenced YAP1 using two different specific shRNAs in UTMC-2 MM cell line. As expected, YAP1 re-expression reduces cellular growth and increases apoptosis in all cell lines tested (25.7%, 37.1% and 32.3% apoptotic KMS-20, KMS-18 and MM.1S cells, respectively), condition that was further enhanced by doxorubicin treatment. Previous studies have shown that P73 is expressed in MM even though at low levels and we here show that they inversely correlate with YAP1 protein expression. Importantly, YAP1 re-expression increases p73 stability and promotes transcription of p73-target genes including BAX, PUMA and p21. In contrast, UTMC-2-YAP1−/− cells show improved survival with lower levels of basal apoptosis and higher resistance to treatment with bortezomib or doxorubicin. Conclusion: YAP1 mediates a strong apoptotic signal for MM cells. Thus, activation and/or overexpression of YAP1 represent a novel therapeutic strategy to improve outcome of patients with MM. Disclosures: Anderson: Celgene, Millennium, BMS, Onyx: Membership on an entity's Board of Directors or advisory committees; Acetylon, Oncopep: Scientific Founder and Scientific Founder, Scientific Founder and Scientific Founder Other.
Стилі APA, Harvard, Vancouver, ISO та ін.
9

Raab, Marc S., Iris Breitkreutz, Giovanni Tonon, Jing Zhang, Johannes Fruehauf, Boris K. Lin, Dharminder Chauhan, et al. "Targeting PKC: A Novel Role for Beta-catenin in ER Stress and Apoptotic Signaling." Blood 112, no. 11 (November 16, 2008): 2763. http://dx.doi.org/10.1182/blood.v112.11.2763.2763.

Повний текст джерела
Анотація:
Abstract Targeting protein kinase C (PKC) isoforms by the small molecule inhibitor enzastaurin has shown promising pre-clinical activity in a wide range of tumor cells. In this study, we further delineated its mechanism of action in multiple myeloma (MM) cells and found a novel role of b-catenin in regulating growth and survival of tumor cells. Inhibition of PKC leads to rapid accumulation of b-catenin by preventing the phosphorylation required for its proteasomal degradation. Specifically, b-catenin was dephosphorylated at Ser33,37,41 and accumulated in a dose- and time-dependent manner in all cell lines tested (including primary MM cells and 10 MM cell lines, 3 colon cancer, HeLa, as well as HEK 293 cells). Microarray analysis and siRNA-mediated gene silencing in MM cells revealed that accumulated b-catenin activates early ER stress signaling via eIF2a, CHOP and p21, leading to immediate inhibition of proliferation. Conversely, knock-down of components of the ER stress response pathway by siRNA (i.e., CHOP) abrogated the inhibitory effect of enzastaurin on MM cell proliferation. Importantly, accumulated b-catenin also contributes to enzastaurin-induced cell death, since inhibition of b-catenin by siRNA partially rescued HeLa, HEK 293, and MM cells from cell death induced by enzastaurin. Analysis of downstream target molecules revealed a b-catenin -dependent up-regulation of c-Jun, but not of c-Myc or Cyclin D1. c-Jun has been reported to stabilize p73, a pro-apoptotic p53-family member; b-catenin induction by enzastaurin led to p73 (but not p53) activation, which was also abrogated by b-catenin -specific siRNA. In turn, specific knockdown of p73 by siRNA rescued cells from enzastaurin-induced apoptosis. Finally, ectopic overexpression of b-catenin in HeLa and MM cells induced c-Jun expression and p73 activation, followed by apoptotic cell death. In summary, our data reveal a novel role of b-catenin in ER stress-mediated growth inhibition and a new pro-apoptotic mechanism triggered by b-catenin upon inhibition of PKC isoforms, and further demonstrate that p73 represents a novel therapeutic target in MM. Based on these and previous data, enzastaurin is currently under clinical investigation in a variety of hematologic malignancies including MM.
Стилі APA, Harvard, Vancouver, ISO та ін.
10

Raab, Marc S., Iris Breitkreutz, Giovanni Tonon, Jing Zhang, Patrick J. Hayden, Thu Nguyen, Johannes H. Fruehauf, et al. "Targeting PKC: a novel role for beta-catenin in ER stress and apoptotic signaling." Blood 113, no. 7 (February 12, 2009): 1513–21. http://dx.doi.org/10.1182/blood-2008-05-157040.

