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: Oncotarget, Impact Journals, LLC, Vol. 6, No. 27 ( 2015-09-15), p. 24218-24229
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2560162-3
    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. 2015-2015
    Abstract: Multiple myeloma (MM) is the most frequently occurring bone cancer and is characterized by malignant antibody-producing cell accumulation in bone marrow (BM). MM is still incurable, and the disease management is complicated by crosstalk between MM cells and the BM microenvironment. Why MM cells predominantly locate in bone is not well understood. The process by which MM cells are recruited into BM and reside in bone for colonization is called MM BM homing. BM homing is also critical for MM metastasis to distal BM sites. Therefore, MM BM homing is an active process throughout the disease pathogenesis. Here for the first time, we report that macrophage migration inhibitory factor (MIF) regulates MM BM homing via a mechanism that involves MM adhesion in the BM microenvironment. More importantly, we also show that MIF might be a promising target for MM treatment. MIF is an inflammatory cytokine, secreted by various human cell types. Our results here showed that BM biopsies from MM patients had significantly higher MIF expression, compared with that from healthy donors as determined by both immunohistochemistry of BM biopsies and ELISA of BM aspirates (P 〈 0.05). Animal studies using a human MM SCID mouse model (ARP-1 in the SCID mouse) and a murine MM mouse model (5-TGM1 in the C57BL/KawRij mouse) showed that intravenously inoculated (IV) MIF knocked-down (MIF-KD) MM cells did NOT form tumors in bone, whereas control knocked-down cells did form tumors in bone, as determined by both in vivo bioluminescent assay and FACS analysis of BM cells. Unlike the previously identified SDF-1/CXCR4 axis, which regulates MM chemotaxis to BM, MIF knock-down did NOT affect MM cell migration to BM. However, MIF-KD MM cells showed decreased adhesion to BM stromal cells. A mechanistic study suggested that MIF regulation of MM BM homing was mediated by MIF and its receptor CXCR4, but was independent of its primary receptor CD74. The MIF/CXCR4 axis regulated a panel of adhesion molecules via NFκB and JNK intracellular signaling, and MIF-KD and CXCR4-KD MM cells had similar downstream cell signaling and adhesion molecule expression. In addition, exposing MM cells to a MIF inhibitor (4-IPP) down-regulated expression of those adhesion molecules. Further, in the murine MM model with established MM tumors in bone, administering 4-IPP resulted in extramedullary metastasis. Overall, the findings above suggest that MIF regulates MM cell homing to the BM. We also explored MIF-targeting therapy for MM treatment. In the human MM SCID model, IV-inoculated ctrl-KD MM cells formed tumors both in BM and outside of the bone, such as in the abdomen and lung. When tumor-bearing mice were treated with the chemotherapeutic melphalan, the extramedullary tumors regressed significantly (P 〈 0.05), whereas the BM tumors were not sensitive to treatment, probably because the BM microenvironment conferred MM chemoresistance. In the same mouse model inoculated with MIF-KD MM cells, because the MIF-KD MM cells had impaired attachment and could NOT form tumors in bone, the tumor-bearing mice had a complete response to the drug treatment as determined by tumor burden and mouse survival (P 〈 0.05). These encouraging results suggested that MIF might be a valid target for MM treatment. To test this hypothesis, we used the murine MM mouse model and treated the tumor-bearing mice with both melphalan and MIF inhibitor. This combined therapy significantly repressed the MM tumor growth in vivo and lengthened survival (P 〈 0.05). Thus, MIF might be a promising target for MM treatment, in combination with conventional chemotherapy. To summarize, our findings suggest a novel function for MIF in MM BM homing regulation. MIF regulates a panel of adhesion molecules and promotes MM cell attachment within the BM. MIF inhibition disrupts the boundary between MM cells and the BM microenvironment and may raise MM cell drug sensitivity. 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 114, No. 17 ( 2009-10-22), p. 3625-3628
    Abstract: Multiple myeloma remains an incurable disease. One of the major problems is that myeloma cells develop drug resistance on interaction with bone marrow stromal cells. In this study, we examined the effects of macrophages (Mφs), a type of stromal cells, on myeloma cell survival and response to chemotherapy. We showed that Mφ, in particular tumor-associated Mφ, is a protector of myeloma cells. The protective effect was dependent on direct contact between Mφs and myeloma cells. Mφs protected both myeloma cell lines and primary myeloma cells from spontaneous and chemotherapy drug-induced apoptosis by attenuating the activation and cleavage of caspase-dependent apoptotic signaling. These findings are clinically relevant because we found that CD68+ Mφs heavily infiltrate the bone marrow of patients with myeloma but not the bone marrow of control patients. Thus, our results indicate that Mφs may contribute to myeloma cell survival and resistance to chemotherapeutic treatment in vivo.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 136, No. 22 ( 2020-11-26), p. 2557-2573
    Abstract: Multiple myeloma (MM) remains largely incurable despite significant advances in biotherapy and chemotherapy. The development of drug resistance is a major problem in MM management. Macrophage migration inhibitory factor (MIF) expression was significantly higher in purified MM cells from relapsed patients than those with sustained response, and MM patients with high MIF had significantly shorter progression-free survival (PFS) and overall survival (OS). MM cell lines also express high levels of MIF, and knocking out MIF made them more sensitive to proteasome inhibitor (PI)-induced apoptosis not observed with other chemotherapy drugs. Mechanistic studies showed that MIF protects MM cells from PI-induced apoptosis by maintaining mitochondrial function via suppression of superoxide production in response to PIs. Specifically, MIF, in the form of a homotrimer, acts as a chaperone for superoxide dismutase 1 (SOD1) to suppress PI-induced SOD1 misfolding and to maintain SOD1 activity. MIF inhibitor 4-iodo-6-phenylpyrimidine and homotrimer disrupter ebselen, which do not kill MM cells, enhanced PI-induced SOD1 misfolding and loss of function, resulting in significantly more cell death in both cell lines and primary MM cells. More importantly, inhibiting MIF activity in vivo displayed synergistic antitumor activity with PIs and resensitized PI-resistant MM cells to treatment. In support of these findings, gene-profiling data showed a significantly negative correlation between MIF and SOD1 expression and response to PI treatment in patients with MM. This study shows that MIF plays a crucial role in MM sensitivity to PIs and suggests that targeting MIF may be a promising strategy to (re)sensitize MM to the treatment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3380-3380
    Abstract: Drug resistance is the most common reason for treatment failure in multiple myeloma (MM), a plasma cell cancer of the bone marrow (BM). Our previous studies have shown that macrophage (MΦ) infiltration is increased in MM BM, and these MM-associated MΦs induce MM drug resistance by protecting MM cells from chemotherapeutic-induced cell death. A recently published clinical study also suggests that the number of BM MΦs negatively correlates with MM drug response and patient survival. Why MM BM harbors more MΦ is not well understood. In general, MΦs are derived from circulating monocytes, and monocytes are recruited to tissues by chemokines and differentiate into MΦs. In this study, we examined monocyte/MΦ chemokine expression in MM BM and determined that CCL14 and CCL3 were functional chemokines that regulated monocyte/MΦ infiltration in MM BM. To identify chemokines that regulate monocyte/MΦ infiltration in the MM tumor bed, we examined a panel of chemokines expressed in MM patient BM cells. Specifically, total BM cells (both CD138+ and CD138- cells) from MM patients were assayed by qPCR for target gene expression analysis. MM BM cells highly expressed CCL2 (MCP-1), CCL3 (MIP-1α), CCL4 (MIP-1β), CCL5 (RANTES), and CCL14 (HCC-1), but not CCL8 (MCP-2), CCL7 (MCP-3) or CCL13 (MCP-4). Next, we compared expression of those chemokines in MM BM vs. healthy BM aspirates by ELISA. MIP-1 α and HCC-1 were highly expressed in MM BM aspirates (n=11), but not in BM from healthy donors (n=7; P 〈 0.05). Immunohistochemistry staining also confirmed that MM BM (n=5 patients) highly expressed MIP-1 α and HCC-1. Based on our findings, we hypothesized that elevated MΦ infiltration in MM BM might be due to MM BM overexpression of MIP-1α and HCC-1. To test this hypothesis, we first examined MIP-1α and HCC-1 function in monocyte migration. In vitro chemotactic assay showed that adding MIP-1α or HCC-1 neutralizing antibody inhibited monocyte migration to cocultured BM stromal cells (BMSCs) and MM cells (P 〈 0.05). The antibodies also inhibited monocyte migration to ex vivo cultured BM cells from MM patients (P 〈 0.05). In a murine MM mouse model, C57BL/KawRij mice inoculated with 5TGM1, a murine MM cell line, developed tumors in hind leg bones. Both flow cytometry analysis and immunohistochemistry staining suggested that the tumor-bearing mice had an increased number of MΦs in the BM, compared with healthy mice (P 〈 0.05). Because HCC-1 does not have murine homology but it has the highest amino acid sequence similarity to MIP-1α, we treated the mice with MIP-1α neutralization antibody. Intra-peritoneal injection of mouse MIP-1α antibody decreased the MΦ number in BM (P 〈 0.05). Such in vivo findings strongly suggest that MIP-1α regulates MΦ infiltration in MM BM. We also examined the source cells that contributed to high MIP-1 α and HCC-1 expression in MM BM. Primary MM cells (CD138+) and non-malignant BM cells (CD138-) were isolated from patient (n=5) BM aspirates. qPCR analysis showed that both CD138+ and CD138- cells had high MIP-1α and HCC-1 expression. Further, BMSC/MM coculture stimulated MIP-1α overexpression in BMSCs. Finally, we examined the association between MIP-1α or HCC-1 expression and the number of BM MΦ in MM patients. The chemokine expression in BM aspirates was determined by ELISA, and the number of MΦ was measured by flow cytometry analysis for CD14+/CD68+ cells. We found that MIP-1α expression was positively associated with the number of BM MΦs (P 〈 0.05). To summarize, our findings suggest that CCL14 and CCL3 facilitate monocyte/MΦ infiltration in MM BM. Inhibiting these 2 chemokines may decrease the number of MM-associated MΦs, therefore increasing MM cell vulnerability to chemotherapy. 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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 4804-4804
    Abstract: Abstract 4804 Multiple myeloma is a B-cell malignancy characterized by the proliferation of plasma cells in the bone marrow. It is the second most common hematological malignancy and is still largely incurable. One of the major problems is that myeloma cells develop drug resistance upon interaction with bone marrow stromal cells. To better understand the importance of different stromal cell components in the bone marrow microenvironment, we examined the effects of macrophages on myeloma cell survival and myeloma cell response to chemotherapy. We report here that macrophages, in particular tumor-associated macrophages obtained by culturing macrophages with myeloma cell culture supernatants, are a protector of myeloma cells. Macrophages protected both myeloma cell lines and primary myeloma cells isolated from patients from spontaneous and chemotherapy drug-induced apoptosis via attenuating the activation of caspase-dependent apoptotic signaling. The protective effect was dependent on direct contact between macrophages and myeloma cells. Although tumor-associated macrophages secreted large amounts of IL-6, which is the most important survival factor for myeloma cells, our results showed that IL-6 neutralizing antibodies fail to significantly affect the protective effects of tumor-associated macrophages. The reduced numbers of apoptotic tumor cells in the cocultures were not the result of macrophage-uptake of apoptotic cells, because macrophages with or without the ability to phagocytose apoptotic cells provide similar protection to myeloma cells against chemotherapy-induced apoptosis. These findings are clinically relevant, because we examined bone marrow biopsies of patients by immunochemical analysis and found that CD68+ macrophages are heavily infiltrated in the bone marrow (tumor bed) of patients with myeloma but not control patients. Thus, our results indicate that macrophages are an important component of the bone marrow stromal cells and may contribute to myeloma cell survival and resistance to chemotherapeutic treatment in vivo. 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: 2009
    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: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 2643-2643
    Abstract: Abstract 2643 Mantle cell lymphoma (MCL) is a unique subtype of incurable B-cell, non-Hodgkin lymphoma, and its overall survival currently remains only 4–5 years. Management of relapsed or refractory MCL patients is still challenging. Immunotherapy may provide an alternative treatment for patients with MCL. Recent studies demonstrated that PD-1/B7-H1 signaling plays a crucial role in T-cell regulation in various immune responses and is involved in peripheral tolerance, autoimmunity, infection, and antitumor immunity. In the present study, we examined whether B7-H1 plays an important role in immune evasion in MCL. We demonstrated that B7-H1 gene and protein were expressed in most MCL cell lines and primary MCL cells from all patients examined. CD3+ T cells were cultured with irradiated MCL cell lines and primary cells, which were pre-incubated with or without anti-B7-H1 monoclonal antibody or control antibody. The presence of anti-B7-H1 blocking antibody, but not control antibody, increased CD3+ T cell proliferation. We confirmed the effect of B7-H1 in suppression of T cell proliferation by knockdown of B7-H1 gene expression using B7-H1 specific and non-specific control shRNA lentiviral particles. Upon transfection, the B7-H1-specific shRNA reduced both B7-H1 gene and surface protein expression, while the control shRNA did not. The B7-H1 specific shRNA, but not control shRNA, augmented CD3+ T cell proliferation. To address whether B7-H1 contributed to the suppression of host antitumor immunity in MCL, allogeneic CD3+ T cells isolated from normal donors were cocultured with irradiated MCL cell line SP53, control shRNA SP53 (SP53-ctl), or B7-H1 targeted shRNA SP53 (SP53-kd), respectively. After 7 days of coculture, CD3+ T cells were harvested and restimulated with newly irradiated SP53, SP53-ctl or SP53-kd cells. After at least 3 repeated cycles of in vitro restimulation, three cytotoxic T lymphocyte (CTL) lines were generated, and named CTL-SP53, CTL-SP53-ctl and CTL-SP53-kd. CTL-SP53-kd showed increased killing of target cells as compared with CTL-SP53 (P 〈 .01) or CTL-SP53-ctl (P 〈 .01). We further showed that B7-H1 targeted shRNA MCL cell line (SP53-kd cells) displayed more specific lysis than SP53 (P 〈 .01) or SP53-trl (P 〈 .01). When SP53 cells were pre-incubated with a blocking anti-B7-H1 monoclonal antibody, it also showed more specific lysis as compared to the control antibody pre-treated cells. In these experiments, purified autologous blood B cells and PBMCs were used as target cells to demonstrate whether T cell lines were cytolytic to normal cells. The results showed that the three CTL cell lines did not kill B cells or PBMCs. Intracellular cytokine staining and ELISA assay demonstrated that T-cell lines express IFN-γ, but not IL-4, IL-6, IL-10 or IL-17, and were thus type I T cells. Moreover, T cell lines stimulated by SP53-kd cells express more IFN-γ than SP53 and SP53-ctl. The T cells also expressed CD45RO, CD28, CD44, but not CD45RA, CD27 or CD62L, indicating that they were memory effector cells. In conclusion, B7-H1 expression may be involved in immune evasion mechanism of MCL. Therefore, B7-H1 may be a promising target for immunotherapy in MCL. 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: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 566-566
    Abstract: Abstract 566 Bone destruction is a hallmark of multiple myeloma (MM). More than 80% of MM patients have osteolysis, which is characterized by pathological fractures, severe bone pain, spinal cord compression, and hypercalcemia. These symptoms can severely compromise a patient's quality of life and performance status. It has been proposed that MM cells activate osteoclast (OC)-mediated bone resorption and inhibit osteoblast (OB)-mediated bone formation. However, the mechanism underlying the association of MM cells with development of bone lesions remains poorly elucidated. Our previous studies showed that p38 mitogen-activated protein kinase (MAPK), which is constitutively activated in MM cells, is a master regulator of MM-mediated bone destruction. Knocking down or inhibiting p38 MAPK activity in MM cells prevented MM-induced bone destruction in vivo. In the present study, we further investigated the mechanism of MM cell p38 MAPK-induced bone destruction. We hypothesized that p38 MAPK activity in MM cells can regulate OB and OC differentiation and activity by upregulating cytokine production by MM cells. In a cytokine array analysis, we examined the expression and secretion of MM-derived cytokines that regulate OB and OC differentiation. Our results showed for the first time that either knockdown or inhibition of p38 MAPK activity by p38 MAPK short hairpin RNAs or inhibitors significantly downregulated the production of dickkopf-1 (DKK-1) and monocyte chemotactic protein-1 (MCP-1) by MM cells. Real-time PCR and ELISA quantified and confirmed the array analysis results. To determine the role of p38 MAPK-upregulated DKK-1 and MCP-1 production in bone destruction, we administered treatment with neutralizing antibodies to SCID mice injected intravenously with ARP-1 or MM.1S cells. Our results showed that neutralization of DKK-1 and MCP-1 led to fewer bone lesions in these mice. Furthermore, we examined the impact of MM cell p38 MAPK activity on OB and/or OC differentiation. Our results showed that knockdown or inhibition of MM cell p38 MAPK significantly downregulated osteoclastogenesis but upregulated osteoblastogenesis in vitro and in vivo. Although DKK-1 is well known to inhibit OB differentiation, we found that DKK-1, together with MCP-1, promoted OC differentiation and bone resorption. Mechanistic studies further showed that MCP-1 upregulated RANK expression in OC precursors and that DKK-1 increased RANKL secretion from stromal cells and mature OBs, all of which led to activation of the NF-kB and MAPK signaling pathways in OCs. Thus, our study uncovered a novel mechanism by which p38 MAPK signaling in MM cells regulates osteoblastogenesis, osteoclastogenesis, and bone destruction in patients with this disease. These findings strongly suggest that disrupting and targeting MM cell p38 signaling are effective approaches to treating osteolytic bone lesions in MM patients. 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: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 24 ( 2012-12-15), p. 6393-6402
    Abstract: p38 mitogen-activated protein kinase (MAPK), which is constitutively activated in human myeloma, has been implicated in bone destruction by this cancer, but the processes it recruits are obscure. In this study, we show that p38 activity in myeloma inhibits osteoblast differentiation and bone formation, but also enhances osteoclast maturation and bone resorption. p38 regulated the expression and secretion of the Wnt pathway antagonist DKK-1 and the monocyte chemoattractant MCP-1. Attenuating p38, DKK-1, or MCP-1 were each sufficient to reduce bone lesions in vivo. Although it is well known that DKK-1 inhibits osteoblast differentiation, we found that together with MCP-1, it could also promote osteoclast differentiation and bone resorption. The latter effects were mediated by enhancing expression of RANK in osteoclast progenitor cells and by upregulating secretion of its ligand RANKL from stromal cells and mature osteoblasts. In summary, our study defined the mechanisms by which p38 signaling in myeloma cells regulates osteoblastogenesis, osteoclastogenesis, and bone destruction. Our findings, which may have implications for bone invasion by other cancers where p38 is elevated, strongly suggests that targeting p38 for inhibition may offer an effective therapeutic approach to treat osteolytic bone lesions in patients with myeloma. Cancer Res; 72(24); 6393–402. ©2012 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer, Wiley, Vol. 119, No. 4 ( 2013-02-15), p. 782-791
    Type of Medium: Online Resource
    ISSN: 0008-543X
    Language: English
    Publisher: Wiley
    Publication Date: 2013
    detail.hit.zdb_id: 1479932-7
    detail.hit.zdb_id: 1429-1
    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...