Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
1.
PLoS One ; 15(2): e0225820, 2020.
Article En | MEDLINE | ID: mdl-32040476

NK cell migration and activation are crucial elements of tumor immune surveillance. In mammary carcinomas, the number and function of NK cells is diminished, despite being positively associated with clinical outcome. MicroRNA-155 (miR-155) has been shown to be an important regulator of NK cell activation through its interaction with SHIP-1 downstream of inhibitory NK receptor signaling, but has not been explored in regard to NK cell migration. Here, we explored the migratory potential and function of NK cells in subcutaneous AT3 in mice lacking miR-155. Without tumor, these bic/miR-155-/- mice possess similar numbers of NK cells that exhibit comparable surface levels of cytotoxic receptors as NK cells from wild-type (WT) mice. Isolated miR-155-/- NK cells also exhibit equivalent cytotoxicity towards tumor targets in vitro compared to isolated WT control NK cells, despite overexpression of known miR-155 gene targets. NK cells isolated from miR-155-/- mice exhibit impaired F-actin polymerization and migratory capacity in Boyden-chamber assays in response chemokine (C-C motif) ligand 2 (CCL2). This migratory capacity could be normalized in the presence of SHIP-1 inhibitors. Of note, miR-155-/- mice challenged with mammary carcinomas exhibited heightened tumor burden which correlated with a lower number of tumor-infiltrating NK1.1+ cells. Our results support a novel, physiological role for SHIP-1 in the control of NK cell tumor trafficking, and implicate miR-155 in the regulation of NK cell chemotaxis, in the context of mammary carcinoma. This may implicate dysfunctional NK cells in the lack of tumor clearance in mice.


Killer Cells, Natural/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Mammary Neoplasms, Experimental/metabolism , MicroRNAs/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Animals , Cell Line, Tumor , Cell Movement/genetics , Chemotaxis/genetics , Female , Gene Knockout Techniques , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Signal Transduction/genetics
2.
Oncoimmunology ; 8(4): e1557372, 2019.
Article En | MEDLINE | ID: mdl-30906652

Immune escape is a hallmark of cancer. In human lung cancer, we have identified a unique microRNA (miR)-based pathway employed by tumor cells to repress detection by immune cells via the NKG2D-MICA/B receptor-ligand system. MICA/B is readily induced by cell transformation and serves as a danger signal and ligand to alert NK and activated CD8+ T cells. However, immunohistochemical analysis indicated that human lung adenocarcinoma and squamous cell carcinoma specimens express little MICA/B while high levels of miR-183 were detected in both tumor types in a TCGA database. Human lung tumor cell lines confirmed the reverse relationship in expression of MICA/B and miR-183. Importantly, a miR-183 binding site was identified on the 3'untranslated region (UTR) of both MICA and MICB, suggesting its role in MICA/B regulation. Luciferase reporter constructs bearing the 3'UTR of MICA or MICB in 293 cells supported the function of miR-183 in repressing MICA/B expression. Additionally, anti-sense miR-183 transfection into H1355 or H1299 tumor cells caused the upregulation of MICA/B. Abundant miR-183 expression in tumor cells was traced to transforming growth factor-beta (TGFß), as evidenced by antisense TGFß transfection into H1355 or H1299 tumor cells which subsequently lost miR-183 expression accompanied by MICA/B upregulation. Most significantly, anti-sense miR-183 transfected tumor cells became more sensitive to lysis by activated CD8+ T cells that express high levels of NKG2D. Thus, high miR-183 triggered by TGFß expressed in lung tumor cells can target MICA/B expression to circumvent detection by NKG2D on immune cells.

3.
Front Immunol ; 9: 2887, 2018.
Article En | MEDLINE | ID: mdl-30574153

Acute graft- vs. -host disease (GVHD) is an important cause of morbidity and death after allogeneic hematopoietic cell transplantation (HCT). We identify a new approach to prevent GVHD that impairs monocyte-derived dendritic cell (moDC) alloactivation of T cells, yet preserves graft- vs.-leukemia (GVL). Exceeding endoplasmic reticulum (ER) capacity results in a spliced form of X-box binding protein-1 (XBP-1s). XBP-1s mediates ER stress and inflammatory responses. We demonstrate that siRNA targeting XBP-1 in moDCs abrogates their stimulation of allogeneic T cells. B-I09, an inositol-requiring enzyme-1α (IRE1α) inhibitor that prevents XBP-1 splicing, reduces human moDC migration, allo-stimulatory potency, and curtails moDC IL-1ß, TGFß, and p40 cytokines, suppressing Th1 and Th17 cell priming. B-I09-treated moDCs reduce responder T cell activation via calcium flux without interfering with regulatory T cell (Treg) function or GVL effects by cytotoxic T lymphocytes (CTL) and NK cells. In a human T cell mediated xenogeneic GVHD model, B-I09 inhibition of XBP-1s reduced target-organ damage and pathogenic Th1 and Th17 cells without impacting donor Tregs or anti-tumor CTL. DC XBP-1s inhibition provides an innovative strategy to prevent GVHD and retain GVL.


Dendritic Cells/immunology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Immunosuppression Therapy/methods , Leukemia/therapy , X-Box Binding Protein 1/antagonists & inhibitors , Animals , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Knockdown Techniques , Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Isoantibodies/immunology , Isoantibodies/metabolism , Isoantigens/immunology , Leukemia/immunology , Lymphocyte Activation/drug effects , Male , Mice , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Skin Transplantation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation Chimera , Transplantation, Homologous/adverse effects , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/immunology , X-Box Binding Protein 1/metabolism , Xenograft Model Antitumor Assays
4.
Sci Rep ; 6: 29521, 2016 07 13.
Article En | MEDLINE | ID: mdl-27405665

Myeloid-derived suppressor cells (MDSCs) constitute a key checkpoint that impedes tumor immunity against cancer. Chemotherapeutic intervention of MDSCs has gained ground as a strategy for cancer therapy but its mechanism remains obscure.We report here a unique mechanism by which monocytic (M)-MDSCs are spared, allowing them to polarize towards M1 macrophages for reactivation of immunity against breast cancer. We first demonstrated that curcumin, like docetaxel (DTX), can selectively target CD11b(+)Ly6G(+)Ly6C(low) granulocytic (G)-MDSCs, sparing CD11b(+)Ly6G(-)Ly6C(high) M-MDSCs, with reduced tumor burden in 4T1-Neu tumor-bearing mice. Curcumin treatment polarized surviving M-MDSCs toward CCR7(+) Dectin-1(-)M1 cells, accompanied by IFN-γ production and cytolytic function in T cells. Selective M-MDSC chemoresistence to curcumin and DTX was mediated by secretory/cytoplasmic clusterin (sCLU). sCLU functions by trapping Bax from mitochondrial translocation, preventing the apoptotic cascade. Importantly, sCLU was only found in M-MDSCs but not in G-MDSCs. Knockdown of sCLU in M-MDSCs and RAW264.7 macrophages was found to reverse their natural chemoresistance. Clinically, breast cancer patients possess sCLU expression only in mature CD68(+) macrophages but not in immature CD33(+) immunosuppressive myeloid cells infiltrating the tumors. We thus made the seminal discovery that sCLU expression in M-MDSCs accounts for positive immunomodulation by chemotherapeutic agents.


Antineoplastic Agents/administration & dosage , Clusterin/metabolism , Curcumin/administration & dosage , Mammary Neoplasms, Experimental/drug therapy , Myeloid-Derived Suppressor Cells/drug effects , Animals , Antigens, Ly/metabolism , Antineoplastic Agents/pharmacology , CD11b Antigen/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/pharmacology , Female , Interferon-gamma/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/immunology , RAW 264.7 Cells , Xenograft Model Antitumor Assays
5.
Cancer Res ; 76(5): 1009-18, 2016 Mar 01.
Article En | MEDLINE | ID: mdl-26833128

It remains unclear how localized radiotherapy for cancer metastases can occasionally elicit a systemic antitumor effect, known as the abscopal effect, but historically, it has been speculated to reflect the generation of a host immunotherapeutic response. The ability to purposefully and reliably induce abscopal effects in metastatic tumors could meet many unmet clinical needs. Here, we describe a mathematical model that incorporates physiologic information about T-cell trafficking to estimate the distribution of focal therapy-activated T cells between metastatic lesions. We integrated a dynamic model of tumor-immune interactions with systemic T-cell trafficking patterns to simulate the development of metastases. In virtual case studies, we found that the dissemination of activated T cells among multiple metastatic sites is complex and not intuitively predictable. Furthermore, we show that not all metastatic sites participate in systemic immune surveillance equally, and therefore the success in triggering the abscopal effect depends, at least in part, on which metastatic site is selected for localized therapy. Moreover, simulations revealed that seeding new metastatic sites may accelerate the growth of the primary tumor, because T-cell responses are partially diverted to the developing metastases, but the removal of the primary tumor can also favor the rapid growth of preexisting metastatic lesions. Collectively, our work provides the framework to prospectively identify anatomically defined focal therapy targets that are most likely to trigger an immune-mediated abscopal response and therefore may inform personalized treatment strategies in patients with metastatic disease.


Cell Movement , Lymphocyte Activation , Neoplasms/radiotherapy , T-Lymphocytes/immunology , Humans , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/physiology
6.
Sci Rep ; 5: 15085, 2015 Oct 15.
Article En | MEDLINE | ID: mdl-26469759

Development of chemoresistance, especially to docetaxel (DTX), is the primary barrier to the cure of castration-resistant prostate cancer but its mechanism is obscure. Here, we report a seminal crosstalk between dying and residual live tumor cells during treatment with DTX that can result in outgrowth of a chemoresistant population. Survival was due to the induction of secretory/cytoplasmic clusterin (sCLU), which is a potent anti-apoptotic protein known to bind and sequester Bax from mitochondria, to prevent caspase 3 activation. sCLU induction in live cells depended on HMGB1 release from dying cells. Supernatants from DTX-treated DU145 tumor cells, which were shown to contain HMGB1, effectively induced sCLU from newly-plated DU145 tumor cells and protected them from DTX toxicity. Addition of anti-HMBG1 to the supernatant or pretreatment of newly-plated DU145 tumor cells with anti-TLR4 or anti-RAGE markedly abrogated sCLU induction and protective effect of the supernatant. Mechanistically, HMGB1 activated NFκB to promote sCLU gene expression and prevented the translocation of activated Bax to mitochondria to block cell death. Importantly, multiple currently-used chemotherapeutic drugs could release HMGB1 from tumor cells. These results suggest that acquisition of chemoresistance may involve the HMGB1/TLR4-RAGE/sCLU pathway triggered by dying cells to provide survival advantage to remnant live tumor cells.


Clusterin/metabolism , Drug Resistance, Neoplasm , HMGB1 Protein/metabolism , Prostatic Neoplasms/metabolism , Apoptosis/genetics , Cell Line, Tumor , Clusterin/genetics , Down-Regulation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , HMGB1 Protein/pharmacology , Humans , Male , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Recombinant Proteins/pharmacology , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/metabolism
7.
Proc Natl Acad Sci U S A ; 111(11): 4203-8, 2014 Mar 18.
Article En | MEDLINE | ID: mdl-24586048

Transforming growth factor ß1 (TGF-ß), enriched in the tumor microenvironment and broadly immunosuppressive, inhibits natural killer (NK) cell function by yet-unknown mechanisms. Here we show that TGF-ß-treated human NK cells exhibit reduced tumor cytolysis and abrogated perforin polarization to the immune synapse. This result was accompanied by loss of surface expression of activating killer Ig-like receptor 2DS4 and NKp44, despite intact cytoplasmic stores of these receptors. Instead, TGF-ß depleted DNAX activating protein 12 kDa (DAP12), which is critical for surface NK receptor stabilization and downstream signal transduction. Mechanistic analysis revealed that TGF-ß induced microRNA (miR)-183 to repress DAP12 transcription/translation. This pathway was confirmed with luciferase reporter constructs bearing the DAP12 3' untranslated region as well as in human NK cells by use of sense and antisense miR-183. Moreover, we documented reduced DAP12 expression in tumor-associated NK cells in lung cancer patients, illustrating this pathway to be consistently perturbed in the human tumor microenvironment.


Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Killer Cells, Natural/immunology , Membrane Proteins/antagonists & inhibitors , MicroRNAs/metabolism , Neoplasms/immunology , Receptors, Natural Killer Cell/antagonists & inhibitors , Transforming Growth Factor beta/immunology , Adaptor Proteins, Signal Transducing/metabolism , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Killer Cells, Natural/metabolism , Luciferases , Membrane Proteins/metabolism , Microscopy, Fluorescence , Receptors, Natural Killer Cell/metabolism , Signal Transduction/immunology , Transforming Growth Factor beta/metabolism
8.
Cancer Immunol Immunother ; 61(4): 523-33, 2012 Apr.
Article En | MEDLINE | ID: mdl-21983879

Large granular lymphocyte (LGL) leukemia is a chronic lymphoproliferative disease in which T-bet [T-box transcription factor 21 gene (tbx21)] overexpression may play a pathogenic role. T-bet orchestrates the differentiation of mature peripheral T-cells into interferon-γ (IFN-γ) and tumor necrosis factor-α producing CD4+ T-helper type I (Th1) and CD8+ T cytotoxic cells that are necessary for antiviral responses. When IL-12 is produced by antigen-presenting cells, T-bet expression is induced, causing direct stimulation of ifng gene transcription while simultaneously acting as a transcriptional repressor of the IL4 gene, which then leads to Th1 dominance and T-helper type 2 differentiation blockade. Additionally, T-bet has been shown to regulate histone acetylation of the ifng promoter and enhancer to loosen condensed DNA, creating greater accessibility for other transcription factor binding, which further amplifies IFNγ production. We found that treatment with a farnesyltransferase inhibitor tipifarnib reduced Th1 cytokines in LGL leukemia patient T-cells and blocked T-bet protein expression and IL-12 responsiveness in T-cells from healthy donors. The mechanism of suppression was based on modulation of histone acetylation of the ifng gene, which culminated in Th1 blockade.


Antineoplastic Agents/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Leukemia, Large Granular Lymphocytic/immunology , Quinolones/pharmacology , T-Box Domain Proteins/metabolism , Th1 Cells/drug effects , Th2 Cells/drug effects , Acetylation/drug effects , Adult , Aged , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunosuppression Therapy , Leukemia, Large Granular Lymphocytic/pathology , Male , Middle Aged , Signal Transduction/drug effects , T-Box Domain Proteins/genetics , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th1-Th2 Balance/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology
9.
PLoS One ; 6(6): e21173, 2011.
Article En | MEDLINE | ID: mdl-21738617

Several reports link cigarette smoking with leukemia. However, the effects of cigarette smoke extract (CSE) on bone marrow hematopoiesis remain unknown. The objective of this study was to elucidate the direct effects of cigarette smoke on human bone marrow hematopoiesis and characterize the inflammatory process known to result from cigarette smoking. Bone marrow mononuclear cells (BMCs) from healthy individuals when exposed to CSE had significantly diminished CFU-E, BFU-E and CFU-GM. We found increased nuclear translocation of the NF-κB p65 subunit and, independently, enhanced activation of AKT and ERK1/2. Exposure of BMCs to CSE induced IL-8 and TGF-ß1 production, which was dependent on NF-κB and ERK1/2, but not on AKT. CSE treatment had no effect on the release of TNF-α, IL-10, or VEGF. Finally, CSE also had a significant induction of TLR2, TLR3 and TLR4, out of which, the up-regulation of TLR2 and TLR3 was found to be dependent on ERK1/2 and NF-κB activation, but not AKT. These results indicate that CSE profoundly inhibits the growth of erythroid and granulocyte-macrophage progenitors in the bone marrow. Further, CSE modulates NF-κB- and ERK1/2-dependent responses, suggesting that cigarette smoking may impair bone marrow hematopoiesis in vivo as well as induce inflammation, two processes that proceed malignant transformation.


Bone Marrow Cells/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Smoking/adverse effects , Toll-Like Receptors/metabolism , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interleukin-10/metabolism , Interleukin-8/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Int Immunopharmacol ; 11(7): 890-8, 2011 Jul.
Article En | MEDLINE | ID: mdl-21244860

3, 5,7-trihydroxy-4'-methoxy-8-(3-hydroxy-3-methylbutyl)-flavone (ICT) is a novel derivative of Icariin (ICA), the major active ingredient of Herba Epimedii, a herb used in traditional Chinese and alternative medicine. We previously demonstrated its anti-inflammatory effect in murine innate immune cells and activated human PBMCs. We report herein that ICA or ICT treatment reduces the expression of MRP8/MRP14 and toll-like receptor 4 (TLR4) on human PBMCs. Administration of ICA or ICT inhibited tumor growth in 4T1-Neu tumor-bearing mice and considerably decreased MDSC numbers in the spleen of these mice. Further, we saw a restoration of IFN-γ production by CD8+ T cells in tumor bearing mice when treated with ICA or ICT. ICA and ICT significantly decreased the amounts of nitric oxide and reactive oxygen species in MDSC in vivo. When MDSC were treated in vitro with ICT, we saw a significant reduction in the percent of these cells with concomitant differentiation into dendritic cells and macrophages. Concomitant with this cell type conversion was a down-regulation of IL-10, IL-6 and TNF-α production. Decreased expression of S100A8/9 and inhibition of activation of STAT3 and AKT may in part be responsible for the observed results. In conclusion, our results showed that ICA, and more robustly, ICT, directly modulate MDSC signaling and therefore altered the phenotype and function of these cells, in vitro and in vivo.


CD8-Positive T-Lymphocytes/metabolism , Flavonoids/administration & dosage , Myeloid Cells/drug effects , Neoplasms, Experimental/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Antineoplastic Agents/administration & dosage , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Cell Differentiation/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , Epimedium/immunology , Flavones/administration & dosage , Humans , Immunosuppression Therapy , Mice , Mice, Nude , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 16(18): 4583-94, 2010 Sep 15.
Article En | MEDLINE | ID: mdl-20702612

PURPOSE: Myeloid-derived suppressor cells (MDSC) accumulate in tumor-bearing hosts and are associated with immune suppression. To date, there have only been few studies that evaluate the direct effect of chemotherapeutic agents on MDSCs. Agents that inhibit MDSCs may be useful in the treatment of patients with various cancers. EXPERIMENTAL DESIGN: We investigated the in vivo effects of docetaxel on immune function in 4T1-Neu mammary tumor-bearing mice to examine if a favorable immunomodulatory effect accompanies tumor suppression. Primary focus was on the differentiation status of MDSCs and their ability to modulate T-cell responses. RESULTS: Docetaxel administration significantly inhibited tumor growth in 4T1-Neu tumor-bearing mice and considerably decreased MDSC proportion in the spleen. The treatment also selectively increased CTL responses. Docetaxel-pretreated MDSCs cocultured with OT-II splenocytes in the presence of OVA(323-339) showed OT-II-specific CD4 activation and expansion in vitro. In characterizing the phenotype of MDSCs for M1 (CCR7) and M2 [mannose receptor (CD206)] markers, MDSCs from untreated tumor bearers were primarily MR(+) with few CCR7(+) cells. Docetaxel treatment polarized MDSCs toward an M1-like phenotype, resulting in 40% of MDSCs expressing CCR7 in vivo and in vitro, and macrophage differentiation markers such as MHC class II, CD11c, and CD86 were upregulated. Interestingly, docetaxel induced cell death selectively in MR(+) MDSCs while sparing the M1-like phenotype. Finally, inhibition of signal transducer and activator of transcription 3 may in part be responsible for the observed results. CONCLUSIONS: These findings suggest potential clinical benefit for the addition of docetaxel to current immunotherapeutic protocols.


Immunomodulation/drug effects , Myeloid Cells/drug effects , Neoplasms/immunology , T-Lymphocytes/drug effects , Taxoids/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic/drug effects , Docetaxel , Female , Immune Tolerance/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myeloid Cells/immunology , Myeloid Cells/physiology , Neoplasm Transplantation , Neoplasms/drug therapy , T-Lymphocytes/immunology , Taxoids/therapeutic use
12.
Mol Cancer Ther ; 9(6): 1831-41, 2010 Jun.
Article En | MEDLINE | ID: mdl-20501799

Clusterin (CLU), in its cytoplasmic form, is abundant in many advanced cancers and has been established to be cytoprotective against chemotherapeutic agents including docetaxel. However, little is known of the mechanism of its induction. Here, we provide evidence that AKT plays a critical role in upregulating cytoplasmic/secretory sCLU, which is responsible for docetaxel resistance. Western blot analysis indicated that docetaxel-resistant sublines derived from DU145 and PC3 prostate tumor cell lines displayed a markedly increased phospho-AKT level closely accompanied by heightened sCLU expression when compared with parental cells. To examine if AKT has a role in sCLU expression, AKT blockade was done by treatment with a specific inhibitor, API-2, or dominant-negative AKT transduction before analysis of sCLU gene expression. Loss of AKT function resulted in loss of sCLU and was accompanied by chemosensitization to docetaxel and increased cell death via a caspase-3-dependent pathway. To confirm that AKT affected resistance to docetaxel through sCLU and not through other mediators, tumor cells were first transfected with full-length CLU for overexpression and then treated with the AKT inhibitor API-2. We found that once sCLU was overexpressed, API-2 could not chemosensitize the tumor cells to docetaxel. Thus, the chemoresistance to docetaxel is mediated by sCLU and it can be induced by AKT. Lastly, AKT was found to mediate sCLU induction via signal transducer and activator of transcription 1 activation, which we have earlier shown to drive sCLU gene expression. These results identify a previously unrecognized pathway linking AKT to cytoprotection by sCLU in tumor cells.


Antineoplastic Agents/pharmacology , Clusterin/metabolism , Cytoprotection/drug effects , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Taxoids/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Clusterin/genetics , Docetaxel , Down-Regulation/drug effects , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Dominant , Humans , Male , Phenotype , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , STAT1 Transcription Factor/metabolism , Up-Regulation/drug effects
13.
Adv Cancer Res ; 105: 77-92, 2009.
Article En | MEDLINE | ID: mdl-19879424

Resistance to anticancer agents is one of the primary impediments to effective cancer therapy. Chemoresistance occurs not only to clinically established therapeutic agents but also to novel targeted therapeutics. Both intrinsic and acquired mechanisms have been implicated in drug resistance but it remains controversial which mechanisms are responsible that lead to failure of therapy in cancer patients. Recent focus has turned to clusterin (CLU) as a key contributor to chemoresistance to anticancer agents. Its role has been documented in prostate cancer for paclitaxel/docetaxel resistance as well as in renal, breast, and lung tumor cells. Moreover, it is abnormally upregulated in numerous advanced stage and metastatic cancers spanning prostate, renal, bladder, breast, head and neck, colon, cervical, pancreatic, lung carcinomas, melanoma, and lymphoma. It is noteworthy that only the cytoplasmic/secretory clusterin form (sCLU), and not the nuclear form, is expressed in aggressive late stage tumors, which is in line with its antiapoptotic function. Most significantly, sCLU expression is documented to lead to broad-based resistance to other unrelated chemotherapeutic agents such as doxorubicin, cisplatin, etoposide, and camphothecin. Resistance to targeted death-inducing molecules, tumor necrosis factor, Fas and TRAIL, or histone deacetylase inhibitors can also be mediated by sCLU. Expression of sCLU may be an adaptive response to genotoxic and oxidative stresses but this adaptive response could pose a threat in malignant cells being treated with cytotoxic agents by enhancing their survival potential. The actual mechanisms for sCLU induction are unclear but STAT1 is required for its constitutive upregulation in docetaxel-resistant tumor cells. Known as a protein chaperone, sCLU appears to stabilize Ku70/Bax complexes, sequestering Bax from its ability to induce mitochondrial release of cytochrome c that triggers cell apoptosis. Thus, sCLU has a key role in preventing apoptosis induced by cytotoxic agents and has the potential to be targeted for cancer therapy.


Clusterin/physiology , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Clusterin/antagonists & inhibitors , Clusterin/genetics , Drug Resistance, Multiple , Humans , Neoplasms/metabolism , Neoplasms/pathology , Oxidative Stress
14.
Proc Natl Acad Sci U S A ; 106(31): 12974-9, 2009 Aug 04.
Article En | MEDLINE | ID: mdl-19470455

Lenalidomide is the first karyotype-selective therapeutic approved for the treatment of myelodysplastic syndromes (MDS) owing to high rates of erythroid and cytogenetic response in patients with chromosome 5q deletion [del(5q)]. Although haploinsufficiency for the RPS14 gene and others encoded within the common deleted region (CDR) have been implicated in the pathogenesis of the del(5q) phenotype, the molecular basis of the karyotype specificity of lenalidomide remains unexplained. We focused our analysis on possible haplodeficient enzymatic targets encoded within the CDR that play key roles in cell-cycle regulation. We show that the dual specificity phosphatases, Cdc25C and PP2Acalpha, which are coregulators of the G(2)-M checkpoint, are inhibited by lenalidomide. Gene expression was lower in MDS and acute myeloid leukemia (AML) specimens with del(5q) compared with those with alternate karyotypes. Lenalidomide inhibited phosphatase activity either directly (Cdc25C) or indirectly (PP2A) with corresponding retention of inhibitory phospho-tyrosine residues. Treatment of del(5q) AML cells with lenalidomide induced G(2) arrest and apoptosis, whereas there was no effect in nondel(5q) AML cells. Small interfering RNA (shRNA) suppression of Cdc25C and PP2Acalpha gene expression recapitulated del(5q) susceptibility to lenalidomide with induction of G(2) arrest and apoptosis in both U937 and primary nondel(5q) MDS cells. These data establish a role for allelic haplodeficiency of the lenalidomide inhibitable Cdc25C and PP2Acalpha phosphatases in the selective drug sensitivity of del(5q) MDS.


Antineoplastic Agents/pharmacology , Chromosome Deletion , Chromosomes, Human, Pair 5 , Myelodysplastic Syndromes/drug therapy , Protein Phosphatase 2/antagonists & inhibitors , Thalidomide/analogs & derivatives , cdc25 Phosphatases/antagonists & inhibitors , Apoptosis/drug effects , G2 Phase/drug effects , Humans , Lenalidomide , Myelodysplastic Syndromes/genetics , Protein Phosphatase 2/genetics , Thalidomide/pharmacology , U937 Cells , cdc25 Phosphatases/genetics
15.
Int Immunol ; 21(4): 423-32, 2009 Apr.
Article En | MEDLINE | ID: mdl-19261694

NK cell cytotoxicity requires two positive signals for killing of tumors. Activation receptors induce polarization of the microtubule organization center and degranulation, while leukocyte function-associated antigen (LFA)-1 is required for conjugate formation and actin polymerization and under some circumstances may be sufficient for NK cell cytotoxicity. Although the receptor for direct killing of fungi is not known, CD18, the beta2 chain of LFA-1, binds components of the capsule and cell wall of the opportunistic pathogen Cryptococcus neoformans, namely the polysaccharides glucoronoxylomannan and galactoxylomannan. Herein, we also demonstrate that LFA-1 was concentrated in regions of the NK cell surface interacting with C. neoformans. Consequently, there was compelling evidence to hypothesize that NK cells would also use LFA-1 to recognize and kill C. neoformans. Using a combination of NK cell lines that did or did not express LFA-1 or by using a CD18-specific functional blocking antibody, we confirm that NK cell anti-tumor activity is critically dependent upon the expression of LFA-1. Duplicating the events of tumor cytotoxicity, NK cells form conjugates with cryptococcal targets, rearrange the cell cytoskeleton to develop an NK immunologic synapse and release perforin-containing granules; however, each of these events occurred independently of LFA-1. Furthermore, NK cell-mediated killing of C. neoformans was detectable in both NK cells pre-treated with CD18-blocking antibodies and in NK cells lacking cell surface LFA-1 expression. These results demonstrate that in the absence of LFA-1 expression, NK cells are fully capable of recognizing a target (C. neoformans) and retain all of the events required for cytotoxicity.


CD18 Antigens/immunology , Cryptococcus neoformans/immunology , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Neoplasms/immunology , Actins/immunology , Actins/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , CD18 Antigens/metabolism , Cell Degranulation/immunology , Cell Line, Tumor , Cytoskeleton/immunology , Cytoskeleton/metabolism , Humans , Killer Cells, Natural/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Perforin/immunology , Perforin/metabolism , Signal Transduction/immunology
17.
Blood ; 113(14): 3226-34, 2009 Apr 02.
Article En | MEDLINE | ID: mdl-19075187

Large granular lymphocyte (LGL) leukemia, or LGLL, is characterized by increased numbers of circulating clonal LGL cells in association with neutropenia, anemia, rheumatoid arthritis, and pulmonary artery hypertension (PAH). Emerging evidence suggests that LGLL cells with a CD8(+)CD28(null) phenotype induce these clinical manifestations through direct destruction of normal tissue. Compared with CD8(+)CD28(null) T cells from healthy controls, CD8(+)CD28(null) T cells from LGLL patients have acquired the ability to directly lyse pulmonary artery endothelial cells and human synovial cells. Here, we show that LGLL cells from patients possess enhanced cytotoxic characteristics and express elevated levels of activating natural killer receptors as well as their signaling partners, DAP10 and DAP12. Moreover, downstream targets of DAP10 and DAP12 are constitutively activated in LGLL cells, and expression of dominant-negative DAP10 and DAP12 dramatically reduces their lytic capacity. These are the first results to show that activating NKR-ligand interactions play a critical role in initiating the DAP10 and DAP12 signaling events that lead to enhanced lytic potential of LGLL cells. Results shown suggest that inhibitors of DAP10 and DAP12 or other proteins involved in this signaling pathway will be attractive therapeutic targets for the treatment of LGLL and other autoimmune diseases and syndromes.


Adaptor Proteins, Signal Transducing/physiology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/genetics , Leukemia, Large Granular Lymphocytic/immunology , Membrane Proteins/physiology , Receptors, Immunologic/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Endothelial Cells/immunology , Endothelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , K562 Cells , Leukemia, Large Granular Lymphocytic/genetics , Leukemia, Large Granular Lymphocytic/metabolism , Leukemia, Large Granular Lymphocytic/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pulmonary Artery/immunology , Pulmonary Artery/pathology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Natural Killer Cell/metabolism , Signal Transduction/genetics , Tumor Cells, Cultured
18.
Clin Genitourin Cancer ; 6(2): 117-21, 2008 Sep.
Article En | MEDLINE | ID: mdl-18824435

BACKGROUND: Bax-interacting factor (Bif)-1 protein is a member of the endophilin B family that binds to and activates the proapoptotic Bax protein in response to apoptotic signals. Loss of Bif-1 suppresses the intrinsic pathway of apoptosis and promotes tumorigenesis. We examined the expression levels of Bif-1 protein in human prostate cancer. MATERIALS AND METHODS: Thirty-nine archival tissue specimens of human prostate cancer, and a human prostate cancer tissue microarray containing 19 samples of normal prostate, 26 samples of benign prostatic hyperplasias (BPHs), 30 samples of high-grade prostatic intraepithelial neoplasia (PIN), and 153 samples of prostate cancer, were selected for immunohistochemical staining with Bif-1 antibody. The slides were scored by 2 independent observers. RESULTS: Nontissue microarray samples: moderate to strong Bif-1 staining was identified in 38 of 39 prostate cancer samples. In 32 cases, foci of PIN were identified adjacent to prostate cancer samples. Of these, 29 samples (90.6%) showed strong and diffuse Bif-1 staining. Benign prostatic hyperplasias, identified in 27 cases, was weakly Bif-1 positive in 88.9% of cases. Tissue microarray samples: 38.6% (59 of 153) of prostate cancer samples showed moderate to strong Bif-1 expression, and 21.6% (33 of 153) were Bif-1 negative. Bif-1 expression was moderate to strong in 76.7% (23 of 30) of PIN. Bif-1 was weak to moderate in 53.8% (14 of 26) of BPH and negative in 46.2% (12 of 26) of them. Low to moderate Bif-1 was seen in 89.5% of normal prostate samples. CONCLUSION: The loss of Bif-1 expression in a subset of prostate cancer samples is in agreement with the proapoptotic function of Bif-1. The significance of the increased Bif-1 in a subgroup of prostate cancer samples and in PIN remains to be determined. It seems that Bif-1 has a role in prostate cancer, providing the rationale for using Bif-1 as a target for prostate anticancer therapy.


Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/metabolism , Prostatic Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antibody Specificity , Humans , Immunohistochemistry , Male , Middle Aged , Prostatic Hyperplasia/metabolism , Tissue Array Analysis
19.
Blood ; 112(12): 4694-8, 2008 Dec 01.
Article En | MEDLINE | ID: mdl-18791165

Large granular lymphocyte (LGL) leukemia is commonly associated with poor hematopoiesis. The first case of pulmonary artery hypertension (PAH) was observed in a 57-year-old woman with natural killer (NK)-LGL leukemia and transfusion-dependent anemia. Using a genetic approach, we demonstrated that killing of pulmonary endothelial cells by patient NK cells was mediated by dysregulated balance in activating and inhibitory NK-receptor signaling. Elevated pulmonary artery pressure and erythroid differentiation improved after disrupting the NK-receptor signaling pathway with 4 courses of a farnesyltransferase inhibitor, tipifarnib. Coincidental association between PAH and LGL leukemia suggest a causal relationship between the expanded lymphocyte population and these clinical manifestations. This trial is registered at www.ClinicalTrials.gov as NCI 6823.


Antineoplastic Agents/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Leukemia, Large Granular Lymphocytic/drug therapy , Receptors, Natural Killer Cell/genetics , Cells, Cultured , Enzyme Inhibitors/therapeutic use , Female , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , K562 Cells , Leukemia, Large Granular Lymphocytic/complications , Leukemia, Large Granular Lymphocytic/genetics , Middle Aged , Quinolones/therapeutic use , Signal Transduction/genetics , Treatment Outcome
20.
Blood ; 111(2): 954-62, 2008 Jan 15.
Article En | MEDLINE | ID: mdl-17928532

Graft-versus-host disease (GVHD) remains a major cause of morbidity and mortality in allogeneic hematopoietic stem cell transplantation. Migration of donor-derived T cells into GVHD target organs plays an essential role in the development of GVHD. beta2 integrins are critically important for leukocyte extravasation through vascular endothelia and for T-cell activation. We asked whether CD18-deficient T cells would induce less GVHD while sparing the graft-versus-leukemia (GVL) effect. In murine allogeneic bone marrow transplantation models, we found that recipients of CD18-/- donor T cells had significantly less GVHD morbidity and mortality compared with recipients of wild-type (WT) donor T cells. Analysis of alloreactivity showed that CD18-/- and WT T cells had comparable activation, expansion, and cytokine production in vivo. Reduced GVHD was associated with a significant decrease in donor T-cell infiltration of recipient intestine and with an overall decrease in pathologic scores in intestine and liver. Finally, we found that the in vivo GVL effect of CD18-/- donor T cells was largely preserved, because mortality of the recipients who received transplants of CD18-/- T cells plus tumor cells was greatly delayed or prevented. Our data suggest that strategies to target beta2 integrin have clinical potential to alleviate or prevent GVHD while sparing GVL activity.


CD18 Antigens/immunology , Cell Movement/immunology , Graft vs Host Disease/therapy , Graft vs Leukemia Effect/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Bone Marrow Transplantation , CD18 Antigens/genetics , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Humans , Intestines/immunology , Intestines/pathology , Liver/immunology , Liver/pathology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , T-Lymphocytes/pathology , Transplantation, Homologous
...