Повний текст джерела
Анотація:
Abstract Targeting protein kinase C (PKC) isoforms by the small molecule inhibitor enzastaurin has shown promising preclinical activity in a wide range of tumor cells. We further delineated its mechanism of action in multiple myeloma (MM) cells and found a novel role of β-catenin in regulating growth and survival of tumor cells. Specifically, inhibition of PKC leads to rapid accumulation of β-catenin by preventing the phosphorylation required for its proteasomal degradation. Microarray analysis and small-interfering RNA (siRNA)–mediated gene silencing in MM cells revealed that accumulated β-catenin activates early endoplasmic reticulum stress signaling via eIF2α, C/EBP-homologous protein (CHOP), and p21, leading to immediate growth inhibition. Furthermore, accumulated β-catenin contributes to enzastaurin-induced cell death. Sequential knockdown of β-catenin, c-Jun, and p73, as well as overexpression of β-catenin or p73 confirmed that accumulated β-catenin triggers c-Jun–dependent induction of p73, thereby conferring MM cell apoptosis. Our data reveal a novel role of β-catenin in endoplasmic reticulum (ER) stress-mediated growth inhibition and a new proapoptotic mechanism triggered by β-catenin on inhibition of PKC isoforms. Moreover, we identify p73 as a potential novel therapeutic target in MM. Based on these and previous data, enzastaurin is currently under clinical investigation in a variety of hematologic malignancies, including MM.
Стилі APA, Harvard, Vancouver, ISO та ін.

Дисертації з теми "P73, multiple myeloma"

1

LUCANI, BENEDETTA. "EXPRESSION OF P73 IN MYELOMA CELL LINES AND PLASMA CELLS OF MULTIPLE MYELOMA PATIENTS." Doctoral thesis, Università di Siena, 2016. http://hdl.handle.net/11365/1004869.

Повний текст джерела
Анотація:
Background: Multiple myeloma (MM) Is a B-cell malignancy characterised by the accumulation of clonal plasma cells in the bone marrow. Myeloma biology is heterogeneous and this lead to a different clinical behavior. As for other type of cancers, hypermethylation could be a mechanism of resistance in myeloma. Hence, DNA methylation of p73 tumor-suppressor gene can potentially be biomarker for diagnosis or relapse in MM. Aim: The goal of this study was to identify the expression of p73 in myeloma evaluating it in 24 different myeloma cell lines and in multiple myeloma patients. Methods: we evaluated the expression of p73 by q-PCR in 24 HMCL and 6 patients, 2 with MGUS, 2 with MM at diagnosis and 2 with relapsed/refractory MM. We tested the expression of p73 in some HMCL, one per category, by western blot. Moreover, we analyzed the expression of p73 before and after treatment with 5- aza-2-deoxycytidine. Results: 14/21 HMCL (66.7%) did not express p73 to a significant level, 4/21 (19%) have been shown to express the gene and its encoded protein to low levels and only 3/21 (14.3%) expressed p73 to an intermediate level. By western blot, we observed that the protein has a very low concentration in all three categories, considered unexpressed. We have observed that 6/12 (50%) cell lines treated increased the expression of the gene p73 after treatment with 5-aza-2- deoxycytidine, but is not reflected in increasing production of the protein detected by western blot. Finally, we observed that 2/6 (33%) patients expressed the gene at low level and 4/6 (67%) patients did not express p73. Conclusions: Our data do not provide evidence that the expression of p73 may be useful as prognostic indicators at MM diagnosis, because our study is limited to few patients tested. To better define and assess the clinical impact of the p73 expression in MM, it is therefore necessary to test the expression of p73 also in a larger number of patient samples with multiple myeloma. This knowledge may help to develop new markers for detection of minimal residual disease and determine candidate patients for epigenetically targeted salvage therapy regimens.
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Kil, Hyun Joo. "Design & Synthesis of Peptidomimetics Adopting Secondary Structures for Inhibition of p53/MDM2 Protein-protein Interaction and Multiple Myeloma Cell Adhesion." Scholar Commons, 2014. https://scholarcommons.usf.edu/etd/5051.

