Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters

Publication year range
1.
J Immunol ; 209(6): 1212-1223, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35995507

ABSTRACT

Chronic lymphocytic leukemia (CLL) is the most common adult leukemia, but, despite advances in treatment, many patients still experience relapse. CLL cells depend on interactions with supportive cells, and nurse-like cells (NLCs) are the major such cell type. However, little is known about how NLCs develop. Here, we performed DNA methylation analysis of CLL patient-derived NLCs using the 850K Illumina array, comparing CD14+ cells at day 1 (monocytes) versus day 14 (NLCs). We found a strong loss of methylation in AP-1 transcription factor binding sites, which may be driven by MAPK signaling. Testing of individual MAPK pathways (MEK, p38, and JNK) revealed a strong dependence on MEK/ERK for NLC development, because treatment of patient samples with the MEK inhibitor trametinib dramatically reduced NLC development in vitro. Using the adoptive transfer Eµ-TCL1 mouse model of CLL, we found that MEK inhibition slowed CLL progression, leading to lower WBC counts and to significantly longer survival time. There were also lower numbers of mouse macrophages, particularly within the M2-like population. In summary, NLC development depends on MEK signaling, and inhibition of MEK leads to increased survival time in vivo. Hence, targeting the MEK/ERK pathway may be an effective treatment strategy for CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Animals , Cell Differentiation , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Monocytes/metabolism , Transcription Factor AP-1/metabolism
2.
Int J Mol Sci ; 24(8)2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37108786

ABSTRACT

Overactivation of immune responses is a hallmark of autoimmune disease pathogenesis. This includes the heightened production of inflammatory cytokines such as Tumor Necrosis Factor α (TNFα), and the secretion of autoantibodies such as isotypes of rheumatoid factor (RF) and anticitrullinated protein antibody (ACPA). Fcγ receptors (FcγR) expressed on the surface of myeloid cells bind Immunoglobulin G (IgG) immune complexes. Recognition of autoantigen-antibody complexes by FcγR induces an inflammatory phenotype that results in tissue damage and further escalation of the inflammatory response. Bromodomain and extra-terminal protein (BET) inhibition is associated with reduced immune responses, making the BET family a potential therapeutic target for autoimmune diseases such as rheumatoid arthritis (RA). In this paper, we examined the BET inhibitor PLX51107 and its effect on regulating FcγR expression and function in RA. PLX51107 significantly downregulated expression of FcγRIIa, FcγRIIb, FcγRIIIa, and the common γ-chain, FcϵR1-γ, in both healthy donor and RA patient monocytes. Consistent with this, PLX51107 treatment attenuated signaling events downstream of FcγR activation. This was accompanied by a significant decrease in phagocytosis and TNFα production. Finally, in a collagen-induced arthritis model, PLX51107-treatment reduced FcγR expression in vivo accompanied by a significant reduction in footpad swelling. These results suggest that BET inhibition is a novel therapeutic approach that requires further exploration as a treatment for patients with RA.


Subject(s)
Arthritis, Rheumatoid , Receptors, IgG , Humans , Arthritis, Rheumatoid/metabolism , Inflammation/metabolism , Monocytes/metabolism , Receptors, IgG/metabolism , Tumor Necrosis Factor-alpha/metabolism , Nerve Tissue Proteins/metabolism
3.
J Immunol ; 204(7): 1988-1997, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32094205

ABSTRACT

TLRs, a family of membrane-bound pattern recognition receptors found on innate immune cells, have been well studied in the context of cancer therapy. Activation of these receptors has been shown to induce inflammatory anticancer events, including differentiation and apoptosis, across a wide variety of malignancies. In contrast, intracellular pattern recognition receptors such as NOD-like receptors have been minimally studied. NOD2 is a member of the NOD-like receptor family that initiates inflammatory signaling in response to the bacterial motif muramyl dipeptide. In this study, we examined the influence of NOD2 in human acute myeloid leukemia (AML) cells, demonstrating that IFN-γ treatment upregulated the expression of NOD2 signaling pathway members SLC15A3 and SLC15A4, downstream signaling kinase RIPK2, and the NOD2 receptor itself. This priming allowed for effective induction of caspase-1-dependent cell death upon treatment with muramyl tripeptide phosphatidylethanolamine (MTP-PE), a synthetic ligand for NOD2. Furthermore, the combination of MTP-PE and IFN-γ on AML blasts generated an inflammatory cytokine profile and activated NK cells. In a murine model of AML, dual treatment with MTP-PE and IFN-γ led to a significant increase in mature CD27- CD11b+ NK cells as well as a significant reduction in disease burden and extended survival. These results suggest that NOD2 activation, primed by IFN-γ, may provide a novel therapeutic option for AML.


Subject(s)
Apoptosis/physiology , Leukemia, Myeloid, Acute/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptors, Pattern Recognition/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Interferon-gamma/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
4.
J Immunol ; 203(12): 3216-3224, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31732534

ABSTRACT

Monocytes and macrophages express FcγR that engage IgG immune complexes such as Ab-opsonized pathogens or cancer cells to destroy them by various mechanisms, including phagocytosis. FcγR-mediated phagocytosis is regulated by the concerted actions of activating FcγR and inhibitory receptors, such as FcγRIIb and SIRPα. In this study, we report that another ITIM-containing receptor, PECAM1/CD31, regulates FcγR function and is itself regulated by FcγR activation. First, quantitative RT-PCR and flow cytometry analyses revealed that human monocyte FcγR activation leads to a significant downregulation of CD31 expression, both at the message level and at surface expression, mainly mediated through FcγRIIa. Interestingly, the kinetics of downregulation between the two varied, with surface expression reducing earlier than the message. Experiments to analyze the mechanism behind this discrepancy revealed that the loss of surface expression was because of internalization, which depended predominantly on the PI3 kinase pathway and was independent of FcγR internalization. Finally, functional analyses showed that the downregulation of CD31 expression in monocytes by small interfering RNA enhanced FcγR-mediated phagocytic ability but have little effect on cytokine production. Together, these results suggest that CD31 acts as a checkpoint receptor that could be targeted to enhance FcγR functions in Ab-mediated therapies.


Subject(s)
Monocytes/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, IgG/metabolism , Antigen-Antibody Complex/immunology , Blood Donors , Cytokines/metabolism , Down-Regulation , Gene Knockdown Techniques , Humans , Immunoglobulin G/metabolism , Phagocytosis/genetics , Phosphatidylinositol 3-Kinases/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics , RNA, Small Interfering/genetics , Signal Transduction/immunology
5.
Proc Natl Acad Sci U S A ; 114(36): 9629-9634, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827320

ABSTRACT

Activating mutations in BRAF are found in 50% of melanomas and although treatment with BRAF inhibitors (BRAFi) is effective, resistance often develops. We now show that recently discovered NRAS isoform 2 is up-regulated in the setting of BRAF inhibitor resistance in melanoma, in both cell lines and patient tumor tissues. When isoform 2 was overexpressed in BRAF mutant melanoma cell lines, melanoma cell proliferation and in vivo tumor growth were significantly increased in the presence of BRAFi treatment. shRNA-mediated knockdown of isoform 2 in BRAFi resistant cells restored sensitivity to BRAFi compared with controls. Signaling analysis indicated decreased mitogen-activated protein kinase (MAPK) pathway signaling and increased phosphoinositol-3-kinase (PI3K) pathway signaling in isoform 2 overexpressing cells compared with isoform 1 overexpressing cells. Immunoprecipitation of isoform 2 validated a binding affinity of this isoform to both PI3K and BRAF/RAF1. The addition of an AKT inhibitor to BRAFi treatment resulted in a partial restoration of BRAFi sensitivity in cells expressing high levels of isoform 2. NRAS isoform 2 may contribute to resistance to BRAFi by facilitating PI3K pathway activation.


Subject(s)
GTP Phosphohydrolases/genetics , Melanoma/drug therapy , Melanoma/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Drug Resistance, Neoplasm/genetics , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/metabolism , Gene Knockdown Techniques , Humans , Indoles/therapeutic use , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Melanoma/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Skin Neoplasms/metabolism , Sulfonamides/therapeutic use , Up-Regulation , Vemurafenib
6.
Int Immunol ; 30(8): 375-383, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29868798

ABSTRACT

Acute myeloid leukemia (AML) remains a significant health problem, with poor outcomes despite chemotherapy and bone marrow transplants. Although one form of AML, acute promyelocytic leukemia (APL), is successfully treated with all-trans retinoic acid (ATRA), this drug is seemingly ineffective against all other forms of AML. Here, we show that ATRA up-regulates CD38 expression on AML blasts to sufficient levels that promote antibody-mediated fratricide following the addition of anti-CD38 daratumumab (DARA). The combination of ATRA plus DARA induced Fc-dependent conjugate formation and cytotoxicity among AML blasts in vitro. Combination treatment also led to reduction in tumor volume and resulted in increased overall survival in murine engraftment models of AML. These results suggest that, although ATRA does not induce differentiation of non-APL, it may be effective as a therapy in conjunction with DARA.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Tretinoin/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Drug Therapy, Combination , Humans , Leukemia, Myeloid, Acute/pathology , Tretinoin/chemistry , Tretinoin/therapeutic use , Tumor Cells, Cultured
7.
J Biol Chem ; 291(8): 3895-904, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26694610

