Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 116(33): 16541-16550, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31346082

ABSTRACT

Non-Hodgkin lymphomas (NHLs) make up the majority of lymphoma diagnoses and represent a very diverse set of malignancies. We sought to identify kinases uniquely up-regulated in different NHL subtypes. Using multiplexed inhibitor bead-mass spectrometry (MIB/MS), we found Tyro3 was uniquely up-regulated and important for cell survival in primary effusion lymphoma (PEL), which is a viral lymphoma infected with Kaposi's sarcoma-associated herpesvirus (KSHV). Tyro3 was also highly expressed in PEL cell lines as well as in primary PEL exudates. Based on this discovery, we developed an inhibitor against Tyro3 named UNC3810A, which hindered cell growth in PEL, but not in other NHL subtypes where Tyro3 was not highly expressed. UNC3810A also significantly inhibited tumor progression in a PEL xenograft mouse model that was not seen in a non-PEL NHL model. Taken together, our data suggest Tyro3 is a therapeutic target for PEL.


Subject(s)
Lymphoma, Non-Hodgkin/enzymology , Lymphoma, Primary Effusion/enzymology , Molecular Targeted Therapy , Proteome/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/virology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Up-Regulation/drug effects
2.
Cancer Epidemiol Biomarkers Prev ; 28(6): 1036-1044, 2019 06.
Article in English | MEDLINE | ID: mdl-30593457

ABSTRACT

BACKGROUND: Despite extensive research to identify biomarkers of response in patients with non-muscle-invasive bladder cancer (NMIBC), there is no biomarker to date that can serve this purpose. Herein, we report how we leveraged serial urine samples to query a panel of cytokines at varying time points in an attempt to identify predictive biomarkers of response in NMIBC. METHODS: Serial urine samples were collected from 50 patients with intermediate- or high-risk NMIBC enrolled in a phase II study, evaluating intravesical BCG ± intradermal HS-410 therapy. Samples were collected at baseline, week 7, week 13, week 28, and at end of treatment. A total of 105 cytokines were analyzed in each sample. To predict outcome of time-to-event (recurrence or progression), univariate and multivariable Cox analyses were performed. RESULTS: Fifteen patients developed recurrence and 4 patients progressed during the follow-up period. Among clinicopathologic variables, ever-smoker versus nonsmoker status was associated with an improved response rate (HR 0.38; 95% confidence interval (CI), 0.14-0.99; P = 0.04). In the most clinically relevant model, the percent change (for 100 units) of IL18-binding protein-a (HR 1.995; 95% CI, 1.16-3.44; P = 0.01), IL23 (HR 1.12; 95% CI, 1.01-1.23; P = 0.03), IL8 (HR 0.27; 95% CI, 0.07-1.08; P = 0.06), and IFNγ-induced protein-10 (HR 0.95; 95% CI, 0.91-0.99; P = 0.04) at week 13 from baseline best predicted time to event. CONCLUSIONS: Urinary cytokines provided additional value to clinicopathologic features to predict response to immune-modulating agents in patients with NMIBC. IMPACT: This study serves as a hypothesis-generating report for future studies to evaluate the role of urine cytokines as a predictive biomarker of response to immune treatments.


Subject(s)
BCG Vaccine/administration & dosage , Cancer Vaccines/administration & dosage , Cytokines/urine , Neoplasm Recurrence, Local/therapy , Neoplasm Recurrence, Local/urine , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/urine , Administration, Intravesical , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biomarkers, Tumor/urine , Clinical Trials, Phase II as Topic , Drug Synergism , Female , Follow-Up Studies , Humans , Injections, Intradermal , Male , Models, Statistical , Neoplasm Invasiveness , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Predictive Value of Tests , Prognosis , Randomized Controlled Trials as Topic , Survival Rate , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
3.
PLoS Pathog ; 12(11): e1005985, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27829024

ABSTRACT

Kaposi's sarcoma herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS) as well as two lymphoproliferative diseases, primary effusion lymphoma and multicentric Castleman's disease. KSHV encodes viral proteins, such as K1, that alter signaling pathways involved in cell survival. Expression of K1 has been reported to transform rodent fibroblasts, and K1 transgenic mice develop multiple tumors, suggesting that K1 has an important role in KSHV pathogenesis. We found that cells infected with a KSHV virus containing a WT K1 gene had a survival advantage under conditions of nutrient deprivation compared to cells infected with KSHV K1 mutant viruses. 5' adenosine monophosphate-activated protein kinase (AMPK) responds to nutrient deprivation by maintaining energy homeostasis, and AMPK signaling has been shown to promote cell survival in various types of cancers. Under conditions of AMPK inhibition, we also observed that cells infected with KSHV containing a WT K1 gene had a survival advantage compared to KSHV K1 mutant virus infected cells. To explore the underpinnings of this phenotype, we identified K1-associated cellular proteins by tandem affinity purification and mass spectrometry. We found that the KSHV K1 protein associates with the gamma subunit of AMPK (AMPKγ1). We corroborated this finding by independently confirming that K1 co-immunoprecipitates with AMPKγ1. Co-immunoprecipitations of wild-type K1 (K1WT) or K1 domain mutants and AMPKγ1, revealed that the K1 N-terminus is important for the association between K1 and AMPKγ1. We propose that the KSHV K1 protein promotes cell survival via its association with AMPKγ1 following exposure to stress.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Herpesvirus 8, Human/metabolism , Viral Proteins/metabolism , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Confocal
4.
Cancer Immunol Res ; 4(9): 766-78, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27364122

ABSTRACT

T-cell costimulation typically occurs in a defined microenvironment that is not recapitulated by agonistic antibody therapy. To deliver such stimulation under more favorable conditions, we investigated whether an allogeneic cell-based vaccine that secreted Fc-OX40L, Fc-ICOSL, or Fc-4-1BBL would activate and expand T cells comparably with systemically administered agonist antibodies. Among these costimulators, locally secreted Fc-OX40L provided superior priming of antigen-specific CD8(+) T cells, compared with combinations with OX40 antibodies or vaccine alone. Vaccine-expressed Fc-OX40L also stimulated IFNγ, TNFα, granzyme B, and IL2 by antigen-specific CD8(+) T cells similarly to OX40 antibodies, without off-target consequences such as proinflammatory cytokine induction. Vaccine-secreted Fc-OX40L increased CD127(+)KLRG-1(-) memory precursor cells during the contraction phase, resulting in improved proliferation upon secondary antigen challenge, as compared with OX40 antibody. A cell-based vaccine cosecreting gp96-Ig and Fc-OX40L led to even more pronounced tumor control, complete tumor rejection, and increased tumor antigen-specific T-cell proliferation, including in tumor-infiltrating lymphocytes, as compared with combinations of gp96-Ig vaccine and OX40 antibodies, in mice with established melanoma or colorectal carcinoma. These data suggest that local modulation of the vaccine microenvironment has unexpected advantages over systemic costimulation with agonistic antibodies, which may simplify the clinical translation of such combination immunotherapies into humans. Cancer Immunol Res; 4(9); 766-78. ©2016 AACR.


Subject(s)
Cancer Vaccines/immunology , Immunity , Immunologic Memory , Neoplasms/immunology , T-Lymphocytes/immunology , 4-1BB Ligand/antagonists & inhibitors , 4-1BB Ligand/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Disease Models, Animal , Melanoma, Experimental , Membrane Glycoproteins , Mice , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes/metabolism
5.
Proc Natl Acad Sci U S A ; 113(28): 7876-81, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27342859

ABSTRACT

Viruses depend upon the host cell for manufacturing components of progeny virions. To mitigate the inextricable dependence on host cell protein synthesis, viruses can modulate protein synthesis through a variety of mechanisms. We report that the viral protein kinase (vPK) encoded by open reading frame 36 (ORF36) of Kaposi's sarcoma-associated herpesvirus (KSHV) enhances protein synthesis by mimicking the function of the cellular protein S6 kinase (S6KB1). Similar to S6KB1, vPK phosphorylates the ribosomal S6 protein and up-regulates global protein synthesis. vPK also augments cellular proliferation and anchorage-independent growth. Furthermore, we report that both vPK and S6KB1 phosphorylate the enzyme 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase 2 (PFKFB2) and that both kinases promote endothelial capillary tubule formation.


Subject(s)
Herpesvirus 8, Human/enzymology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Viral Proteins/metabolism , Computer Simulation , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Models, Molecular , Ribosomal Protein S6 Kinases, 70-kDa/chemistry , Substrate Specificity , Viral Proteins/chemistry
6.
mBio ; 6(6): e01499-15, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26646010

ABSTRACT

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of human Kaposi's sarcoma, a tumor that arises from endothelial cells, as well as two B cell lymphoproliferative diseases, primary effusion lymphoma and multicentric Castleman's disease. KSHV utilizes a variety of mechanisms to evade host immune responses and promote cellular transformation and growth in order to persist for the life of the host. A viral homolog of human interleukin-6 (hIL-6) named viral interleukin-6 (vIL-6) is encoded by KSHV and expressed in KSHV-associated cancers. Similar to hIL-6, vIL-6 is secreted, but the majority of vIL-6 is retained within the endoplasmic reticulum, where it can initiate functional signaling through part of the interleukin-6 receptor complex. We sought to determine how intracellular vIL-6 modulates the host endothelial cell environment by analyzing vIL-6's impact on the endothelial cell transcriptome. vIL-6 significantly altered the expression of many cellular genes associated with cell migration. In particular, vIL-6 upregulated the host factor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) at the protein and message levels. CEACAM1 has been implicated in tumor invasion and metastasis and promotes migration and vascular remodeling in endothelial cells. We report that vIL-6 upregulates CEACAM1 by a STAT3-dependent mechanism and that CEACAM1 promotes vIL-6-mediated migration. Furthermore, latent and de novo KSHV infections of endothelial cells also induce CEACAM1 expression. Collectively, our data suggest that vIL-6 modulates endothelial cell migration by upregulating the expression of cellular factors, including CEACAM1. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is linked with the development of three human malignancies, Kaposi's sarcoma, multicentric Castleman's disease, and primary effusion lymphoma. KSHV expresses many factors that enable the virus to manipulate the host environment in order to persist and induce disease. The viral interleukin-6 (vIL-6) produced by KSHV is structurally and functionally homologous to the human cytokine interleukin-6, except that vIL-6 is secreted slowly and functions primarily from inside the host cell. To investigate the unique intracellular role of vIL-6, we analyzed the impact of vIL-6 on endothelial cell gene expression. We report that vIL-6 significantly alters the expression of genes associated with cell movement, including that for CEACAM1. The gene for CEACAM1 was upregulated by vIL-6 and by latent and primary KSHV infection and promotes vIL-6-mediated endothelial cell migration. This work advances the field's understanding of vIL-6 function and its contribution to KSHV pathogenesis.


Subject(s)
Antigens, CD/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement , Endothelial Cells/virology , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , Interleukin-6/metabolism , Viral Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans
7.
J Virol ; 89(22): 11572-83, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26355087

ABSTRACT

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) is a gammaherpesvirus known to establish lifelong latency in the human host. We and others have previously shown that three KSHV homologs of cellular interferon regulatory factors (IRFs), known as viral IRFs (vIRFs), participate in evasion of the host interferon (IFN) response. We report that vIRF1 interacts with the cellular interferon-stimulated gene 15 (ISG15) E3 ligase, HERC5, in the context of Toll-like receptor 3 (TLR3) activation and IFN induction. The ISG15 protein is covalently conjugated to target proteins upon activation of the interferon response. Interaction between vIRF1 and HERC5 was confirmed by immunoprecipitation, and the region between amino acids 224 and 349 of vIRF1 was required for interaction with HERC5. We further report that expression of vIRF1 in the context of TLR3 activation results in decreased ISG15 conjugation of proteins. Specifically, TLR3-induced ISG15 conjugation and protein levels of cellular IRF3, a known ISG15 target, were decreased in the presence of vIRF1 compared to the control. vIRF1 itself was also identified as a target of ISG15 conjugation. KSHV-infected cells exhibited increased ISG15 conjugation upon reactivation from latency in coordination with increased IFN. Furthermore, knockdown of ISG15 in latently infected cells resulted in a higher level of KSHV reactivation and an increase in infectious virus. These data suggest that the KSHV vIRF1 protein affects ISG15 conjugation and interferon responses and may contribute to effective KSHV replication. IMPORTANCE: The KSHV vIRF1 protein can inhibit interferon activation in response to viral infection. We identified a cellular protein named HERC5, which is the major ligase for ISG15, as a vIRF1 binding partner. vIRF1 association with HERC5 altered ISG15 modification of cellular proteins, and knockdown of ISG15 augmented reactivation of KSHV from latency.


Subject(s)
Cytokines/metabolism , Herpesvirus 8, Human/physiology , Interferon Regulatory Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Toll-Like Receptor 3/metabolism , Ubiquitins/metabolism , Viral Proteins/metabolism , Virus Activation , Amino Acid Sequence , Cytokines/genetics , Enzyme Activation , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , RNA Interference , RNA, Small Interfering , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/genetics , Virus Latency , Virus Replication
8.
J Virol ; 88(16): 9429-41, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24920810

ABSTRACT

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV, also called human herpesvirus 8) is linked to the development of Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease (MCD). KSHV expresses several proteins that modulate host cell signaling pathways. One of these proteins is viral interleukin-6 (vIL-6), which is a homolog of human IL-6 (hIL-6). vIL-6 is able to prevent apoptosis and promote proinflammatory signaling, angiogenesis, and cell proliferation. Although it can be secreted, vIL-6 is mainly an intracellular protein that is retained in the endoplasmic reticulum (ER). We performed affinity purification and mass spectrometry to identify novel vIL-6 binding partners and found that a cellular ER chaperone, hypoxia-upregulated protein 1 (HYOU1), interacts with vIL-6. Immunohistochemical staining reveals that both PEL and KS tumor tissues express significant amounts of HYOU1. We also show that HYOU1 increases endogenous vIL-6 protein levels and that HYOU1 facilitates vIL-6-induced JAK/STAT signaling, migration, and survival in endothelial cells. Furthermore, our data suggest that HYOU1 also modulates vIL-6's ability to induce CCL2, a chemokine involved in cell migration. Finally, we investigated the impact of HYOU1 on cellular hIL-6 signaling. Collectively, our data indicate that HYOU1 is important for vIL-6 function and may play a role in the pathogenesis of KSHV-associated cancers. IMPORTANCE: KSHV vIL-6 is detectable in all KSHV-associated malignancies and promotes tumorigenesis and inflammation. We identified a cellular protein, called hypoxia-upregulated protein 1 (HYOU1), that interacts with KSHV vIL-6 and is present in KSHV-infected tumors. Our data suggest that HYOU1 facilitates the vIL-6-induced signaling, migration, and survival of endothelial cells.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Herpesvirus 8, Human/metabolism , Interleukin-6/metabolism , Viral Proteins/metabolism , Cell Line , Chemokine CCL2/metabolism , HEK293 Cells , Herpesviridae Infections/metabolism , Humans , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/virology , Signal Transduction/physiology , Up-Regulation/physiology
9.
Adv Virus Res ; 88: 111-59, 2014.
Article in English | MEDLINE | ID: mdl-24373311

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV; also known as human herpesvirus 8) is the etiologic agent of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. These cancers often occur in the context of immunosuppression, which has made KSHV-associated malignancies an increasing global health concern with the persistence of the AIDS epidemic. KSHV has also been linked to several acute inflammatory diseases. KSHV exists between a lytic and latent lifecycle, which allows the virus to transition between active replication and quiescent infection. KSHV encodes a number of proteins and small RNAs that are thought to inadvertently transform host cells while performing their functions of helping the virus persist in the infected host. KSHV also has an arsenal of components that aid the virus in evading the host immune response, which help the virus establish a successful lifelong infection. In this comprehensive chapter, we will discuss the diseases associated with KSHV infection, the biology of latent and lytic infection, and individual proteins and microRNAs that are known to contribute to host cell transformation and immune evasion.


Subject(s)
Carcinogenesis , Herpesvirus 8, Human/physiology , Host-Pathogen Interactions , Immune Evasion , Virus Latency , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...