Повний текст джерела
Анотація:
The protein-protein interactions (PPIs) occur when two or more proteins are bound together. Also, this protein-protein interactions (PPIs) cause the various biological processes in the body. Due to this reason, abilities of controlling or inhibiting PPIs can give us promising advantages like (1) better understanding of biological systems, (2) development of new diagnostic approaches for health or disease, and (3) establishment of novel molecular therapeutics. Many proteins adopt the secondary structures, where most of protein-protein interactions take place. -Helices and -sheets are the prevalent secondary conformations, but there are extended secondary structures such as -hairpins, -turns, 310 helix, and so on. As a result, construction of molecules mimicking these protein secondary structures is tractable target for drug design. Moreover, in drug discovery, designing peptidomimetics or non-peptidic mimetics is a popular strategy instead using peptides or truncated peptides because peptides or truncated peptides are prone to proteolysis and degraded in the body. Also, peptidomimetics and non-peptidic mimetics have not only the similar topology as peptides but also resistance to proteolysis. Due to these advantages, in this study, peptidomimetics or non-peptidic mimetics were synthesized and tested for different targets: (1) synthesis of non-peptidic -helical mimetics for p53-MDM2 inhibition, (2) solution-phase synthesis of -hairpin peptide for the inhibition of multiple myeloma cells (MM) adhesion, and (3) synthesis of -hairpin peptoid-peptide hybrids. The synthesis in all three different studies was succeeded, but they still need some improvements. For instance, non-peptidic -helical mimetics, terpyrimidyl derivatives, were synthesized successfully, but they did not show any bioactivity against p53-MDM2. Also, they have a solubility problem. Based on these results, it is necessary to improve the pharmacokinetic properties and bioactivity by changing the substituents on the rings or structures. The -hairpin peptide for the second case already showed good bioactivity against multiple myeloma (MM). For the next level of bio-study, the considerable amount of a -hairpin peptide was demanded. In order to make the substantial -hairpin peptide, the solution phase peptide synthesis was chosen instead of the solid phase peptide synthesis because of the cost-effect. Two methodology were tried for the solution-phase peptide synthesis: (1) segment ligation and (2) continuous synthesis. In the former case, the -hairpin peptide synthesis was successful, but, in the latter case, it is necessary to investigate the appropriate coupling reagents for each step. Peptoid-peptide hybrids has been one of the popular peptidomimetics in the last two decades. Also, mimicking the peptide secondary structure in peptoids has been studied extensively these days. The combination of these two factors was the goal for the third case. Because peptoid-peptide hybrids with a secondary structure can be recognizable by native proteins and resistant to proteolysis. So far, three sets of peptoid-peptide hybrids were synthesize and checked the secondary structure formation by using NMR. However, there was no indication of the secondary structure formation in the three sets of peptoid-peptide hybrids. This result suggests that it is necessary to introduce the more constrained components in peptoid-peptide hybrids. In the above three chapters, it has been tried to find the new drug candidates by synthesizing peptidomimetics or non-peptidic mimetics. Even though the synthesis was successful, some intended results such as the bioactivity or the secondary structure formation were not obtained. However, these results can give us the inspirations to improve properties of peptidomimetics or non-peptidic mimetics for a certain purpose, which leads to earn the intended results and eventually find new drug candidates.
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Jain, Priyesh. "Design and Synthesis of Beta-Hairpin Peptidomimetics for Modulating Integrin Mediated Cell Adhesion, Abeta Fibrillogenesis and p53-MDM2 Protein-Protein Interactions." Scholar Commons, 2010. http://scholarcommons.usf.edu/etd/3458.