ABSTRACT

Monocytes and macrophages are critical for the effectiveness of monoclonal antibody therapy. Responses to antibody-coated tumor cells are largely mediated by Fcγ receptors (FcγRs), which become activated upon binding to immune complexes. FcγRIIb is an inhibitory FcγR that negatively regulates these responses, and it is expressed on monocytes and macrophages. Therefore, deletion or down-regulation of this receptor may substantially enhance therapeutic outcomes. Here we screened a panel of Toll-like receptor (TLR) agonists and found that those selective for TLR4 and TLR8 could significantly down-regulate the expression of FcγRIIb. Upon further examination, we found that treatment of monocytes with TLR4 agonists could lead to the ubiquitination of FcγRIIb protein. A search of our earlier microarray database of monocytes activated with the TLR7/8 agonist R-848 (in which FcγRIIb was down-regulated) revealed an up-regulation of membrane-associated ring finger (C3HC4) 3 (MARCH3), an E3 ubiquitin ligase. Therefore, we tested whether LPS treatment could up-regulate MARCH3 in monocytes and whether this E3 ligase was involved with LPS-mediated FcγRIIb down-regulation. The results showed that LPS activation of TLR4 significantly increased MARCH3 expression and that siRNA against MARCH3 prevented the decrease in FcγRIIb following LPS treatment. These data suggest that activation of TLR4 on monocytes can induce a rapid down-regulation of FcγRIIb protein and that this involves ubiquitination.


Subject(s)
Carrier Proteins/metabolism , Down-Regulation/drug effects , Lipopolysaccharides/pharmacology , Membrane Proteins/metabolism , Monocytes/metabolism , Receptors, IgG/biosynthesis , Toll-Like Receptor 4/agonists , Ubiquitination/drug effects , Down-Regulation/physiology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Ubiquitin-Protein Ligases , Ubiquitination/physiology
8.
J Biol Chem ; 291(27): 14356-14362, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27226587

ABSTRACT

Nurse-like cells (NLCs) play a central role in chronic lymphocytic leukemia (CLL) because they promote the survival and proliferation of CLL cells. NLCs are derived from the monocyte lineage and are driven toward their phenotype via contact-dependent and -independent signals from CLL cells. Because of the central role of NLCs in promoting disease, new strategies to eliminate or reprogram them are needed. Successful reprogramming may be of extra benefit because NLCs express Fcγ receptors (FcγRs) and thus could act as effector cells within the context of antibody therapy. IFNγ is known to promote the polarization of macrophages toward an M1-like state that is no longer tumor-supportive. In an effort to reprogram the phenotype of NLCs, we found that IFNγ up-regulated the M1-related markers CD86 and HLA-DR as well as FcγRIa. This corresponded to enhanced FcγR-mediated cytokine production as well as rituximab-mediated phagocytosis of CLL cells. In addition, IFNγ down-regulated the expression of CD31, resulting in withdrawal of the survival advantage on CLL cells. These results suggest that IFNγ can re-educate NLCs and shift them toward an effector-like state and that therapies promoting local IFNγ production may be effective adjuvants for antibody therapy in CLL.


Subject(s)
Cell Survival , Interferon-gamma/administration & dosage , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , B7-2 Antigen/metabolism , Cells, Cultured , HLA-DR Antigens/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Phagocytosis , Receptors, IgG/metabolism
9.
J Biol Chem ; 291(49): 25656-25666, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27780867

ABSTRACT

Acute myeloid leukemia (AML) is characterized by the proliferation of immature myeloid lineage blasts. Due to its heterogeneity and to the high rate of acquired drug resistance and relapse, new treatment strategies are needed. Here, we demonstrate that IFNγ promotes AML blasts to act as effector cells within the context of antibody therapy. Treatment with IFNγ drove AML blasts toward a more differentiated state, wherein they showed increased expression of the M1-related markers HLA-DR and CD86, as well as of FcγRI, which mediates effector responses to therapeutic antibodies. Importantly, IFNγ was able to up-regulate CD38, the target of the therapeutic antibody daratumumab. Because the antigen (CD38) and effector receptor (FcγRI) were both simultaneously up-regulated on the AML blasts, we tested whether IFNγ treatment of the AML cell lines THP-1 and MV4-11 could stimulate them to target one another after the addition of daratumumab. Results showed that IFNγ significantly increased daratumumab-mediated cytotoxicity, as measured both by 51Cr release and lactate dehydrogenase release assays. We also found that the combination of IFNγ and activation of FcγR led to the release of granzyme B by AML cells. Finally, using a murine NSG model of subcutaneous AML, we found that treatment with IFNγ plus daratumumab significantly attenuated tumor growth. Taken together, these studies show a novel mechanism of daratumumab-mediated killing and a possible new therapeutic strategy for AML.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cytotoxins/pharmacology , Interferon-gamma/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Animals , Cell Line, Tumor , Female , Granzymes/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/metabolism , Receptors, IgG/metabolism , Xenograft Model Antitumor Assays
10.
J Biol Chem ; 291(6): 3043-52, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26627823

ABSTRACT

The irreversible Bruton's tyrosine kinase (Btk) inhibitor ibrutinib has shown efficacy against B-cell tumors such as chronic lymphocytic leukemia and B-cell non-Hodgkin lymphoma. Fcγ receptors (FcγR) on immune cells such as macrophages play an important role in tumor-specific antibody-mediated immune responses, but many such responses involve Btk. Here we tested the effects of ibrutinib on FcγR-mediated activities in monocytes. We found that ibrutinib did not affect monocyte FcγR-mediated phagocytosis, even at concentrations higher than those achieved physiologically, but suppressed FcγR-mediated cytokine production. We confirmed these findings in macrophages from Xid mice in which Btk signaling is defective. Because calcium flux is a major event downstream of Btk, we tested whether it was involved in phagocytosis. The results showed that blocking intracellular calcium flux decreased FcγR-mediated cytokine production but not phagocytosis. To verify this, we measured activation of the GTPase Rac, which is responsible for actin polymerization. Results showed that ibrutinib did not inhibit Rac activation, nor did the calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester). We next asked whether the effect of ibrutinib on monocyte FcγR-mediated cytokine production could be rescued by IFNγ priming because NK cells produce IFNγ in response to antibody therapy. Pretreatment of monocytes with IFNγ abrogated the effects of ibrutinib on FcγR-mediated cytokine production, suggesting that IFNγ priming could overcome this Btk inhibition. Furthermore, in monocyte-natural killer cell co-cultures, ibrutinib did not inhibit FcγR-mediated cytokine production despite doing so in single cultures. These results suggest that combining ibrutinib with monoclonal antibody therapy could enhance chronic lymphocytic leukemia cell killing without affecting macrophage effector function.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Macrophages/metabolism , Monocytes/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, IgG/metabolism , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Animals , Calcium Signaling/drug effects , Calcium Signaling/genetics , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Macrophages/pathology , Mice , Monocytes/pathology , Piperidines , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptors, IgG/genetics
11.
J Immunol ; 194(6): 2786-95, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25667415

ABSTRACT

FcγRs are critical mediators of mAb cancer therapies, because they drive cytotoxic processes upon binding of effector cells to opsonized targets. Along with NK cells, monocytes are also known to destroy Ab-coated targets via Ab-dependent cellular cytotoxicity (ADCC). However, the precise mechanisms by which monocytes carry out this function have remained elusive. In this article, we show that human monocytes produce the protease granzyme B upon both FcγR and TLR8 activation. Treatment with TLR8 agonists elicited granzyme B and also enhanced FcγR-mediated granzyme B production in an additive fashion. Furthermore, monocyte-mediated ADCC against cetuximab-coated tumor targets was enhanced by TLR8 agonist treatment, and this enhancement of ADCC required granzyme B. Hence we have identified granzyme B as an important mediator of FcγR function in human monocytes and have uncovered another mechanism by which TLR8 agonists may enhance FcγR-based therapies.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Granzymes/metabolism , Monocytes/metabolism , Toll-Like Receptor 8/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Angiotensinogen/genetics , Angiotensinogen/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Cells, Cultured , Cetuximab , Cluster Analysis , Dose-Response Relationship, Drug , Granzymes/antagonists & inhibitors , Granzymes/genetics , Humans , Imidazoles/pharmacology , Interleukin-2/genetics , Interleukin-2/metabolism , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , Perforin/genetics , Perforin/metabolism , Quinolines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Thiazoles/pharmacology , Time Factors , Toll-Like Receptor 8/agonists , Transcriptome/drug effects
12.
J Immunol ; 195(5): 1995-2005, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26238487

ABSTRACT

Sorafenib is an oral multikinase inhibitor that was originally developed as a Raf kinase inhibitor. We hypothesized that sorafenib would also have inhibitory effects on cytokine signaling pathways in immune cells. PBMCs from normal donors were treated with varying concentrations of sorafenib and stimulated with IFN-α or IL-2. Phosphorylation of STAT1 and STAT5 was measured by flow cytometry and confirmed by immunoblot analysis. Changes in IFN-α- and IL-2-stimulated gene expression were measured by quantitative PCR, and changes in cytokine production were evaluated by ELISA. Cryopreserved PBMCs were obtained from cancer patients before and after receiving 400 mg sorafenib twice daily. Patient PBMCs were thawed, stimulated with IL-2 or IFN-α, and evaluated for phosphorylation of STAT1 and STAT5. Pretreatment of PBMCs with 10 µM sorafenib decreased STAT1 and STAT5 phosphorylation after treatment with IFN-α or IL-2. This inhibitory effect was observed in PBMCs from healthy donors over a range of concentrations of sorafenib (5-20 µM), IL-2 (2-24 nM), and IFN-α (10(1)-10(6) U/ml). This effect was observed in immune cell subsets, including T cells, B cells, NK cells, regulatory T cells, and myeloid-derived suppressor cells. Pretreatment with sorafenib also inhibited PBMC expression of IFN-α- and IL-2-regulated genes and inhibited NK cell production of IFN-γ, RANTES, MIP1-α, and MIG in response to IFN-α stimulation. PBMCs from patients receiving sorafenib therapy showed decreased responsiveness to IL-2 and IFN-α treatment. Sorafenib is a Raf kinase inhibitor that could have off-target effects on cytokine-induced signal transduction in immune effector cells.


Subject(s)
Janus Kinase 1/metabolism , Leukocytes, Mononuclear/drug effects , Protein Kinase Inhibitors/pharmacology , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression/drug effects , Humans , Immunoblotting , Interferon-alpha/pharmacology , Interleukin-2/pharmacology , K562 Cells , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice, Inbred BALB C , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sorafenib , Thyroid Neoplasms/blood , Thyroid Neoplasms/drug therapy , raf Kinases/antagonists & inhibitors , raf Kinases/metabolism
13.
Nanomedicine ; 13(3): 909-920, 2017 04.
Article in English | MEDLINE | ID: mdl-27993723

ABSTRACT

Fluorescent nanodiamonds (FNDs) are nontoxic, infinitely photostable, and emit fluorescence in the near infrared region. Natural killer (NK) cells and monocytes are part of the innate immune system and are crucial to the control of carcinogenesis. FND-mediated stimulation of these cells may serve as a strategy to enhance anti-tumor activity. FNDs were fabricated with a diameter of 70±28 nm. Innate immune cell FND uptake, viability, surface marker expression, and cytokine production were evaluated in vitro. Evaluation of fluorescence emission from the FNDs was conducted in an animal model. In vitro results demonstrated that treatment of immune cells with FNDs resulted in significant dose-dependent FND uptake, no compromise in cell viability, and immune cell activation. FNDs were visualized in an animal model. Hence, FNDs may serve as novel agents with "track and trace" capabilities to stimulate innate immune cell anti-tumor responses, especially as FNDs are amenable to surface-conjugation with immunomodulatory molecules.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Fluorescent Dyes/therapeutic use , Immunity, Cellular/drug effects , Nanodiamonds/therapeutic use , Adjuvants, Immunologic/pharmacokinetics , Animals , Cells, Cultured , Fluorescent Dyes/pharmacokinetics , Humans , Immunity, Innate/drug effects , Immunotherapy , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Monocytes/immunology , Nanodiamonds/analysis , Neoplasms/immunology , Neoplasms/therapy , RAW 264.7 Cells
14.
Kidney Int ; 90(3): 568-79, 2016 09.
Article in English | MEDLINE | ID: mdl-27401534

ABSTRACT

Diabetes mellitus is a systemic disease associated with a deficiency of insulin production or action. Diabetic patients have an increased susceptibility to infection with the urinary tract being the most common site. Recent studies suggest that Ribonuclease 7 (RNase 7) is a potent antimicrobial peptide that plays an important role in protecting the urinary tract from bacterial insult. Because the impact of diabetes on RNase 7 expression and function are unknown, we investigated the effects of insulin on RNase 7 using human urine specimens. The urinary RNase 7 concentrations were measured in healthy control patients and insulin-deficient type 1 diabetics before and after starting insulin therapy. Compared with controls, diabetic patients had suppressed urinary RNase 7 concentrations, which increased with insulin. Using primary human urothelial cells, the mechanisms by which insulin stimulates RNase 7 synthesis were next explored. Insulin induced RNase 7 production via the phosphatidylinositide 3-kinase signaling pathway (PI3K/AKT) to shield urothelial cells from uropathogenic E. coli. In contrast, uropathogenic E. coli suppressed PI3K/AKT activity and RNase 7 production. Thus, insulin and PI3K/AKT signaling are essential for RNase 7 expression and increased infection risks in diabetic patients may be secondary to suppressed RNase 7 production. Our data may provide unique insight into novel urinary tract infection therapeutic strategies in at-risk populations.


Subject(s)
Diabetes Mellitus, Type 1/complications , Escherichia coli Infections/metabolism , Insulin/metabolism , Ribonucleases/metabolism , Urinary Tract Infections/metabolism , Urinary Tract/metabolism , Adolescent , Antigens, CD/metabolism , Cell Line, Tumor , Child , Child, Preschool , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/urine , Escherichia coli/isolation & purification , Escherichia coli Infections/etiology , Escherichia coli Infections/microbiology , Escherichia coli Infections/urine , Female , Humans , Insulin/therapeutic use , Male , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Ribonucleases/urine , Signal Transduction , Urinary Tract/microbiology , Urinary Tract Infections/etiology , Urinary Tract Infections/microbiology , Urinary Tract Infections/urine
15.
Cancer Immunol Immunother ; 65(11): 1353-1364, 2016 11.
Article in English | MEDLINE | ID: mdl-27581603

ABSTRACT

Prime-boost vaccination with recombinant (r) vaccinia(V)-CEA(6D)-TRICOM (triad of co-stimulatory molecules B7.1, ICAM-1 and LFA-3) and rFowlpox(F)-CEA(6D)-TRICOM infect antigen-presenting cells and direct expression of co-stimulatory molecules. We hypothesized that co-administration of vaccine with GM-CSF and interferon alpha (IFN-α) would have efficacy in CEA-expressing cancers. Patients with CEA-expressing cancers received the rV-CEA(6D)-TRICOM vaccine subcutaneously (s.c.) on day 1 followed by GM-CSF s.c. to the injection site on days 1-4. In Cycle 1, patients received thrice weekly s.c. injections of IFN-α-2b the week after rV-CEA(6D)-TRICOM. In Cycles 2-4, patients received thrice weekly s.c. injections of IFN-α-2b the same week that rF-CEA(6D)-TRICOM was given. The first cohort received no IFN followed by dose escalation of IFN-α in subsequent cohorts. Thirty-three patients were accrued (mean 59.8 years). Grade 3 toxicities included fatigue and hyperglycemia. Grade 4-5 adverse events (unrelated to treatment) were confusion (1), elevated aspartate transaminase (AST)/alanine transaminase (ALT) (1), and sudden death (1). No patients had a partial response, and eight patients exhibited stable disease of ≥3 months. Median progression-free survival and overall survival (OS) were 1.8 and 6.3 months, respectively. Significantly higher serum CD27 levels were observed after vaccine therapy (p = 0.006 post 1-2 cycles, p = 0.003 post 3 cycles, p = 0.03 post 4-7 cycles) and 42 % of patients assayed developed CEA-specific T cell responses. Pre-treatment levels of myeloid-derived suppressor cells correlated with overall survival (p = 0.04). Administration of IFN-α led to significantly increased OS (p = 0.02) compared to vaccine alone. While the vaccine regimen produced no clinical responses, IFN-α administration was associated with improved survival.


Subject(s)
Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Interferon-alpha/administration & dosage , T-Lymphocytes/immunology , B7-H1 Antigen/genetics , CD58 Antigens/genetics , Carcinoembryonic Antigen/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Female , Humans , Hyperglycemia/etiology , Intercellular Adhesion Molecule-1/genetics , Male , Middle Aged , Survival Analysis , Treatment Outcome , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Vaccination , Vaccines, Synthetic , Vaccinia virus/genetics
16.
Blood ; 121(6): 984-95, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23144169

ABSTRACT

Microvesicles are small membrane-bound particles comprised of exosomes and various-sized extracellular vesicles. These are released by several cell types. Microvesicles have a variety of cellular functions from communication to mediating growth and differentiation. Microvesicles contain proteins and nucleic acids. Previously, we showed that plasma microvesicles contain microRNAs (miRNAs). Based on our previous report, the majority of peripheral blood microvesicles are derived from platelets, while mononuclear phagocytes, including macrophages, are the second most abundant population. Here, we characterized macrophage-derived microvesicles and explored their role in the differentiation of naive monocytes. We also identified the miRNA content of the macrophage-derived microvesicles. We found that RNA molecules contained in the macrophage-derived microvesicles were transported to target cells, including mono cytes, endothelial cells, epithelial cells, and fibroblasts. Furthermore, we found that miR-223 was transported to target cells and was functionally active. Based on our observations, we hypothesize that microvesicles bind to and activate target cells. Furthermore, we find that microvesicles induce the differentiation of macrophages. Thus, defining key components of this response may identify novel targets to regulate host defense and inflammation.


Subject(s)
Cell Differentiation , Cell-Derived Microparticles/metabolism , Exosomes/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Cell Communication , Cell Line , Cell Line, Tumor , Cells, Cultured , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/cytology , Macrophages/ultrastructure , MicroRNAs/genetics , Microscopy, Confocal , Microscopy, Electron, Transmission , Monocytes/cytology , Monocytes/metabolism , Monocytes/ultrastructure , Oligonucleotide Array Sequence Analysis , RNA Transport/drug effects
17.
J Immunol ; 190(6): 2702-11, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23418626

ABSTRACT

CD20 is a widely validated, B cell-specific target for therapy in B cell malignancies. Rituximab is an anti-CD20 Ab that prolongs survival of chronic lymphocytic leukemia (CLL) patients when combined with chemotherapy. Ofatumumab and GA101 (obinutuzumab) are CD20-directed Abs currently being developed as alternative agents to rituximab in CLL based upon different properties of enhanced direct cell death, NK cell-mediated Ab-dependent cellular cytotoxicity, or complement-dependent cytotoxicity. Despite widespread study, ofatumumab and GA101 have not been compared with each other, nor studied for their interactions with monocytes and macrophages which are critical for the efficacy of anti-CD20 Abs in murine models. In CLL cells, we show that direct cell death and complement-dependent cytotoxicity are greatest with GA101 and ofatumumab, respectively. GA101 promotes enhanced NK cell activation and Ab-dependent cellular cytotoxicity at high Ab concentrations. Ofatumumab elicits superior Ab-dependent cellular phagocytosis with monocyte-derived macrophages. GA101 demonstrated reduced activation of monocytes with diminished pERK, TNF-α release, and FcγRIIa recruitment to lipid rafts. These data demonstrate that GA101 and ofatumumab are both superior to rituximab against CLL cells via different mechanisms of potential tumor elimination. These findings bear relevance to potential combination strategies with each of these anti-CD20 Abs in the treatment of CLL.


Subject(s)
Antibodies, Neoplasm/toxicity , Antigens, CD20/immunology , Drug Delivery Systems/methods , Killer Cells, Natural/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Macrophages/immunology , Monocytes/immunology , Antibodies, Neoplasm/therapeutic use , Antigens, CD20/metabolism , Cell Line, Tumor , Cytotoxicity Tests, Immunologic , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Macrophages/metabolism , Macrophages/pathology , Monocytes/metabolism , Monocytes/pathology , Tumor Cells, Cultured
18.
Mol Ther ; 22(9): 1678-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24895995

ABSTRACT

Glioblastoma is a devastating disease, and there is an urgent need to develop novel therapies, such as oncolytic HSV1 (OV) to effectively target tumor cells. OV therapy depends on tumor-specific replication leading to destruction of neoplastic tissues. Host responses that curtail virus replication limit its efficacy in vivo. We have previously shown that cysteine-rich 61 protein (CCN1) activates a type 1 IFN antiviral defense response in glioblastoma cells. Incorporating TCGA data, we found CCN1 expression to be a negative prognostic factor for glioblastoma patients. Based on this, we used neutralizing antibodies against CCN1 to investigate its effect on OV therapy. Use of an anti-CCN1 antibody in mice bearing glioblastomas treated with OV led to enhanced virus expression along with reduced immune cell infiltration. OV-induced CCN1 increases macrophage migration toward infected glioblastoma cells by directly binding macrophages and also by enhancing the proinflammatory activation of macrophages inducing MCP-1 expression in glioblastoma cells. Activation of macrophages by CCN1 also increases viral clearance. Neutralization of integrin αMß2 reversed CCN1-induced macrophage activation and migration, and reduced MCP-1 expression by glioblastoma cells. Our findings reveal that CCN1 plays a novel role in pathogen clearance; increasing macrophage infiltration and activation resulting in increased virus clearance in tumors.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Glioblastoma/immunology , Herpesvirus 1, Human/genetics , Macrophages/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Chemokine CCL2/metabolism , Female , Genetic Vectors/administration & dosage , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Macrophage Activation , Mice , Neoplasm Transplantation , Oncolytic Viruses/genetics
19.
J Biol Chem ; 288(17): 12345-52, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23504312

ABSTRACT

Fcγ receptor (FcγR) clustering on monocytes/macrophages results in phagocytosis and inflammatory cytokine production, which serve to eliminate antibody-opsonized targets and activate neighboring immune cells. Toll-like receptor 2 (TLR2), which recognizes a range of both bacterial and fungal components, elicits strong proinflammatory responses in these cells when stimulated by ligands, either natural or synthetic. Thus, we explored the possibility that TLR2 agonists could strengthen FcγR activity within the context of antibody therapy. Human peripheral blood monocytes treated with the TLR2 agonist Pam2CSK4 showed significantly enhanced FcγR-mediated cytokine production as well as phagocytic ability. An examination of the molecular mechanism behind this enhancement revealed increased expression of both FcγRIIa and the common γ subunit following Pam2CSK4 treatment. Interestingly however, expression of the inhibitory receptor FcγRIIb was also modestly increased. Further investigation revealed that Pam2CSK4 also dramatically decreased the expression of SHIP, the major mediator of FcγRIIb inhibitory activity. Using a murine Her2/neu solid tumor model of antibody therapy, we found that Pam2CSK4 significantly enhanced the ability of anti-Her2 antibody to reduce the rate of tumor growth. To verify that the FcγR enhancement was not unique to the diacylated Pam2CSK4, we also tested Pam3CSK4, a related triacylated TLR2 agonist. Results showed significant enhancement in FcγR function and expression. Taken together, these findings indicate that TLR2 activation can positively modulate FcγR and suggest that TLR2 agonists should be considered for testing as adjuvants for antitumor antibody therapy.


Subject(s)
Lipopeptides/pharmacology , Monocytes/metabolism , Receptors, IgG/biosynthesis , Toll-Like Receptor 2/metabolism , Animals , Antibodies, Neoplasm/pharmacology , Humans , Male , Mice , Mice, Inbred BALB C , Monocytes/pathology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, IgG/genetics , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics
20.
J Biol Chem ; 288(6): 3691-5, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23269671

ABSTRACT

Burkholderia cenocepacia, the causative agent of cepacia syndrome, primarily affects cystic fibrosis patients, often leading to death. In the lung, epithelial cells serve as the initial barrier to airway infections, yet their responses to B. cenocepacia have not been fully investigated. Here, we examined the molecular responses of human airway epithelial cells to B. cenocepacia infection. Infection led to early signaling events such as activation of Erk, Akt, and NF-κB. Further, TNFα, IL-6, IL-8, and IL-1ß were all significantly induced upon infection, but no IL-1ß was detected in the supernatants. Because caspase-1 is required for IL-1ß processing and release, we examined its expression in airway epithelial cells. Interestingly, little to no caspase-1 was detectable in airway epithelial cells. Transfection of caspase-1 into airway epithelial cells restored their ability to secrete IL-1ß following B. cenocepacia infection, suggesting that a deficiency in caspase-1 is responsible, at least in part, for the attenuated IL-1ß secretion.


Subject(s)
Bronchi/metabolism , Burkholderia Infections/metabolism , Burkholderia cenocepacia , Epithelial Cells/metabolism , Interleukin-1beta/metabolism , Respiratory Mucosa/metabolism , Bronchi/microbiology , Bronchi/pathology , Burkholderia Infections/genetics , Burkholderia Infections/microbiology , Burkholderia Infections/pathology , Caspase 1/biosynthesis , Caspase 1/genetics , Cell Line , Cytokines/biosynthesis , Cytokines/genetics , Epithelial Cells/microbiology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-1beta/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL