Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Journal of Translational Medicine, Springer Science and Business Media LLC, Vol. 20, No. 1 ( 2022-03-10)
    Abstract: Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in nicotinamide adenine dinucleotide (NAD) biosynthesis, is up-regulated in several cancers, including metastatic melanoma (MM). The BRAF oncogene is mutated in different cancer types, among which MM and thyroid carcinoma (THCA) are prominent. Drugs targeting mutant BRAF are effective, especially in MM patients, even though resistance rapidly develops. Previous data have linked NAMPT over-expression to the acquisition of BRAF resistance, paving the way for therapeutic strategies targeting the two pathways. Methods Exploiting the TCGA database and a collection of MM and THCA tissue microarrays we studied the association between BRAF mutations and NAMPT expression. BRAF wild-type (wt) cell lines were genetically engineered to over-express the BRAF V600E construct to demonstrate a direct relationship between over-activation of the BRAF pathway and NAMPT expression. Responses of different cell line models to NAMPT (i)nhibitors were studied using dose–response proliferation assays. Analysis of NAMPT copy number variation was performed in the TCGA dataset. Lastly, growth and colony forming assays were used to study the tumorigenic functions of NAMPT itself. Results The first finding of this work is that tumor samples carrying BRAF -mutations over-express NAMPT, as demonstrated by analyzing the TCGA dataset, and MM and THC tissue microarrays. Importantly, BRAF wt MM and THCA cell lines modified to over-express the BRAF V600E construct up-regulated NAMPT, confirming a transcriptional regulation of NAMPT following BRAF oncogenic signaling activation. Treatment of BRAF -mutated cell lines with two different NAMPTi was followed by significant reduction of tumor growth, indicating NAMPT addiction in these cells. Lastly, we found that several tumors over-expressing the enzyme, display NAMPT gene amplification. Over-expression of NAMPT in BRAF wt MM cell line and in fibroblasts resulted in increased growth capacity, arguing in favor of oncogenic properties of NAMPT. Conclusions Overall, the association between BRAF mutations and NAMPT expression identifies a subset of tumors more sensitive to NAMPT inhibition opening the way for novel combination therapies including NAMPTi with BRAFi/MEKi, to postpone and/or overcome drug resistance. Lastly, the over-expression of NAMPT in several tumors could be a key and broad event in tumorigenesis, substantiated by the finding of NAMPT gene amplification.
    Type of Medium: Online Resource
    ISSN: 1479-5876
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2118570-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 126, No. 1 ( 2015-11-30), p. 181-194
    Type of Medium: Online Resource
    ISSN: 0021-9738 , 1558-8238
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2015
    detail.hit.zdb_id: 2018375-6
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 10 ( 2019-7-25)
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2019
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3286-3286
    Abstract: Lenalidomide is an immunomodulatory agent clinically active in CLL patients. The specific mechanism of action is still undefined, but includes the modulation of microenvironment. In CLL patients, nurse-like cells (NLCs) differentiate from CD14+ mononuclear cells and nurture/protect CLL cells from apoptosis. NLCs resemble M2 macrophages with potent immunosuppressive functions. We examined the ability of lenalidomide to mediate a pro-inflammatory switch of NLCs affecting the protective microenvironment generated by CLL into tissues. NLCs were generated in presence or absence of lenalidomide: cell surface markers, phagocytosis and induction of T cell proliferation were analyzed after 10 days. NLCs activation was measured using a yellow tetrazolium MTT assay after 5 days of culture and NLCs proliferation was measured by CFSE staining. Microarray-based gene expression profiles of NLCs treated or not with lenalidomide were evaluated after 10 days and data were confirmed by real time PCR. Lenalidomide modifies the immunophenotype and the biological characteristics of NLCs. First, treatment with lenalidomide 0.5µM and 1µM increased the number of NLCs to 268% and 309% compared to untreated control (100%) respectively (p 〈 0.05). To explain the high number of NLCs generated by lenalidomide, we analyzed cell activation and proliferation. We observed a strong increase in NLCs activation after treatment with lenalidomide that correlated with stimulation of NLCs proliferation from 44% to 55% (% of dividing cells) (p 〈 0.05). In contrast with the high number of NLCs generated in presence of lenalidomide, we found that NLCs lost the ability to nurture and protect CLL cell from apoptosis reducing their viability from 54.2% to 44.5% (p 〈 0.05), but they strongly attracted CLL cells reaching an increase of adhesion to 227% and 212% with the addition of 0.5µM and 1µM lenalidomide (p 〈 0.05). Accordingly with these results, we investigated the ability of lenalidomide to interfere with leukemia-promoting activity of NLCs. Lenalidomide improved the ability of NLCs to engulf zymogen particles to 141% and 155% with dosage of 0.5µM and 1µM compared to control (100%) (p 〈 0.05), further confirmed analyzing the uptake of FITC-dextran by NLCs that increased to 252% and 356% compared to untreated control (p 〈 0.01). Moreover lenalidomide strongly improved the ability of NLCs to induce T cells proliferation from 19.5% to 35.0% (% of dividing cells) (p 〈 0.05). Lastly, gene expression profiling showed a switch to a pro-inflammatory profile in NLCs induced by treatment with lenalidomide involving a modulation of pivotal genes for the immune response, activation/proliferation of T cells, complement activation as well as regulation of cellular movement, cytokine and chemokine activation. In particular, down-regulation of CCL2, CXCL12, IL-10, CD163 and up-regulation of IL-2 were apparent. Collectively, our data provide new insights into the mechanism of action of lenalidomide that reverts NLCs polarization from M2 to M1-skewed phenotype affecting the supporting and protective microenvironment generated by CLL into tissues. Disclosures Maffei: Celgene: support for travel to congresses Other. Marasca:Celgene: Honoraria, 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3316-3316
    Abstract: Cancer-associated microenvironment provides to malignant cells the right cocktail of signals to increase survival, reprogram their metabolism and escape the control of the immune system. Nicotinamide phosphoribosyltransferase (NAMPT) is the only human enzyme that metabolizes nicotinamide, the main NAD precursor, in a rate-limiting step in NAD biosynthesis. Beside this canonical activity, NAMPT can be secreted in the extracellular milieu (eNAMPT), where it exerts cytokine/adipokine-like actions in different tumor models, as well as in acute and chronic inflammatory-metabolic diseases. Here we investigated the functional role of eNAMPT in the microenvironment of chronic lymphocytic leukemia (CLL), an indolent lymphoproliferative disorder, strongly dependent on a growth supportive environment. Results indicate that: i) B-CLL lymphocytes express higher levels of NAMPT mRNA and intracellular protein than normal circulating B lymphocytes obtained from age- and sex-matched donors; ii) plasma levels of eNAMPT are also significantly higher in CLL patients (n=95) compared to controls (n=20) and iii) eNAMPT is produced by CLL lymphocytes upon B-cell receptor, toll-like receptor and NF-KB signaling pathway activation. We then asked whether this cytokine plays an active role in the leukemic microenvironment. Data shows that eNAMPT acts on resting monocytes, polarizing them towards tumor-supporting M2 macrophages, known as nurse-like cells (NLC) in CLL. These cells express high levels of the scavenger receptor CD163, mannose receptor CD206 and indoleamine 2,3-dioxygenase (IDO), and secrete immunosuppressive (IL-10, CCL18) and tumor-promoting (IL-6, IL-8) cytokines. NAMPT-primed NLCs activate ERK1/2, STAT3 and NF-kB signaling, promote leukemic cell survival and reduce T cell responses. These effects are independent of the enzymatic activity of NAMPT, as inferred from the use of an enzymatically inactive mutant. From the translational point of view, drugs interfering with CLL signaling, such as Ibrutinib, can efficiently suppress NAMPT transcription, reducing NAMPT protein levels. Furthermore, NLCs differentiated in the presence of the immunomodulatory drug lenalidomide express lower levels of NAMPT, again suggesting that drugs that restore immune functions interfere with the production of eNAMPT. To conclude, we propose that a vicious circle based on CLL cell activation through antigen and accessory signals increase eNAMPT and CCL3 production. CCL3 serves as an attractant for circulating monocytes, which - in the presence of high levels of eNAMPT - differentiate into NLC, with an enhanced M2 phenotype and increased functional characteristics, contributing to CLL survival, activation and proliferation and inhibition of T cell responses. 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: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 125, No. 1 ( 2015-01-01), p. 111-123
    Abstract: CLL lymphocytes show high intracellular and extracellular NAMPT levels, further increased upon activation. eNAMPT prompts differentiation of CLL monocytes into M2 macrophages that sustain CLL survival and reduce T-cell proliferation.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 613-613
    Abstract: Introduction: In lymphoid organs, nurse-like cells (NLCs) show properties of tumor-associated macrophages with a M2-like phenotype, associated to prominent immunosuppressive functions, and play a crucial role in CLL survival. Ibrutinib is a potent inhibitor of BTK and ITK kinases and is able to counteract the microenvironmental-derived pro-survival effects in CLL cells. One of the main effects of ibrutinib treatment is attenuated retention and homing of CLL cells to tissue compartments, which ultimately leads to mobilization of CLL cells from microenvironmental niches into the peripheral blood. Aim: Here, we investigated whether ibrutinib treatment may have an effect on the immunosuppressive properties of NLCs. Methods: NLCs were generated in complete medium after 12 days of in vitro culture of CLL-PBMCs. Cell activation status, cell surface markers, real time PCR and functional studies were performed after 1h or 24h of ibrutinib treatment, to test whether ibrutinib influenced M1 or M2-polarization of NLCs. Results: We first observed that, after ibrutinib treatment, the activation status and viability of NLCs were not affected, the morphology of NLCs in vitro analyzed by flow-cytometric FCS and SSC plots was not modified and the number of NLCs generated after 12 days of culture was preserved. Furthermore, treatment with ibrutinib stimulated the expression of the M2-associated markers hemoglobin scavenger receptor CD163 and the mannose receptor C type 1 CD206, and induced expression of the monocyte differentiation antigen CD14. At the gene expression level, ibrutinib induced strong expression of genes involved in M2 polarization such as CD163 (293%, p 〈 0.01), IL10 (279%, p 〈 0.05), MRC1/CD206 (180%, p 〈 0.05) and CCL18 (271%, p 〈 0.05), and concomitantly decreased the expression of M1 polarization genes such as IL1 (53%, p 〈 0.01), TNFα (57%, p 〈 0.01) and IL2 (11%, p 〈 0.01), as compared to the untreated control (100%). Of interest, treatment with ibrutinib stimulated also the expression of the enzyme nicotinamide phosphoribosyltransferase (NAMPT) to 141% (p 〈 0.05) and the programmed death-ligand 1 (PD-L1) to 133% (p 〈 0.01) in NLCs, both important in the induction of an immunosuppressive and protective microenvironment in CLL. Overall these data suggest that ibrutinib could stimulate features related to suppression of the immune system. Next, we tested the ability of ibrutinib to regulate actin cytoskeletal reorganization, which is an essential process during macrophages phagocytosis. Upon ibrutinib stimulation, NLCs showed low actin polymerization levels, since F-actin formation decreased to 66% (p 〈 0.05) as compared to unstimulated control (100%). In addition, ibrutinib impaired the phagocytic activity of NLCs and functionally affected the expression of MAC-1 (CD11b/CD18), which is required for optimal phagocytosis, from 100% to 86% (p 〈 0.01). Conclusions: Collectively, these preliminary data provide new insights into the effects of ibrutinib treatment on the modulation of immune elements in CLL tissue microenvironment. Noteworthy, our data demonstrate that ibrutinib further promotes M2-polarization features of NLCs, suggesting that this drug not only has an effect on the CLL clone but also extensively influences the cellular components of the CLL microenvironment, whether these effects may be related to ibrutinib-resistance warrants further investigations. Disclosures Burger: Boehringer Ingelheim Pharma: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Noxxon: Membership on an entity's Board of Directors or advisory committees; Portola Pharmaceuticals: Research Funding; Gilead: Research Funding; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees, Research Funding. Marasca:Janssen: Honoraria, Other: support for travel to congresses.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 133, No. 2 ( 2019-01-10), p. 156-167
    Abstract: Proteasome inhibitors (PI) are extensively used for the therapy of multiple myeloma (MM) and mantle cell lymphoma. However, patients continuously relapse or are intrinsically resistant to this class of drugs. Here, to identify targets that synergize with PI, we carried out a functional screening in MM cell lines using a short hairpin RNA library against cancer driver genes. Isocitrate dehydrogenase 2 (IDH2) was identified as a top candidate, showing a synthetic lethal activity with the PI carfilzomib (CFZ). Combinations of US Food and Drug Administration–approved PI with a pharmacological IDH2 inhibitor (AGI-6780) triggered synergistic cytotoxicity in MM, mantle cell lymphoma, and Burkitt lymphoma cell lines. CFZ/AGI-6780 treatment increased death of primary CD138+ cells from MM patients and exhibited a favorable cytotoxicity profile toward peripheral blood mononuclear cells and bone marrow–derived stromal cells. Mechanistically, the CFZ/AGI-6780 combination significantly decreased tricarboxylic acid cycle activity and adenosine triphosphate levels as a consequence of enhanced IDH2 enzymatic inhibition. Specifically, CFZ treatment reduced the expression of nicotinamide phosphoribosyltransferase (NAMPT), thus limiting IDH2 activation through the NAD+-dependent deacetylase SIRT3. Consistently, combination of CFZ with either NAMPT or SIRT3 inhibitors impaired IDH2 activity and increased MM cell death. Finally, inducible IDH2 knockdown enhanced the therapeutic efficacy of CFZ in a subcutaneous xenograft model of MM, resulting in inhibition of tumor progression and extended survival. Taken together, these findings indicate that NAMPT/SIRT3/IDH2 pathway inhibition enhances the therapeutic efficacy of PI, thus providing compelling evidence for treatments with lower and less toxic doses and broadening the application of PI to other malignancies.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 240-240
    Abstract: Background. The tumor microenvironment is typically an immunosuppressive niche often characterized by low oxygen tension, representing an intrinsic limitation to the success of immunotherapeutic approaches. Several lines of evidence indicate hypoxia is a master regulator of the adenosinergic axis, up-regulating on one side expression of CD39 and CD73, the two enzymes that generate adenosine starting from ATP/ADP and on the other side the adenosine receptors, which are powerful inhibitors of immune responses. Our previous studies using primary samples indicate that CLL cells can produce extracellular adenosine and that the CLL microenvironment is rich in expression of the A2A receptor, which potently inhibits T cell responses and skews macrophages towards type 2 responses(1, 2). Aim of the work. The main aims of this work are i) to dynamically study expression and activity of the adenosinergic axis using an in vivo model of CLL, and ii) to investigate whether its targeting may restore immune responses. Results. We exploited the TCL1 mouse model of CLL by adoptively transferring different leukemias into immunocompetent wild-type C57BL/6 mice and following the re-organization of the microenvironment and the reshaping of immune responses during disease progression. The first observation is that the appearance of large leukemic nodules that subvert normal splenic architecture is accompanied by increased staining with the hypoxia marker pimonidazole and by lactic acidosis, as witnessed by progressive increase in LDH activity within the tissue. The second observation concerns the onset of progressive immunosuppression with appearance of terminally differentiated and dysfunctional T lymphocytes and skewing of classical inflammatory monocytes to anti-inflammatory patrolling monocytes. Leukemia development is also accompanied by increased adenosine bio-synthetic potential due to the up-regulation of CD39 and CD73 and to the marked and generalized up-regulation of the A2A adenosine receptor, which is evident in all cells of the microenvironment, including leukemic cells, T lymphocytes and macrophages, as documented by real-time PCR studies on purified populations and by flow cytometry and immunohistochemistry analyses. This finding suggests that A2A may be a common mediator of immune suppression and that it may be a suitable therapeutic target. To test this hypothesis, we adoptively transferred different TCL1 leukemias in naïve C57BL/6 mice, allowed engraftment for 10 days and then treated with the commercially available SCH58621 A2A inhibitor every other day for two weeks (1mg/kg, intra-peritoneally). At the end of treatment, mice were euthanized and immune features examined as above. Results very consistently showed that A2A inhibition increased the naïve component of both CD4+ and CD8+ subsets, with a concomitant partial reduction in effector T lymphocytes and in regulatory T cells. Moreover, both CD4+ and CD8+ subsets recovered their cytotoxic functions as indicated by the production of IFN-γ and IL-2. When examining the macrophage compartment, SCH58261 repolarized monocytes, by increasing the inflammatory subset at the expense of patrolling monocytes. Conclusions. Taken together, these results highlight the relevance of the adenosinergic axis in the creation and maintenance of a tumor microenvironment that favors immune escape. This could be due, at least in part, to the presence of a highly hypoxic leukemic niche. Interrupting this network using an A2A receptor antagonist restores the functions of the different immune cell subsets. Therefore, these data suggest that the adenosinergic axis may represent a good target in treatment strategies that combine anti-leukemic drugs with agents able to repolarize the CLL environment towards immune competence.Serra S, et al. (2016) Adenosine signaling mediates hypoxic responses in the chronic lymphocytic leukemia microenvironment. 1(1).doi:10.1182/bloodadvances.2016000984.The.Serra S, et al. (2011) CD73-generated extracellular adenosine in chronic lymphocytic leukemia creates local conditions counteracting drug-induced cell death. 118(23):6141-6153. Disclosures Deaglio: iTeos therapeutics: Research Funding; Verastem: Research Funding; VelosBio inc: Research Funding.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal of the American Society of Nephrology, Ovid Technologies (Wolters Kluwer Health), Vol. 32, No. 5 ( 2021-5), p. 1114-1130
    Abstract: FSGS is an important cause of end-stage renal failure, but the mechanisms leading to podocyte injury or conferring protection against FSGS remain poorly understood. The cytosolic protein M-Sec has been involved in the formation of tunneling nanotubes (TNTs), membrane channels that transiently connect cells and allow intercellular organelle transfer. The authors found that the cytosolic protein M-Sec, which is involved in the development of TNTs, is induced in FSGS and allows replacement of dysfunctional podocyte organelles via TNTs, whereas M-Sec abrogation leads to FSGS onset. These findings reveal the M-Sec–TNT system plays an important protective role in podocytes and may represent a novel therapeutic target in FSGS. Demonstration that dysfunctional podocyte mitochondria can be replaced through horizontal TNT-mediated transfer may set the stage for strategies of regenerative cell-based therapy. Background Podocyte dysfunction and loss are major determinants in the development of proteinuria. FSGS is one of the most common causes of proteinuria, but the mechanisms leading to podocyte injury or conferring protection against FSGS remain poorly understood. The cytosolic protein M-Sec has been involved in the formation of tunneling nanotubes (TNTs), membrane channels that transiently connect cells and allow intercellular organelle transfer. Whether podocytes express M-Sec is unknown and the potential relevance of the M-Sec–TNT system in FSGS has not been explored. Methods We studied the role of the M-Sec–TNT system in cultured podocytes exposed to Adriamycin and in BALB/c M-Sec knockout mice. We also assessed M-Sec expression in both kidney biopsies from patients with FSGS and in experimental FSGS (Adriamycin-induced nephropathy). Results Podocytes can form TNTs in a M-Sec–dependent manner. Consistent with the notion that the M-Sec–TNT system is cytoprotective, podocytes overexpressed M-Sec in both human and experimental FSGS. Moreover, M-Sec deletion resulted in podocyte injury, with mitochondrial abnormalities and development of progressive FSGS. In vitro , M-Sec deletion abolished TNT-mediated mitochondria transfer between podocytes and altered mitochondrial bioenergetics. Re-expression of M-Sec reestablishes TNT formation and mitochondria exchange, rescued mitochondrial function, and partially reverted podocyte injury. Conclusions These findings indicate that the M-Sec–TNT system plays an important protective role in the glomeruli by rescuing podocytes via mitochondrial horizontal transfer. M-Sec may represent a promising therapeutic target in FSGS, and evidence that podocytes can be rescued via TNT-mediated horizontal transfer may open new avenues of research.
    Type of Medium: Online Resource
    ISSN: 1046-6673 , 1533-3450
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2021
    detail.hit.zdb_id: 2029124-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages