Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 111(11): 4203-8, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24586048

ABSTRACT

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.


Subject(s)
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
2.
Cancer Immunol Immunother ; 61(4): 523-33, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21983879

ABSTRACT

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.


Subject(s)
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
3.
J Immunol ; 185(10): 6058-67, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20944005

ABSTRACT

NK cells are major effectors of the innate immune response through cytolysis and bridge to the adaptive immune response through cytokine release. The mediators of activation are well studied; however, little is known about the mechanisms that restrain activation. In this report, we demonstrate that the transcriptional repressor PRDM1 (also known as Blimp-1 or PRDI-BF1) is a critical negative regulator of NK function. Three distinct PRDM1 isoforms are selectively induced in the CD56(dim) NK population in response to activation. PRDM1 coordinately suppresses the release of IFN-γ, TNF-α, and TNF-ß through direct binding to multiple conserved regulatory regions. Ablation of PRDM1 expression leads to enhanced production of IFN-γ and TNF-α but does not alter cytotoxicity, whereas overexpression blocks cytokine production. PRDM1 response elements are defined at the IFNG and TNF loci. Collectively, these data demonstrate a key role for PRDM1 in the negative regulation of NK activation and position PRDM1 as a common regulator of the adaptive and innate immune response.


Subject(s)
Cytokines/biosynthesis , Gene Expression Regulation/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Repressor Proteins/metabolism , Animals , Blotting, Western , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Humans , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction
4.
Proc Natl Acad Sci U S A ; 106(31): 12974-9, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19470455

ABSTRACT

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.


Subject(s)
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
5.
Blood ; 113(14): 3226-34, 2009 Apr 02.
Article in English | MEDLINE | ID: mdl-19075187

ABSTRACT

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.


Subject(s)
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
6.
Blood ; 112(12): 4694-8, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18791165

ABSTRACT

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.


Subject(s)
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
7.
Int Immunol ; 21(4): 423-32, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19261694

ABSTRACT

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.


Subject(s)
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
8.
ACS Biomater Sci Eng ; 6(7): 4179-4199, 2020 07 13.
Article in English | MEDLINE | ID: mdl-33463353

ABSTRACT

A critical hurdle associated with natural killer (NK) cell immunotherapies is inadequate infiltration and function in the solid tumor microenvironment. Well-controlled 3D culture systems could advance our understanding of the role of various biophysical and biochemical cues that impact NK cell migration in solid tumors. The objectives of this study were to establish a biomaterial which (i) supports NK cell migration and (ii) recapitulates features of the in vivo solid tumor microenvironment, to study NK infiltration and function in a 3D system. Using peptide-functionalized poly(ethylene glycol)-based hydrogels, the extent of NK-92 cell migration was observed to be largely dependent on the density of integrin binding sites and the presence of matrix metalloproteinase degradable sites. When lung cancer cells were encapsulated into the hydrogels to create tumor microenvironments, the extent of NK-92 cell migration and functional activity was dependent on the cancer cell type and duration of 3D culture. NK-92 cells showed greater migration into the models consisting of nonmetastatic A549 cells relative to metastatic H1299 cells, and reduced migration in both models when cancer cells were cultured for 7 days versus 1 day. In addition, the production of NK cell-related pro-inflammatory cytokines and chemokines was reduced in H1299 models relative to A549 models. These differences in NK-92 cell migration and cytokine/chemokine production corresponded to differences in the production of various immunomodulatory molecules by the different cancer cells, namely, the H1299 models showed increased stress ligand shedding and immunosuppressive cytokine production, particularly TGF-ß. Indeed, inhibition of TGF-ß receptor I in NK-92 cells restored their infiltration in H1299 models to levels similar to that in A549 models and increased overall infiltration in both models. Relative to conventional 2D cocultures, NK-92 cell mediated cytotoxicity was reduced in the 3D tumor models, suggesting the hydrogel serves to mimic some features of the biophysical barriers in in vivo tumor microenvironments. This study demonstrates the feasibility of a synthetic hydrogel system for investigating the biophysical and biochemical cues impacting NK cell infiltration and NK cell-cancer cell interactions in the solid tumor microenvironment.


Subject(s)
Killer Cells, Natural , Tumor Microenvironment , A549 Cells , Hydrogels , Immunotherapy
9.
PLoS One ; 15(2): e0225820, 2020.
Article in English | MEDLINE | ID: mdl-32040476

ABSTRACT

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.


Subject(s)
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
10.
Oncoimmunology ; 8(4): e1557372, 2019.
Article in English | MEDLINE | ID: mdl-30906652

ABSTRACT

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.

11.
Cancer Causes Control ; 19(8): 805-11, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18344007

ABSTRACT

Concerns have been raised regarding the possible adverse health effects of consumption of unpasteurized milk and risk of cancer. We examined the association of self-reported intake of unpasteurized milk with subsequent risk of cancer in a large population-based cohort study. The Iowa Women's Health Study is a prospective cohort study of 55-69 year old women at baseline in 1986. Of the 41,836 women in the cohort at baseline, 22,808 cancer-free women completed the fourth follow-up questionnaire in 1997. Risk ratios (RR) and 95% confidence intervals (CI) were calculated using Cox proportional hazards regression analysis. Reported intake of unpasteurized milk was high: 59.2% consumed only as a child, 2.5% consumed only as an adult, and 16.5% consumed as a child and an adult. A total of 2,379 cancers were identified in the cohort at risk. Overall, the age-adjusted risk of cancer was lower among women who reported consumption of unpasteurized milk only as a child (RR = 0.90, 95% CI: 0.82-0.99) or as a child and an adult (RR = 0.85; 95% CI: 0.75-0.97). Adjustment for confounding factors attenuated these associations (RR = 0.92, 95% CI: 0.83-1.02 for consumption only as a child, and RR = 0.91; 95% CI: 0.79-1.04 for consumption as a child and an adult). These data suggest that consumption of unpasteurized milk does not increase risk of cancer.


Subject(s)
Diet , Milk , Neoplasms/epidemiology , Aged , Animals , Cohort Studies , Female , Humans , Incidence , Male , Middle Aged , Neoplasms/etiology , Prospective Studies , Risk Assessment , Surveys and Questionnaires
12.
Clin Genitourin Cancer ; 6(2): 117-21, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18824435

ABSTRACT

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.


Subject(s)
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
13.
Biol Res Nurs ; 9(3): 205-14, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18077773

ABSTRACT

BACKGROUND: The use of relaxation and guided imagery to reduce stress and improve immune function has great potential benefits for patients with breast cancer. METHODS: This pilot study used a pretest-posttest experimental design with 28 breast cancer patients, aged 25 to 75 years, with the diagnosis of stage 0, 1, or 2 breast cancer. The experimental group received a relaxation and guided imagery intervention and the control group received standard care. The effects of the intervention on immune function were measured by natural killer (NK) cell cytotoxicity and IL-2-activated NK cell activity prior to surgery and 4 weeks postsurgery. NK cell activity was measured using a 15-hr incubation chromium release assay. Cytotoxicity of NK cells was measured against chromium-labeled K-562 target cells. IL-2 was used to enhance reactivity of NK cells against tumor cells. After incubation for 15 hr, cytotoxicity was measured through the release of radioactive chromium. RESULTS: Significant differences between groups were found at 4 weeks postsurgery. T-tests showed increased NK cell cytotoxicity for the intervention group at 100:1, 50:1, and 25:1 effector cell: target cell ratios (E:T) (p < .01 to p < .05) and increased activation for IL-2 at 100:1, 50:1, 25:1, and 12.5:1 (E:T) (p < .01 to p < .05) for the intervention group as compared to the control group. DISCUSSION: These findings suggest that a relaxation intervention such as guided imagery could have an effect on NK cell cytotoxicity and NK cell cytotoxicity after activation with IL-2 in patients undergoing surgery for breast cancer.


Subject(s)
Breast Neoplasms/immunology , Imagery, Psychotherapy , Adult , Aged , Breast Neoplasms/psychology , Breast Neoplasms/therapy , Female , Humans , Interleukin-2/immunology , Killer Cells, Natural/immunology , Middle Aged
14.
Mol Cancer Ther ; 6(11): 2938-47, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18025278

ABSTRACT

One of the major obstacles in curing prostate cancer is the development of drug resistance to docetaxel, which is the gold standard for the treatment of this disease. It is not only imperative to discover the molecular basis of resistance but also to find therapeutic agents that can disrupt the resistant pathways. Based on initial findings that docetaxel-resistant PC3-DR and DU145-DR prostate tumor cell lines express tumor necrosis factor-related apoptosis inducing ligand (TRAIL) receptors, we examined whether TRAIL could be used as an alternative method to kill PC3-DR and DU145-DR cells. However, these tumor cells were found to be TRAIL resistant. Because PC3-DR and DU-145-DR cells were previously shown by us to be clusterin positive, we examined if clusterin could play a role in TRAIL resistance. We found that resveratrol could sensitize docetaxel-resistant tumor cells to TRAIL, and it worked by blocking clusterin expression. In particular, small interfering RNA clusterin expression in the cell lines was sufficient to produce apoptosis by TRAIL. Further analysis indicated that resveratrol functions as an effective tyrosine kinase inhibitor, similar to its analogue, piceatannol, and could inhibit Src and Jak kinases, thus resulting in loss of Stat1 activation. We have shown earlier that Stat1 is essential for gene transcription of clusterin. These results, taken together, show that resveratrol could be a useful new therapeutic agent to combat docetaxel resistance.


Subject(s)
Clusterin/metabolism , Drug Resistance, Neoplasm/drug effects , Prostatic Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Clusterin/genetics , Docetaxel , Down-Regulation/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Janus Kinases/metabolism , Male , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Resveratrol , STAT Transcription Factors/metabolism , Stilbenes/pharmacology , Taxoids/pharmacology
15.
Front Immunol ; 9: 2887, 2018.
Article in English | MEDLINE | ID: mdl-30574153

ABSTRACT

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.


Subject(s)
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
16.
Mol Cell Biol ; 24(12): 5565-76, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15169915

ABSTRACT

Although recent evidence supports a tumor-suppressive role for the GTPase RhoB, little is known about its regulation by signal transduction pathways. Here we demonstrate that Ras downregulates RhoB expression by a phosphatidylinositol 3-kinase (PI3K)- and Akt- but not Mek-dependent mechanism. Furthermore, genetic and pharmacological blockade of PI3K/Akt results in upregulation of RhoB expression. We also provide evidence for the importance of the downregulation of RhoB in oncogenesis by demonstrating that RhoB antagonizes Ras/PI3K/Akt malignancy. Ectopic expression of RhoB, but not the close relative RhoA, inhibits Ras, PI3K, and Akt induction of transformation, migration, and invasion and induces apoptosis and anoikis. Finally, RhoB inhibits melanoma metastasis to the lung in a mouse model. These studies identify suppression of RhoB as a mechanism by which the Ras/PI3K/Akt pathway induces tumor survival, transformation, invasion, and metastasis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , ras Proteins/metabolism , rhoB GTP-Binding Protein/genetics , rhoB GTP-Binding Protein/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Down-Regulation , Female , Fluorouracil/pharmacology , Genes, ras , MAP Kinase Signaling System/drug effects , Melanoma, Experimental/genetics , Melanoma, Experimental/prevention & control , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins c-akt , Transcription, Genetic
17.
Viruses ; 9(9)2017 08 23.
Article in English | MEDLINE | ID: mdl-28832539

ABSTRACT

After decades of research, oncolytic virotherapy has recently advanced to clinical application, and currently a multitude of novel agents and combination treatments are being evaluated for cancer therapy. Oncolytic agents preferentially replicate in tumor cells, inducing tumor cell lysis and complex antitumor effects, such as innate and adaptive immune responses and the destruction of tumor vasculature. With the availability of different vector platforms and the potential of both genetic engineering and combination regimens to enhance particular aspects of safety and efficacy, the identification of optimal treatments for patient subpopulations or even individual patients becomes a top priority. Mathematical modeling can provide support in this arena by making use of experimental and clinical data to generate hypotheses about the mechanisms underlying complex biology and, ultimately, predict optimal treatment protocols. Increasingly complex models can be applied to account for therapeutically relevant parameters such as components of the immune system. In this review, we describe current developments in oncolytic virotherapy and mathematical modeling to discuss the benefit of integrating different modeling approaches into biological and clinical experimentation. Conclusively, we propose a mutual combination of these research fields to increase the value of the preclinical development and the therapeutic efficacy of the resulting treatments.


Subject(s)
Models, Theoretical , Neoplasms/therapy , Oncolytic Virotherapy/methods , Cell Death/immunology , Combined Modality Therapy , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology , Humans , Immune System , Immunotherapy/methods , Life Cycle Stages , Neoplasms/immunology , Neoplasms/virology , Oncolytic Viruses/immunology , Virus Replication , Viruses/growth & development , Viruses/immunology , Viruses/pathogenicity
18.
Cancer Res ; 63(23): 8523-30, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14679020

ABSTRACT

T cells expressing human leukemia virus (HTLV) type 1, the etiological agent of adult T-cell leukemia, are remarkably resistant to conventional chemotherapy, and the need for drugs that effectively kill these cells is apparent. Here we show that roscovitine, an inhibitor of cyclin-dependent kinases (CDKs), induces the apoptosis of the HTLV-1-transformed T-cell line MT-2. Roscovitine prevented the tyrosine phosphorylation and consequent activation of the transcription factor signal transducer and activator of transcription (STAT) 5 when presented to MT-2 cells in the presence or absence of a caspase-3 inhibitor, and ectopic expression of a dominant-negative form of STAT5 in MT-2 cells induced apoptosis. Roscovitine and dominant-negative STAT5 also reduced the expression of the antiapoptotic protein XIAP, and STAT5 was associated with the XIAP promoter in vivo. Antibody to platelet-derived growth factor (PDGF) alpha receptors coprecipitated STAT5 from extracts of untreated but not roscovitine-treated cells. The tyrosine phosphatase inhibitor sodium orthovanadate ablated the inhibitory effects of roscovitine on STAT5/PDGF alpha receptor interaction, STAT5 activity, and cell survival. We suggest that roscovitine reduces the abundance of tyrosine-phosphorylated PDGF alpha receptors; as a result, STAT5 does not become active, and STAT5 gene products required for cell survival are not expressed.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Human T-lymphotropic virus 1/physiology , Milk Proteins , Purines/pharmacology , T-Lymphocytes/drug effects , Trans-Activators/antagonists & inhibitors , Apoptosis/physiology , Cell Line, Transformed , DNA-Binding Proteins/metabolism , Humans , Promoter Regions, Genetic , Protein Biosynthesis , Proteins/antagonists & inhibitors , Proteins/genetics , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Roscovitine , STAT5 Transcription Factor , T-Lymphocytes/cytology , T-Lymphocytes/virology , Trans-Activators/metabolism , X-Linked Inhibitor of Apoptosis Protein
19.
Cancer Res ; 76(5): 1009-18, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26833128

ABSTRACT

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.


Subject(s)
Cell Movement , Lymphocyte Activation , Neoplasms/radiotherapy , T-Lymphocytes/immunology , Humans , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/physiology
20.
Sci Rep ; 6: 29521, 2016 07 13.
Article in English | MEDLINE | ID: mdl-27405665

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL