GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Discover Oncology, Springer Science and Business Media LLC, Vol. 13, No. 1 ( 2022-11-11)
    Abstract: The NONO protein belongs to the multifunctional family of proteins that can bind DNA, RNA and proteins. It is located in the nucleus of most mammalian cells and can affect almost every step of gene regulation. Dysregulation of NONO has been found in many types of cancer; however, data regarding its expression and relevance in Multiple Myeloma (MM) are virtually absent. Methods We took advantage of a large cohort of MM patients enrolled in the Multiple Myeloma Research Foundation CoMMpass study to elucidate better the clinical and biological relevance of NONO expression in the context of the MM genomic landscape and transcriptome. Results NONO is overexpressed in pathological samples compared to normal controls. In addition, higher NONO expression levels are significant independent prognostic markers of worse clinical outcome in MM. Our results indicate that NONO deregulation may play a pathogenetic role in MM by affecting cell cycle, DNA repair mechanisms, and influencing translation by regulating ribosome biogenesis and assembly. Furthermore, our data suggest NONO involvement in the metabolic reprogramming of glucose metabolism from respiration to aerobic glycolysis, a phenomenon known as the ‘Warburg Effect’ that supports rapid cancer cell growth, survival, and invasion. Conclusion These findings strongly support the need of future investigations for the understanding of the mechanisms of deregulation and the biological role and activity of NONO in MM.
    Type of Medium: Online Resource
    ISSN: 2730-6011
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 3059869-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Haematologica, Ferrata Storti Foundation (Haematologica), Vol. 106, No. 1 ( 2020-02-20), p. 185-195
    Abstract: Multiple Myeloma (MM) is a hematologic malignancy strongly characterized by genomic instability, which promotes disease progression and drug resistance. Since we previously demonstrated that LIG3-dependent repair is involved in the genomic instability, drug resistance and survival of MM cells, we here investigated the biological relevance of PARP1, a driver component of Alternative-Non Homologous End Joining (Alt-NHEJ) pathway, in MM. We found a significant correlation between higher PARP1 mRNA expression and poor prognosis of MM patients. PARP1 knockdown or its pharmacological inhibition by Olaparib impaired MM cells viability in vitro and was effective against in vivo xenografts of human MM. Anti-proliferative effects induced by PARP1-inhibition were correlated to increase of DNA double-strand breaks, activation of DNA Damage Response (DDR) and finally apoptosis. Importantly, by comparing a gene expression signature of PARP inhibitors (PARPi) sensitivity to our plasma cell dyscrasia (PC) gene expression profiling (GEP), we identified a subset of MM patients which could benefit from PARP inhibitors. In particular, Gene Set Enrichment Analysis (GSEA) suggested that high MYC expression correlates to PARPi sensitivity in MM. Indeed, we identified MYC as promoter of PARP1-mediated repair in MM and, consistently, we demonstrate that cytotoxic effects induced by PARP inhibition are mostly detectable on MYC-proficient MM cells. Taken together, our findings indicate that MYC-driven MM cells are addicted to PARP1 Alt-NHEJ repair, which represents therefore a druggable target in this still incurable disease.
    Type of Medium: Online Resource
    ISSN: 1592-8721 , 0390-6078
    Language: Unknown
    Publisher: Ferrata Storti Foundation (Haematologica)
    Publication Date: 2020
    detail.hit.zdb_id: 2333-4
    detail.hit.zdb_id: 2030158-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Experimental Hematology, Elsevier BV, Vol. 38, No. 2 ( 2010-02), p. 141-153
    Type of Medium: Online Resource
    ISSN: 0301-472X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 185107-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Cellular Physiology, Wiley, Vol. 220, No. 2 ( 2009-08), p. 401-409
    Abstract: Bone marrow stromal cells (MSCs) and osteoblasts are the two main non‐haematopoietic cellular components of human bone tissue. To identify novel osteoblast‐related molecules, we performed a gene expression profiling analysis comparing MSCs and osteoblasts isolated from the same donors. Genes differentially overexpressed in osteoblasts were mainly related to the negative control of cell proliferation, pro‐apoptotic processes, protein metabolism and bone remodelling. Notably, we also identified the collagen XV (COL15A1) gene as the most up‐regulated gene in osteoblasts compared with MSCs, previously described as being expressed in the basement membrane in other cell types. The expression of collagen type XV was confirmed at the protein level on isolated osteoblasts and we demonstrated that it significantly increases during the osteogenic differentiation of MSCs in vitro and that free ionised extracellular calcium significantly down‐modulates its expression. Moreover, light and electron microscopy showed that collagen type XV is expressed in bone tissue biopsies mainly by working osteoblasts forming new bone tissue or lining bone trabeculae. To our knowledge, these data represent the first evidence of the expression of collagen type XV in human osteoblasts, a calcium‐regulated protein which correlates to a specific functional state of these cells. J. Cell. Physiol. 220: 401–409, 2009. © 2009 Wiley‐Liss, Inc.
    Type of Medium: Online Resource
    ISSN: 0021-9541 , 1097-4652
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2009
    detail.hit.zdb_id: 1478143-8
    detail.hit.zdb_id: 3116-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 3107-3107
    Abstract: Background- Multiple Myeloma (MM) is a hematologic malignancy strongly characterized by genomic instability, which promotes disease progression and drug resistance. We previously demonstrated that LIG3-dependent Alt-NHEJ repair is involved in genomic instability, drug resistance and survival of MM cells. On these premises, we investigated PARP1 as driver component of Alt-NHEJ pathway and new therapeutic target in MM. Materials and methods- Cell proliferation and apoptosis were evaluated with CellTiter-Glo assay and Annexin V staining. Alt-NHEJ repair was evaluated using EJ2-GFP. PARP1, Caspase 3, MYC and DNA Damage Response protein levels were analyzed by Western blot of whole protein extracts. In vivo anti-MM activity was evaluated in NOD-SCID mice bearing subcutaneous H929 and AMO-1 Bortezomib resistant (ABZB) xenografts, daily treated with Olaparib (Selleckchem) via oral gavage. Results - By interrogating public available datasets, we found significant correlation between higher mRNA expression of PARP1 and shorter survival of MM patients. On these findings, we investigated the effect of available PARP inhibitors (PARPi) on MM cell survival. We found that Olaparib, a clinically available PARPi induced a significant reduction of proliferation and clonogenic growth of MM cell lines at low micromolar concentrations. Importantly, Olaparib impaired viability of MM cell lines or primary malignant plasmacells co-cultured with stromal cells, thus overcoming the bone marrow microenvironment supportive effect for MM survival. As result of PARP-mediated Alt-NHEJ repair inhibition, anti-proliferative effects were associated to increase of DNA double-strand breaks (DSBs), activation of DNA damage response, cell cycle arrest and finally apoptosis. To identify predictive biomarkers for PARPi in MM, a published sensitivity gene expression signature was applied to our MM gene expression profiling (GEP) data. Interestingly, this signature was particularly enriched in TC2 MM and secondary plasma cell leukemia (PCL). Therefore, in order to evaluate concordantly modulated sets of genes that were possibly associated to PARPi signature in MM, PARPi-positive and