Повний текст джерела
Анотація:
Inhibiting therapeutically important protein-protein interactions has been a tremendous challenge for medicinal chemists. The folded 3D structures of peptides and proteins, mainly comprise secondary structural elements i.e α-helices and β-sheet have created an opportunity to design small molecules and peptidomimetic inhibitors of protein-protein interaction (PPI). Hence, information about the formation and stabilization of these secondary structures is vital for designing future drugs. In this dissertation, several cyclic beta-hairpin peptidomimetics that mimic the recognition surface have been designed and synthesized as inhibitors for different targets such as integrin mediated extracellular matrix -cell adhesion in multiple myeloma, p53-MDM2 PPI, amyloid beta fibrillogenesis inhibitor. Cyclization of linear peptides to restrict the number of conformations available to the linear peptide can increase its affinity for the target as well as increase its proteolytic resistance. In this study, different beta turn promoters that increase the propensity of cyclic peptides to adopt beta-sheet structures have been designed and synthesized. Chapter two discusses the design and synthesis of several cyclic III (Integrin Interaction Inhibitor) peptides that block adhesion of integrins to extracellular matrix components in Multiple Myeloma tumor cells. These cyclic peptides, as assayed by TOPRO 3 assay were more potent than the parent linear peptide with a bio-activity of 1.08 μM. We have also studied structure activity relationships (SAR) of these cyclic III peptide analogs to increase the potency and bioavailability of these peptides. Chapter three describes the application of cyclic beta-hairpin peptidomimetics to inhibit abeta fibrillogenesis that is responsible for Alzheimer’s disease. We have successfully designed and synthesized cyclic peptides that target the hydrophobic region (17-21) of abeta fibril which is believed to cause self aggregation and plaque formation. We have also successfully explored these cyclic beta-hairpin peptides to disrupt p53-MDM2 interactions. Chapter five discusses the design and synthesis of novel cysteine based Peptide Nucleic Acid (PNA) monomers that are aimed to increase cellular uptake by introducing positively charged species attached to the cysteine side chain. We have successfully synthesized CPNA monomers and made efforts to make PNA oligomers.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Ok, Claudia Barbara Verfasser], Ralf [Gutachter] Bargou, and Thomas [Gutachter] [Dandekar. "Isoform-spezifische Analyse der PI3-Kinase (Klasse I) im Multiplen Myelom / Claudia Barbara Ok [geb. Hofmann]. Gutachter: Ralf Bargou ; Thomas Dandekar." Würzburg : Universität Würzburg, 2015. http://d-nb.info/1110915373/34.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Ok, Claudia Barbara [Verfasser], Ralf C. [Gutachter] Bargou, and Thomas [Gutachter] Dandekar. "Isoform-spezifische Analyse der PI3-Kinase (Klasse I) im Multiplen Myelom / Claudia Barbara Ok [geb. Hofmann]. Gutachter: Ralf Bargou ; Thomas Dandekar." Würzburg : Universität Würzburg, 2015. http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-108466.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Hensen, Janina. "Die anti-tumorale Wirkung des neuen Phosphoinositid-3-Kinase-Inhibitors BAY 80-6946 auf humane Myelom-Zellen." Doctoral thesis, 2015. http://hdl.handle.net/11858/00-1735-0000-0023-997D-4.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Ok, [geb Hofmann] Claudia Barbara. "Isoform-spezifische Analyse der PI3-Kinase (Klasse I) im Multiplen Myelom." Doctoral thesis, 2014. https://nbn-resolving.org/urn:nbn:de:bvb:20-opus-108466.

Повний текст джерела
Анотація:
Das Multiple Myelom (MM) ist eine unheilbare Erkrankung, die aus einer klonalen Proliferation maligner Plasmazellen im Knochenmark hervorgeht. Dabei liegt ein komplexes Signalnetzwerk vor, das zum Überleben und Wachstum der MM-Zellen führt. Das MM ist durch eine enorme genetische und phänotypische Heterogenität gekennzeichnet. Die konstitutive Aktivierung des PI3K/Akt-Signalwegs spielt bei ungefähr der Hälfte der Patienten mit MM eine wichtige Rolle für das Überleben der MM-Zellen und ist daher ein potentieller therapeutischer Ansatzpunkt. Isoform-spezifische Untersuchungen der katalytischen Untereinheiten der Klasse I-PI3K (p110α, p110β, p110γ, p110δ) sollten zur Erkenntnis führen, welche dieser Isoformen für das MM Zellüberleben wichtig sind, um spezifischere Behandlungen mit möglichst geringen Nebenwirkungen zu erlauben. Dafür wurden zunächst Isoform-spezifische Knockdown-Experimente mit MM Zelllinien durchgeführt und sowohl deren Überleben als auch die Aktivierung der nachgeschalteten Komponenten im PI3K Signalweg untersucht. Zur Verifizierung der Ergebnisse wurden sowohl MM Zelllinien als auch Primärzellen mit Isoform-spezifischen PI3K-Inhibitoren behandelt (BYL 719 für p110α, TGX 221 für p110β, CAY10505 für p110γ und CAL 101 für p110δ) und in gleicher Weise untersucht. In beiden Versuchsansätzen stellte sich die katalytische Untereinheit p110α als wichtigste Isoform für das Überleben von MM Zellen mit konstitutiv phosphoryliertem Akt Signal heraus. Weder der Knockdown noch die pharmakologische Inhibition der anderen drei Isoformen (p110β, p110γ, p110δ) führten in MM-Zelllinien zur Beeinträchtigung des Zellüberlebens. Auch reagierten die Primärzellen von MM Patienten größtenteils nicht mit Apoptose auf eine Behandlung mit TGX 221, CAY10505 oder CAL 101. Aufbauend auf der postulierten Bedeutung von p110α, wurde der dafür spezifische Inhibitor BYL 719 mit bereits klinisch etablierten Therapeutika in Kombination verwendet, woraus eine im Vergleich zur Einzelbehandlung verstärkte Apoptose resultierte. Insgesamt deuten diese Daten darauf hin, dass PI3K/p110α eine therapeutisch nutzbare Zielstruktur zur Behandlung des Multiplen Myeloms darstellt. Daher scheinen weitergehende prä-klinische Studien mit p110α Inhibitoren erfolgversprechend
Multiple myeloma (MM) is an incurable disease, which results from clonal proliferation of malignant plasma cells in the bone marrow. Thereby, a complex signaling network regulates the survival and growth of MM cells. This malignant hematological disease is characterized by profound genetic and phenotypical heterogeneity. The PI3K/Akt signaling pathway is constitutively activated in about 50% of patients with MM and therefore plays an important role for the survival of MM cells. Accordingly, treatment of MM patients with the most isoform-specific drugs may be a desirable goal to achieve therapeutic utility with a minimum of undesired side effects. Therefore, an isoform-specific analysis of the catalytic subunits of the PI3K class I (p110α, p110β, p110γ, p110δ) was undertaken to reveal their individual role(s) for MM cell survival. Initially, isoform-specific knockdown experiments in MM cell lines were performed to assess their survival and the activation states of down-stream components of the PI3K pathway. These experiments were then complemented using isoform-specific pharmacological inhibitors (BYL 719 for p110α, TGX 221 for p110β, CAY10505 for p110γ and CAL 101 for p110δ) in MM cells and primary MM cells. Cell lines with constitutively phosphorylated Akt reduced this signal after p110α knockdown or pharmacologic inhibition and these treatments also affected their survival. Conversely, neither knockdown nor drug-mediated inhibition of any of the other three p110 isoforms influenced MM cell survival. In addition, whereas most primary MM samples were sensitive against BYL-719 only a few samples displayed apoptotic effects when treated with TGX 221, CAY10505 or CAL-101. These results showed that p110α is the major contributor of PI3K-mediated cell survival, and therefore the inhibitor BYL 719 was tested in combination with clinically relevant therapeutics for MM. Such treatment led to increased rates of apoptosis in MM cell lines in comparison to the respective single drug treatments. Taken together, we assume that PI3K/p110α is a therapeutically valuable target structure for the treatment of MM that would warrant more extensive pre-clinical studies
Стилі APA, Harvard, Vancouver, ISO та ін.

Частини книг з теми "P73, multiple myeloma"

1

Harvey, R. Donald, Jeannine Silberman, and Sagar Lonial. "The PI3 Kinase/Akt Pathway as a Therapeutic Target in Multiple Myeloma." In Myeloma Therapy, 309–22. Totowa, NJ: Humana Press, 2008. http://dx.doi.org/10.1007/978-1-59745-564-0_20.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Plesniar, Andrej, Gaj Vidmar, Borut tabuc, and Blanka Kores. "Effects of Recombinant Human Tumor Necrosis Factor-α and Its Combination with Native Human Leukocyte Interferon-α on P3-X63- Ag8.653 Mouse Myeloma Cell Growth." In Multiple Myeloma - An Overview. InTech, 2012. http://dx.doi.org/10.5772/31091.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Haase, AQ, SV Rajkumar, and V. Fatourechi. "Effect of Therapy with Thalidomide Derivatives on Thyroid Function in Multiple Myeloma." In The Endocrine Society's 92nd Annual Meeting, June 19–22, 2010 - San Diego, P3–589—P3–589. Endocrine Society, 2010. http://dx.doi.org/10.1210/endo-meetings.2010.part3.p12.p3-589.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.

Тези доповідей конференцій з теми "P73, multiple myeloma"

1

Matsui, William. "Abstract PL3-2: Translating multiple myeloma stem cells." In Abstracts: AACR International Conference on Translational Cancer Medicine--; Mar 21–24, 2010; Amsterdam, The Netherlands. American Association for Cancer Research, 2010. http://dx.doi.org/10.1158/1078-0432.tcme10-pl3-2.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Cottini, Francesca, Teru Hideshima, Yoshihisa Nozawa, Hiroto Ohguchi, Teruhiro Utsugi, and Kenneth C. Anderson. "Abstract PR13: p53-related protein kinase is a novel prognostic marker and therapeutic target in multiple myeloma." In Abstracts: Second AACR Conference on Hematologic Malignancies: Translating Discoveries to Novel Therapies; May 6-9, 2017; Boston, MA. American Association for Cancer Research, 2017. http://dx.doi.org/10.1158/1557-3265.hemmal17-pr13.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Lajmi, Nesrine, Sara Yousef, Tim Luetkens, Julia Templin, Carsten Bokemeyer, Nicolaus Kröger, and Djordje Atanackovic. "Abstract 4729: MAGE-C2 promotes proliferation and enhances resistance to p53 DNA damage-mediated apoptosis in multiple myeloma." In Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC. American Association for Cancer Research, 2013. http://dx.doi.org/10.1158/1538-7445.am2013-4729.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Surget, Sylvanie, Géraldine Descamps, Carole Brosseau, Philippe Moreau, Steven Le Gouill, Martine Amiot, and Catherine Pellat-Deceunynck. "Abstract 793: Preclinical study of efficacy and specificity of p53-reactivating drugs in multiple myeloma, identification of biomarkers." In Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC. American Association for Cancer Research, 2013. http://dx.doi.org/10.1158/1538-7445.am2013-793.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Teoh, Phaik Ju, Junli Yan, and Wee Joo Chng. "Abstract 786: Genomic and functional characterization identify different manifestations of p53 pathway in 17p13 deletion cases of multiple myeloma." In Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC. American Association for Cancer Research, 2013. http://dx.doi.org/10.1158/1538-7445.am2013-786.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Terragna, Carolina, Marina Martello, Enrica Borsi, Lucia Pantani, Elena Zamagni, Paola Tacchetti, Annamaria Brioli, et al. "Abstract 5595: Impact of p53 impaired function on outcomes of multiple myeloma patients carrying deleted TP53 and/or amplified MDM4." In Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA. American Association for Cancer Research, 2014. http://dx.doi.org/10.1158/1538-7445.am2014-5595.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Borrero, Liz J. Hernandez, David T. Dicker, and Wafik S. El-Deiry. "Abstract 4828: Newly identified p53 pathway-restoring compound CB002 and its derivatives sensitize colorectal and multiple myeloma cancer cell lines to front-line cancer therapies." In Proceedings: AACR Annual Meeting 2018; April 14-18, 2018; Chicago, IL. American Association for Cancer Research, 2018. http://dx.doi.org/10.1158/1538-7445.am2018-4828.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Johnson, Laura, Katelyn Trifilo, Helen Wang, Brian Kudlow, Eric Padron, Peter R. Pappenhausen, and Mohammad Hussaini. "Abstract 3988: Detection of novel t(12;17)(p12;p13) in treatment-refractory/relapsed acute myeloid leukemia by anchored multiplex PCR(AMP)-based next-generation sequencing." In Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA. American Association for Cancer Research, 2016. http://dx.doi.org/10.1158/1538-7445.am2016-3988.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії