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: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3012-3012
    Abstract: The immunomodulatory drugs (IMiDs®) exert an anti-myeloma effect by cereblon-dependent destruction of IKZF proteinseither through a direct action on multiple myeloma (MM) cells or through indirect immunomodulatory and anti-angiogenic effects.Previous data indicated that MM cells overexpress hypoxia inducible factor (HIF)-1α and that HIF-1α suppression significantly blocks MM-induced angiogenesis and reduces in vivo tumoral burden in MM mouse models. Interestingly, it has been recently reported that HIF-1α knock-down in MM cells potentiates the in vitro effect of Lenalidomide (LEN) on cell proliferation without changing cell viability and that LEN is not able to suppress HIF-1α expression in MM cells. These evidences give the rationale design to investigate the in vivo effect of HIF-1α stable suppression in MM cells on LEN sensitivity. Thus, in this study, we assessed the effect of LEN in vivo in combination with HIF-1α inhibition in a non-obese diabetic/severe combined immunodeficiency (NOD/SCID) subcutaneous mouse model using JJN3, a cell line known to be resistant to the cytotoxic effect of LEN. Different groups of animals were injected with JJN3-pLKO.1 (empty vector) or JJN3-anti-HIF1α, obtained by anti-HIF1α lentiviral shRNA pool. When tumors became palpable, mice were treated with LEN (5mg/kg), using the intraperitoneal route. After three weeks, we evaluated tumor volume and weight. Moreover, by immunohistochemistry on ex vivo plasmacytomas, we evaluated the expression of p27 and the microvascular density (MVD), checked by CD34 immunostaining. In addition, the expression of a p27 inhibitor, the S-phase kinase-associated protein 2 (SKP2), the expression of the HIF-1α target key mediator of glycolysis and tumoral growth, Hexokinase II (HK2), and the levels of pERK 1/2, and total Caspase-3 (Casp-3) were evaluated in the ex vivo plasmacytomas lysates by western blot. We found that LEN treatment induced a significant weight and volume reduction of the tumor burden in mice injected with JJN3 anti-HIF1α as compared to JJN3-pLKO.1. The p27 nuclear expression was significantly increased by LEN treatment in JJN3-anti-HIF1α as compared to JJN3 pLKO.1 mice and compared to JJN3-anti-HIF1α mice treated with vehicle. In addition, we demonstrated that LEN in combination with HIF-1α inhibition significantly reduced in vivo the expression of p-ERK1/2, total Casp-3, HK2 and the p27 inhibitor, SKP2. Because it is known that LEN exerts its anti-MM effect targeting the IKZF proteins, we further checked in vitro whether the effect of HIF-1α suppression and LEN treatment combination could be mediated by IKZF proteins modulation. Interestingly, after LEN (2-10µM) treatment we found that both IKZF1 and IKZF3 were not differentially expressed, whereas IRF4 was down regulated, in JJN3-anti-HIF1α as compared to JJN3 pLKO.1. Finally, regarding a possible combinatory effect on the in vivo angiogenesis, we found that both the number of CD34 positive vessels and the MVD were reduced in mice colonized by JJN3-anti-HIF1α as compared to JJN3-pLKO.1. On the other hand, LEN treatment did not further significantly reduce the number of CD34 positive vessels and the MVD. Accordingly, we did not find any significant inhibitory effect by LEN treatment on the main pro-angiogenic molecules in JJN3 anti-HIF1α as compared to JJN3 pLKO.1 even after 72 hours. Overall, our data indicate that HIF-1α suppression in MM cells significantly increases the anti-MM effect of LEN in vivo, mainly through the inhibition of proliferation signaling including the modulation of p27 pathway and the IKZF target protein IRF4, rather than to an anti-angiogenic effect. These data suggest that the combination of LEN and HIF-1α inhibition has a therapeutic rationale in MM. 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: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3373-3373
    Abstract: Galectin-1 (Gal-1) is a lectin, involved in several processes related to cancer, including immunosuppression, angiogenesis, hypoxia, and metastases. However, the expression profiles of Gal-1 and its pathophysiological role in multiple myeloma (MM) cell growth, in the relationship between MM cells and the bone marrow (BM) microenvironment and in the MM-induced angiogenesis are unknown and were investigated in this study. Firstly we evaluatedGal-1 expression by CD138+ cells of a dataset of 133 MM patients at diagnosis (GSE16122) and 23 human myeloma cell lines (HMCLs) (GSE6205) or on a proprietary? dataset of primary mesenchymal stromal cells (MSCs) and osteoblasts (OBs) of 16 MM and 4 MGUS. CD138+ cells and HMCLs were positive for LGALS1 with no statistically significant differences. LGALS1 mRNA expression was positively correlated with 154 genes and negatively with 109 genes including ERG1 and SPARC. MSCs cells showed a higher expression of LGALS1 compared to the OBs and MM-OBs showed a higher expression of LGALS1 mRNA than that obtained from healthy subjects. Gene expression profiling (GEP) data were then validated by Real-Time PCR and western blot in freshly purified primary CD138+ and BM MSCs samples as well as in 6 HMCLs and in both human MSC (HS-5 and hMSC-Tert) and osteoblastic cell lines (HOBIT and HOB-01). Moreover, immunohistochemistry analyses on bone biopsies obtained from 12 MM, 9 smoldering MM, 9 MGUS and 3 plasma cell leukemia samples revealed an high level of Gal-1 protein expression by MM cells, OBs and vessels in all the patients tested. Secondly, we evaluated whether Gal-1 expression was regulated by hypoxia and by Hypoxia Inducible Factor-1a (HIF-1a) checking the effect of hypoxic treatment (1% of O2) and HIF-1α inhibition by shRNA lentivirus. We found that Gal-1 was upregulated in HMCLs upon hypoxic treatment and consistently the re-oxygenation process significantly restored the expression level of Gal-1. Interestingly the stable knock-down of HIF-1a significantly down-regulated Gal-1 expression in HMCLs both in normoxic and hypoxic conditions. Thereafter, we explored the effect of persistent Gal-1 inhibition in MM cells and BM microenvironment cells on cell proliferation, survival and the transcriptional and pro-angiogenic profiles. An anti-Gal-1 Lentivirus shRNA was used for Gal-1 stable knock-down in HMCLs (JJN3-anti-Gal-1 and OPM-2-anti-Gal-1) and MSC cell lines (HS-5 and HMSC-Tert) and the Scramble lentiviral vector (JJN3-Scramble and OPM-2-Scramble) was used as the empty control vector. The stable inhibition of Gal-1 did not affect the proliferation rate and viability of both HMCLs and MSC cell lines. On the other hand Gal-1 inhibition by shRNA lentivirus significantly modified the transcriptional profiles of HMCLs and HS-5, evaluated by U133 Plus2.0 Arrays (Affymetrix®) either in normoxic or hypoxic or re-oxygenation conditions. Among the genes significantly modulated by Gal-1 inhibition in HMCLs, we found that pro-angiogenic (eg. CCL2, MMP9) and adhesion molecules (eg. MCAM and STEAP1) were down-regulated by Gal-1 suppression in both normoxic and hypoxic conditions as well as some putative anti-tumoral genes, including EGR1, SPARC and TGFBI, and anti-angiogenic ones, including SEMA3A, were up-regulated by Gal-1 inhibition. In line with these observations, we found that Gal-1 suppression by shRNA significantly decreased the pro-angiogenic proprieties of HMCLs by an in vitro angiogenesis assay. Finally, we found that mice, injected subcutaneously with JJN3-anti-Gal-1 and OPM-2-anti-Gal-1, showed a reduction in the weight and volume of the tumor burden compared to mice inoculated with the JJN3-Scramble and OPM-2-Scramble. Moreover, a significant reduction in the number of CD34 positive vessels X field was observed. In an intratibial mouse model, JJN3-anti-Gal-1, JJN3-Scramble and JJN3 wild type were injected: in the anti-Gal-1 group tumors grew in reduced number and size compared to the Scramble group, moreover JJN3 anti-Gal-1 mice developed fewer and smaller lytic lesions on x-ray compared to the controls. Overall our data indicate that Gal-1 is highly expressed by MM cells and those of the BM microenvironment and that its expression is regulated by hypoxia. Gal-1 shows a role in MM-induced angiogenesis and its inhibition in MM cells significantly reduced tumor growth in vivo, suggesting that Gal-1 is a potential new therapeutic target in MM. Disclosures Giuliani: Celgene Italy: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancers, MDPI AG, Vol. 12, No. 11 ( 2020-11-05), p. 3267-
    Abstract: Multiple myeloma (MM) cells consume huge amounts of glutamine and, as a consequence, the amino acid concentration is lower-than-normal in the bone marrow (BM) of MM patients. Here we show that MM-dependent glutamine depletion induces glutamine synthetase in stromal cells, as demonstrated in BM biopsies of MM patients, and reproduced in vitro by co-culturing human mesenchymal stromal cells (MSCs) with MM cells. Moreover, glutamine depletion hinders osteoblast differentiation of MSCs, which is also severely blunted by the spent, low-glutamine medium of MM cells, and rescued by glutamine restitution. Glutaminase and the concentrative glutamine transporter SNAT2 are induced during osteoblastogenesis in vivo and in vitro, and both needed for MSCs differentiation, pointing to enhanced the requirement for the amino acid. Osteoblastogenesis also triggers the induction of glutamine-dependent asparagine synthetase (ASNS), and, among non-essential amino acids, asparagine rescues differentiation of glutamine-starved MSCs, by restoring the transcriptional profiles of differentiating MSCs altered by glutamine starvation. Thus, reduced asparagine availability provides a mechanistic link between MM-dependent Gln depletion in BM and impairment of osteoblast differentiation. Inhibition of Gln metabolism in MM cells and supplementation of asparagine to stromal cells may, therefore, constitute novel approaches to prevent osteolytic lesions in MM.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2020
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Bone and Mineral Research, Wiley, Vol. 31, No. 4 ( 2016-04), p. 815-827
    Abstract: Multiple myeloma (MM) is characterized by severely imbalanced bone remodeling. In this study, we investigated the potential effect of proteasome inhibitors (PIs), a class of drugs known to stimulate bone formation, on the mechanisms involved in osteocyte death induced by MM cells. First, we performed a histological analysis of osteocyte viability on bone biopsies on a cohort of 37 MM patients with symptomatic disease. A significantly higher number of viable osteocytes was detected in patients treated with a bortezomib (BOR)‐based regimen compared with those treated without BOR. Interestingly, both osteocyte autophagy and apoptosis were affected in vivo by BOR treatment. Thereafter, we checked the in vitro effect of BOR to understand the mechanisms whereby BOR maintains osteocyte viability in bone from MM patients. We found that osteocyte and preosteocyte autophagic death was triggered during coculturing with MM cells. Our evaluation was conducted by analyzing either autophagy markers microtubule‐associated protein light chain 3 beta (LC3B) and SQSTM1/sequestome 1 (p62) levels, or the cell ultrastructure by transmission electron microscopy. PIs were found to increase the basal levels of LC3 expression in the osteocytes while blunting the myeloma‐induced osteocyte death. PIs also reduced the autophagic death of osteocytes induced by high‐dose dexamethasone (DEX) and potentiated the anabolic effect of PTH(1‐34). Our data identify osteocyte autophagy as a new potential target in MM bone disease and support the use of PIs to maintain osteocyte viability and improve bone integrity in MM patients. © 2015 American Society for Bone and Mineral Research.
    Type of Medium: Online Resource
    ISSN: 0884-0431 , 1523-4681
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2016
    detail.hit.zdb_id: 2008867-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Hematology & Oncology, Springer Science and Business Media LLC, Vol. 13, No. 1 ( 2020-12)
    Abstract: The oncolytic viruses have shown promising results for the treatment of multiple myeloma. However, the use of human viruses is limited by the patients’ antiviral immune response. In this study, we investigated an alternative oncolytic strategy using non-human pathogen viruses as the bovine viral diarrhea virus (BVDV) that were able to interact with CD46. Methods We treated several human myeloma cell lines and non-myeloma cell lines with BVDV to evaluate the expression of CD46 and to study the effect on cell viability by flow cytometry. The possible synergistic effect of bortezomib in combination with BVDV was also tested. Moreover, we infected the bone marrow mononuclear cells obtained from myeloma patients and we checked the BVDV effect on different cell populations, defined by CD138, CD14, CD3, CD19, and CD56 expression evaluated by flow cytometry. Finally, the in vivo BVDV effect was tested in NOD-SCID mice injected subcutaneously with myeloma cell lines. Results Human myeloma cells were selectively sensitive to BVDV treatment with an increase of cell death and, consequently, of apoptotic markers. Consistently, bone marrow mononuclear cells isolated from myeloma patients treated with BVDV, showed a significant selective decrease of the percentage of viable CD138 + cells. Interestingly, bortezomib pre-treatment significantly increased the cytotoxic effect of BVDV in myeloma cell lines with a synergistic effect. Finally, the in vitro data were confirmed in an in vivo myeloma mouse model showing that BVDV treatment significantly reduced the tumoral burden compared to the vehicle. Conclusions Overall, our data indicate, for the first time, a direct oncolytic effect of the BVDV in human myeloma cells suggesting its possible use as novel alternative anti-myeloma virotherapy strategy.
    Type of Medium: Online Resource
    ISSN: 1756-8722
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2429631-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2959-2959
    Abstract: The relationship between multiple myeloma (MM) cells and osteoblast (OB)s and osteoclast (OCL)s into the bone marrow (BM) niche has a critical role in the pathophysiology of MM and in the development of bone disease in MM patients. Recently, Daratumumab (DARA), a human anti-CD38 monoclonal antibody has been developed with broad-spectrum killing activity including complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and, at least in part, modulation of CD38 enzymatic activity. In pre-clinical studies, DARA has been shown to effectively kill MM cells and clinical trials are ongoing. However, the effects of DARA in the context of the MM bone niche and on MM-induced bone remodeling alterations are unknown. In this study, we have firstly evaluated the expression profile of CD38 and its related ectoenzymes by bone niche cells and, thereafter, we investigated the effect of DARA on OCL and OB formation and bone remodeling. The study design included immunohistochemistry for CD38, CD73, CD39, CD203a (PC-1), CD157 and CD31 on bone biopsies in a cohort of 38 patients with newly diagnosed MM and 14 patients with monoclonal gammopathy of uncertain significance (MGUS). Moreover, the expression profile of these antigens was performed by flow cytometry on primary BM CD138+ cells (sample number=16), mesenchymal stromal cells, OBs, monocytes and OCLs and on the related human cell lines. As expected, we found that CD38 was highly expressed by MM cells that were also positive for CD203a and for CD39 and CD31 at variable level but not for CD157 and CD73. CD38 was also expressed by BM monocytes but not by OBs, OCLs and BM stromal cells. Interestingly, we found that OBs expressed CD73 and CD203a. Any significant difference was not observed in the expression of CD38 and related ectoenzymes between MM and MGUS patients. In line with CD38 expression profile by MM cells and the niche, we further demonstrated that DARA binds both MM cells and monocytes, but not OBs and OCLs. Consistently, we lack to find any significant effect of DARA on OB formation from BM stromal cells or OBs proliferation and survival. Thus, we investigated the effect of DARA (1-25 ug/ml) as compared to human IgG isotype control on OCL formation and activity in the presence of RANKL and M-CSF, using either CD138- cell fraction or purified CD14+ cells from MM BM samples. OCLs were evaluated by both TRAP staining and a fluorimetric osteolysis assay. We found that DARA, between 10 and 25 ug/ml, with a dose dependent effect, significantly inhibited OCL formation and activity from BM mononuclear cells and from the CD138- cell fraction, but not from purified CD14+ cells. The inhibitory effect on OCL formation by DARA was observed when the antibody was present for all the culture period (21-28 days). On the other hand DARA did not show any effect on late OCL progenitors and mature OCLs. Accordingly CD38 expression by monocytes cultured in a pro-osteoclastogenic medium disappeared after 7 days. In conclusion, our data indicate that DARA inhibit osteoclastogenesis, likely mediated by ADCC, targeting monocytes and early OCL progenitors. This evidence supports the use of an anti-CD38 based approach as a treatment for MM bone disease. Disclosures Malavasi: Janssen: Honoraria, Research Funding. Giuliani:Celgene Italy: Other: Research Grant; Janssen Pharamceutical: Other: Research Grant.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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: Oncotarget, Impact Journals, LLC, Vol. 8, No. 34 ( 2017-08-22), p. 56598-56611
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Oncotarget, Impact Journals, LLC, Vol. 7, No. 21 ( 2016-05-24), p. 30109-30118
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2016
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancers, MDPI AG, Vol. 11, No. 3 ( 2019-03-06), p. 321-
    Abstract: C-X3-C motif chemokine ligand 1 (CX3CL1)/fractalkine is a chemokine released after cleavage by two metalloproteases, ADAM metallopeptidase domain 10 (ADAM10) and ADAM metallopeptidase domain 17 (ADAM17), involved in inflammation and angiogenesis in the cancer microenvironment. The role of the CX3CL1/ C-X3-C motif chemokine receptor 1(CX3CR1) axis in the multiple myeloma (MM) microenvironment is still unknown. Firstly, we analyzed bone marrow (BM) plasma levels of CX3CL1 in 111 patients with plasma cell disorders including 70 with active MM, 25 with smoldering myeloma (SMM), and 16 with monoclonal gammopathy of undetermined significance (MGUS). We found that BM CX3CL1 levels were significantly increased in MM patients compared to SMM and MGUS and correlated with BM microvessel density. Secondly, we explored the source of CX3CL1 in MM and BM microenvironment cells. Primary CD138+ cells did not express CXC3L1 but up-regulated its production by endothelial cells (ECs) through the involvement of tumor necrosis factor alpha (TNFα). Lastly, we demonstrated the presence of CX3CR1 on BM CD14+CD16+ monocytes of MM patients and on ECs, but not on MM cells. The role of CX3CL1 in MM-induced angiogenesis was finally demonstrated in both in vivo chick embryo chorioallantoic membrane and in vitro angiogenesis assays. Our data indicate that CX3CL1, present at a high level in the BM of MM patients, is a new player of the MM microenvironment involved in MM-induced angiogenesis.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2019
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 5315-5315
    Abstract: The malignant plasma cells (PC) dependence on the bone marrow (BM) microenvironment is a main feature of multiple myeloma (MM), mainly due to high expression of cell adhesion molecules including CD44, CD54 (ICAM1), CD56, CD49d and chemokines receptors such as CXCR4. At the end stage of the disease, a rare MM complication, that may occurs, is a secondary extramedullary involvement without a leukemic phase and, among the extramedullary localizations, the skin is one of the possible sites, due to the physiological homing of PCs. However, the mechanisms behind the extramedullary spread are not completely understood. In a patient with refractory resistant MM, who has developed a cutaneous localization after 16 months from the diagnosis under Bortezomib (BOR) treatment, we analyzed the gene expression profiles (GEPs) by GeneChip U133 Plus 2.0 (Affymetrix), the immunophenotypic and immunohistochemistry (IHC) profiles of MM cells, across the course of the disease. To confirm the results, IHC profiles on selected molecules were then analyzed in other six MM patients with secondary skin involvement without PC leukemia. On BM CD138+ PCs at diagnosis and BM relapse, by GEP analysis, 742 genes resulted differentially expressed. Moreover, a more stringent analysis identified 19 down-regulated and 42 up-regulated genes; data were confirmed by Real Time PCR on selected genes mainly involved in PC homing. At the BM relapse, we found that MM cells expressed CXCR4 and were negative for CD56; moreover, a significant down-regulation of ICAM1 (CD54) and ALCAM was observed together with the up-regulation of MAGE family genes, DKK1 and SEMA3A in the BM relapse sample compared to diagnosis one. At the cutaneous involvement development, 4 months after BM relapse, by IHC analysis, the MM cells localized in the skin showed the expression of both CD56 and CD54. On the other hand, the BM MM cells immunophenotype confirmed the presence of CD56 but showed the lack of CXCR4 expression. IHC for CD54, CD56 and CXCR4 expression was then performed on both skin and bone biopsies of the other patients analyzed. To go further inside on the role of CXCR4 we treated with BOR (5-10nM) for 24-48h human myeloma cell lines (HMCLs) RPMI-8226 and U266, known to be resistant to BOR, evaluating the CXCR4 expression at both mRNA and protein level, by real-time PCR and by cytofluorimetry, respectively. CXCR4 resulted downregulated after BOR treatment in BOR resistant HMCLs. Consistently, the loss of CXCR4 expression was recently associated to BOR resistance and extramedullary disease in a mouse model of MM suggesting that CXCR4 loss can be correlated with BOR resistance and PCs mobilization from BM, leading to an extramedullary disease. In conclusion, our data indicate that the loss of CXCR4 expression by MM cells across the disease progression, together with CD54 and CD56 down-regulation and their re-acquisition at the extramedullary site, are involved driving the escape of PCs from BM to the extramedullary localization in the skin in the context of Bortezomib resistance. 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: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    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...