PARPi-negative MM-TC2 cases were compared by GSEA analysis. Interestingly, groups of genes regulated by MYC or involved in DNA repair resulted among the most significantly up-regulated in PARPi-positive versus PARPi-negative MM-TC2 cases. Accordingly, MYC transcript reached the highest median expression levels in sPCL and HMCLs across PC dyscrasia groups, and in MM-TC2 class. Consistently U266 cells, which was quiet insensitive to PARP knockdown or PARP inhibitor Olaparib, were null for c-MYC as compared to multiple myeloma cell lines evaluated in this study. Conversely, as formal proof of our hypothesis, over-expression of c-MYC in U266 cells (MYC-OE) induced cell death upon PARP silencing or PARP inhibitor treatment. Notably, we found that c-MYC-PARP1 loop was also hyper-activated in Bortezomib resistant cells, thus confirming pivotal role of Alt-NHEJ repair in drug resistance development. Remarkably, to demonstrate the in vivo relevance of our findings, we showed that clinically available Parp-inhibitor Olaparib exerted a significant anti-MM activity on both Bortezomib sensitive (H929) and resistant (ABZB) MM cells injected in immunocompromised mice. Conclusion Taken together, our findings indicate that MM cells are dependent on PARP-mediated Alt-NHEJ repair pathway, which therefore represents a novel druggable target pathway in MYC-driven MM cells. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1585-1585
    Abstract: Although estrogens have been shown to regulate normal B cell proliferation and differentiation, the impact of estrogen receptor (ER) signaling on B cell malignancies, including Waldenström Macroglobulinemia (WM), remains unexplored. To address this issue, we have analyzed, through preclinical WM models, the druggability either of the classical estrogen receptor ERβ, an emerging target for acute leukemias and certain types of lymphomas, and of the novel, more recently deorphanized G-protein coupled ER GPER, whose therapeutic potential in solid tumors has recently emerged. Both ERβ and GPER were found expressed at mRNA and protein level in a panel of WM and IgM-secreting lymphoma cell lines. However, while treatment with ERβ agonists did not elicit any effect, selective GPER activation significantly reduced WM cell viability. On this basis, we investigated GPER expression pattern and therapeutic potential in WM. By interrogating public microarray datasets, we found GPER transcript significantly upregulated in WM as compared to normal B cells; moreover, immunohistochemical analysis showed elevated GPER expression in lymph node biopsies from newly-diagnosed WM patients as compared to healthy lymph nodes. Treatment with the selective GPER agonist G-1 ( (±) -1-[(3aR*,4S*,9bS*)-4-(6-Bromo-1,3-benzodioxol-5-yl) -3a,4,5,9b-tetrahydro-3H cyclopenta [c]quinolin-8-yl] ethanone) led to reduced viability, clonogenicity and migration of WM and IgM-secreting lymphoma cell lines (IC50 at 48h ranging between 2.5 and 5.0 mM), including MYD88 and CXCR4 mutated cell lines, and even in the presence of bone marrow-derived stromal cells, while it did not affect healthy CD19+ B cell viability; conversely, GPER antagonists G-36 and G-15 slightly enhanced cell proliferation. The growth inhibitory activity of G-1 was associated with accumulation in G2/M cell cycle phase and induction of apoptosis, the latter phenomenon assessed by Annexin V/7AAD staining, analysis of mitochondrial membrane depolarization and cleavage of caspase 3, 7 and 9; hybridization of an antibody-array also highlighted G-1-induced down-regulation of anti-apoptotic proteins, including survivin and Bcl2. A significant effect of G-1 was also observed in primary tumor cells from refractory WM patients. Moreover, G-1 synergistically enhanced bortezomib and ibrutinib cytotoxicity in vitro. Importantly, intraperitoneal injection of G-1 (2mg/kg) significantly reduced the growth of BCWM-1 xenografts in NOD/SCID mice. To gain further insight into the consequences of selective GPER activation, we performed gene expression profile and GSEA analysis of WM cell lines treated with G-1. Of note, the p53 signaling pathway was strongly induced upon GPER activation, and upregulation of p53 and of its target genes or microRNAs (p21CIP1, Bax, Bad, PUMA and miR-34a) was confirmed both in G-1 treated WM cell lines and primary WM cells; moreover, G-1 combination with bortezomib or ibrutinib led to stronger increase in p53 and p21CIP1 levels as compared to single agent treatment. Finally, p53 knock-down partially reversed G-1-dependent anti-WM effects, thus suggesting p53 as downstream mediator of GPER. In summary, we report a significant anti-tumor activity upon selective GPER activation in WM cells, with apoptosis induction and a potent synergistic anti-WM activity in combination with ibrutinib and bortezomib. Altogether, these preclinical data suggest GPER agonists as a potential therapeutic option in WM. Disclosures Munshi: OncoPep: Other: Board of director.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Haematologica, Ferrata Storti Foundation (Haematologica), Vol. 108, No. 1 ( 2022-09-08), p. 219-233
    Abstract: Long non-coding RNA NEAT1 is the core structural component of the nuclear paraspeckle (PS) organelles and it has been found to be deregulated in multiple myeloma (MM) patients. Experimental evidence indicated that NEAT1 silencing negatively impacts proliferation and viability of MM cells, both in vitro and in vivo, suggesting a role in DNA damage repair (DDR). In order to elucidate the biological and molecular relevance of NEAT1 upregulation in MM disease we exploited the CRISPR/Cas9 synergistic activation mediator genome editing system to engineer the AMO-1 MM cell line and generate two clones that para-physiologically transactivate NEAT1 at different levels. NEAT1 overexpression is associated with oncogenic and prosurvival advantages in MM cells exposed to nutrient starvation or a hypoxic microenvironment, which are stressful conditions often associated with more aggressive disease phases. Furthermore, we highlighted the NEAT1 involvement in virtually all DDR processes through, at least, two different mechanisms. On one side NEAT1 positively regulates the posttranslational stabilization of essential PS proteins, which are involved in almost all DDR systems, thus increasing their availability within cells. On the other hand, NEAT1 plays a crucial role as a major regulator of a molecular axis that includes ATM and the catalytic subunit of DNA-PK kinase proteins, and their direct targets pRPA32 and pCHK2. Overall, we provided novel important insightsthe role of NEAT1 in supporting MM cells adaptation to stressful conditions by improving the maintenance of DNA integrity. Taken together, our results suggest that NEAT1, and probably PS organelles, could represent a potential therapeutic target for MM treatment.
    Type of Medium: Online Resource
    ISSN: 1592-8721 , 0390-6078
    Language: Unknown
    Publisher: Ferrata Storti Foundation (Haematologica)
    Publication Date: 2022
    detail.hit.zdb_id: 2333-4
    detail.hit.zdb_id: 2030158-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancers, MDPI AG, Vol. 13, No. 17 ( 2021-08-29), p. 4365-
    Abstract: Background: MYC is a master regulator of multiple myeloma (MM) by orchestrating several pro-tumoral pathways, including reprograming of the miRNA transcriptome. MYC is also involved in the acquirement of resistance to anti-MM drugs, including immunomodulatory imide drugs (IMiDs). Methods: In silico analysis was performed on MM proprietary and on public MMRF-CoMMpass datasets. Western blot and chromatin immunoprecipitation (ChIP) experiments were performed to validate miR-22 repression induced by MYC. Cell viability and apoptosis assays were used to evaluate lenalidomide sensitization after miR-22 overexpression. Results: We found an inverse correlation between MYC and miR-22 expression, which is associated with poor outcome in IMiD-treated MM patients. Mechanistically, we showed that MYC represses transcription of miR-22, which, in turn, targets MYC, thus establishing a feed-forward loop. Interestingly, we found that IMiD lenalidomide increases miR-22 expression by reducing MYC repression and, most importantly, that the combination of lenalidomide with miR-22 mimics results in a synergistic direct and NK-mediated cytotoxic activity. Conclusions: Taken together, our findings indicate that: (1) low miR-22 expression could represent a potential predictive biomarker of poor lenalidomide response in MM patients; and (2) miR-22 reduces MYC oncogenic activity, thus triggering a novel synthetic lethality loop, which sensitizes MM cells to lenalidomide.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2021
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 2740-2740
    Abstract: Multiple myeloma (MM) is a plasma cell malignancy with the high capacity to induce osteolytic bone lesions. Whereas previous studies identified genes overexpressed by MM cells related to the bone status, the occurrence of transcriptional alterations in the bone microenvironment cells in the relationship with the bone involvement has not yet investigated. To clarify this issue, in this study we have analyzed the gene expression profiling of mesenchymal (MSC) and osteoblastic (OB) cells obtained from MM patients (n=24; osteolytic n=10; non-osteolytic n=14) in relationship with the presence or absence of osteolytic bone lesions. MGUS subjects (n=7) and healthy donors (n=8) were also included in the study as controls. Both MSC and OB were isolated from trabecular bone biopsies without in vitro differentiation. The presence of potential contaminating cells was excluded by FACS analysis in both MCS and OB, testing CD3, CD14, CD20 and CD138 antigens, as well as the expression of CD105 and CD146; the osteoblast-related markers Osteocalcin, Alkaline Phosphatase, Collagen I and Runx2 were evaluated in OB in comparison with MSC. Thereafter a gene expression profiling analysis of isolated MSC and OB cells was performed using GeneChip® HG-U133A oligonucleotide arrays. The obtained data were validated by real time PCR. Using conventional hierarchical clustering, the unsupervised analyses performed of the whole dataset generated a dendrogram clearly distinguishing MSC and OB cellular types. When considering MSC and OB dataset separately, a preferential clustering in relation to the presence of osteolytic bone lesions was observed for MSC but not OB samples. A supervised multi class analysis identified a total of 84 probe sets differentially expressed in MSC with an intermediate transcriptional profile in MGUS-MSC between osteolytic and non osteolytic MM patients. A supervised analysis performed on MSC MM samples revealed a total of 49 probe-sets (36 up-regulated and 9 down-regulated genes) as differentially expressed in osteolytic vs. non-osteolytic patients. Specifically, genes belonging to Wnt signaling as WNT6 and extracellular matrix structure as decorin (DCN) were found to be down-regulated in osteolytic as compared to and non-osteolytic MSC. Interestingly, no significantly modulated genes were found by comparing osteolytic and non-osteolytic OB samples. Finally, we performed two distinct supervised analyses by comparing the two cellular types (MSC and OB) in the two groups of MM patients in relationship with the bone status. A distinct transcriptional pattern was observed in MSC versus OB between osteolytic and non-osteolytic MM patients (52 vs. 21 differentially expressed probe-sets, respectively), mainly involving cell-cycle realted genes. Our results highlight that in MM bone microenvironment MSC rather than OB show transcriptional alterations in relationship with the presence of osteolytic bone lesions in MM patients.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Hematological Oncology, Wiley, Vol. 27, No. 1 ( 2009-03), p. 23-30
    Abstract: The protein kinase C (PKC) pathway has been shown to play a role in the regulation of cell proliferation in several haematological malignancies, including multiple myeloma (MM). Recent data have shown that a PKC inhibitor, enzastaurin, has antiproliferative and proapoptotic activity in a large panel of human myeloma cell lines (HMCLs). In order to further characterise the effect of enzastaurin in MM, we performed gene expression profiling of enzastaurin‐treated KMS‐26 cell line. We identified 62 upregulated and 32 downregulated genes that are mainly involved in cellular adhesion ( CXCL12 , CXCR4 ), apoptosis ( CTSB , TRAF5 , BCL2L1 ), cell proliferation ( IGF1 , GADD45A , BCMA (B‐cell maturation antigen), CDC20 ), transcription regulation ( MYC , MX11 , IRF4 ), immune and defence responses. Subsequent validation by Western blotting of selected genes in four enzastaurin‐treated HMCLs was consistent with our microarray analysis. Our data indicate that enzastaurin may affect important processes involved in the proliferation and survival of malignant plasma cells as well as in their interactions with the bone marrow microenvironment and provide a preclinical rationale for the potential role of this drug in the treatment of MM. Copyright © 2008 John Wiley & Sons, Ltd.
    Type of Medium: Online Resource
    ISSN: 0278-0232 , 1099-1069
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2009
    detail.hit.zdb_id: 2001443-0
    detail.hit.zdb_id: 604884-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...