Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Sci Rep ; 14(1): 4125, 2024 02 19.
Article in English | MEDLINE | ID: mdl-38374336

ABSTRACT

Primary effusion lymphoma (PEL) and a form of multicentric Castleman's disease (MCD) are both caused by Kaposi sarcoma herpesvirus (KSHV). There is a critical need for improved therapies for these disorders. The IL-6/JAK/STAT3 pathway plays an important role in the pathogenesis of both PEL and KSHV-MCD. We explored the potential of JAK inhibitors for use in PEL and KSHV-MCD, and found that pacritinib was superior to others in inhibiting the growth of PEL cell lines. Pacritinib induced apoptosis in PEL cells and inhibited STAT3 and NF-κB activity as evidenced by reduced amount of phosphorylated moieties. Pacritinib also inhibits FLT3, IRAK1, and ROS1; studies utilizing other inhibitors of these targets revealed that only FLT3 inhibitors exhibited similar cell growth inhibitory effects. FLT3's likely contribution to pacritinib's cell growth inhibition was further demonstrated by siRNA knockdown of FLT3. RNA sequencing and RT-PCR showed that many key host genes including cyclins and IL-6 were downregulated by pacritinib, while KSHV genes were variably altered. Finally, pacritinib suppressed KSHV viral IL-6-induced human IL-6 and IL-10 production in peripheral blood mononuclear cells, which may model an important step in KSHV-MCD pathogenesis. These results suggest that pacritinib warrants testing for the treatment of KSHV-MCD and PEL.


Subject(s)
Bridged-Ring Compounds , Castleman Disease , Herpesvirus 8, Human , Lymphoma, Primary Effusion , Pyrimidines , Humans , Interleukin-6/metabolism , Lymphoma, Primary Effusion/metabolism , Cytokines/metabolism , Leukocytes, Mononuclear/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Viral Proteins/genetics , Herpesvirus 8, Human/genetics , Cell Proliferation
2.
Curr Opin Virol ; 56: 101270, 2022 10.
Article in English | MEDLINE | ID: mdl-36182745

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) causes primary effusion lymphoma (PEL). Here, we review what is known about human gene essentiality in PEL-derived cell lines. We provide an updated list of PEL-specific human gene dependencies, based on the improved definition of core essential genes across human cancer types. The requirements of PEL cell lines for interferon regulatory factor 4 (IRF4), basic leukine zipper ATF-like transcription factor (BATF), G1/S cyclin D2 (CCND2), CASP8 and FADD like apoptosis regulator (CFLAR), MCL1 apoptosis regulator (MCL1), and murine double minute 2 (MDM2) have been confirmed experimentally. KSHV co-opts IRF4 and BATF to drive superenhancer (SE)-mediated expression of IRF4 itself, MYC, and CCND2. IRF4 dependency of SE-mediated gene expression is shared with Epstein-Barr virus-transformed lymphoblastoid cell lines (LCLs) and human T-cell leukemia virus type 1-transformed adult T-cell leukemia/lymphoma (ATLL) cell lines, as well as several B-cell lymphomas of nonviral etiology. LCLs and ATLL cell lines similarly share dependencies on CCND2 and CFLAR with PEL, but also have distinct gene dependencies. Genetic dependencies could be exploited for therapeutic intervention in PEL and other cancers.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 8, Human , Leukemia-Lymphoma, Adult T-Cell , Lymphoma, Primary Effusion , Neoplasms , Adult , Humans , Animals , Mice , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/therapeutic use , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/genetics
3.
Cell Oncol (Dordr) ; 45(5): 967-989, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35964258

ABSTRACT

BACKGROUND: Primary effusion lymphoma (PEL) is an aggressive form of non-Hodgkin lymphoma of B cells caused by Kaposi's Sarcoma-associated Herpes Virus (KSHV). KSHV encoded latent and lytic antigens promote oncogenic transformation and evade apoptosis through the modulation of various host cellular signaling pathways. Nm23-H1 is a known metastatic suppressor whose expression inversely correlates with the metastatic potential of different cancers. Here, we set out to assess the role of Nm23-H1 in PEL development. METHODS: Flow cytometry and real-time PCR assays were performed for Nm23-H1 expression analysis. Induction of apoptosis was assessed using Western blotting and flow cytometry-based assays in Nm23-H1 overexpressing cells. Co-immunoprecipitation assays, confocal microscopy and imaging flow cytometry were performed to determine Nm23-H1 and vFLIP K13 protein-protein interaction. A PEL cell-induced xenograft model was established in non-obese diabetic/severely combined immunodeficient (NOD/SCID) mice to validate the effect of Nm23-H1 overexpression. RESULTS: We found that Nm23-H1 expression was significantly downregulated both at transcriptional and protein levels in PEL cell lines and that its overexpression triggered mitochondrial-mediated caspase-dependent apoptosis. We revealed Nm23-H1 interacts with the latent protein vFLIP K13 and that Nm23-H1 overexpression leads to inhibition of vFLIP K13 driven nuclear factor-kappa B (NF-κB) signaling with concurrent inhibition of autocrine and paracrine growth factors and downregulation of latent KSHV antigens without induction of active lytic reactivation. We also confirmed the effects of Nm23-H1 overexpression in a PEL cell-induced xenograft model in NOD/SCID mice. CONCLUSION: Downregulation of Nm23-H1 expression in KSHV-infected PEL cells and its overexpression trigger apoptosis by impairing vFLIP K13-driven NF-κB signaling, suggesting therapeutic implications of Nm23-H1 for primary effusion lymphomas.


Subject(s)
Herpesvirus 8, Human , Lymphoma, Primary Effusion , Sarcoma, Kaposi , Animals , Humans , Mice , Apoptosis , Herpesvirus 8, Human/metabolism , Lymphoma, Primary Effusion/metabolism , Mice, Inbred NOD , Mice, SCID , NF-kappa B/metabolism , Oncogene Proteins/metabolism , Sarcoma, Kaposi/metabolism , Viral Proteins/metabolism
4.
Leuk Res ; 116: 106826, 2022 05.
Article in English | MEDLINE | ID: mdl-35316715

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive B-cell non-Hodgkin lymphoma in immunocompromised individuals such as AIDS patients. PEL shows a poor prognosis (median survival time < 6 months) compared with other AIDS-related lymphomas, and is generally resistant to conventional treatments. Novel drugs for PEL treatment are required. Midkine inhibitor (iMDK) was previously found to suppress midkine protein expression. Interestingly, iMDK suppressed cell proliferation in PEL cell lines in a time- and dose-dependent manner, regardless of midkine gene expression. We examined the mechanism of iMDK on PEL. Importantly, iMDK strongly induced cell cycle arrest at the G2/M phase within 12 h of incubation and suppressed the p-CDK1 protein level, which is associated with the cell cycle checkpoint at G2/M, resulting in mitotic catastrophe with observation of multipolar division. After mitotic catastrophe, iMDK-treated PEL showed apoptosis with caspase-3, - 8, and - 9 activation at 24 h incubation. However, iMDK showed no effects on viral protein-activated signaling pathways such as JAK-STAT, PI3K-Akt and NF-κB, and HHV-8/KSHV gene expression in PEL. These results indicate that iMDK is a novel CDK1 inhibitor and a promising lead compound for PEL chemotherapy treatment.


Subject(s)
Herpesvirus 8, Human , Lymphoma, Primary Effusion , Apoptosis , G2 Phase Cell Cycle Checkpoints , Humans , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Midkine/pharmacology , Phosphatidylinositol 3-Kinases/pharmacology , Phosphatidylinositol 3-Kinases/therapeutic use
5.
Cancer Immunol Immunother ; 71(10): 2497-2509, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35262781

ABSTRACT

Primary effusion lymphoma (PEL) is a rare aggressive B-cell non-Hodgkin's lymphoma with no optimal treatment. Signaling lymphocytic activation molecule-F7 (SLAMF7, CD319), a type I transmembrane glycoprotein highly expressed in multiple myeloma (MM), represents a promising target for mAb-based immunotherapy. SLAMF7 also expresses on several hematopoietic lineages including NK cells. Elotuzumab (Elo), a humanized antibody targeting SLAMF7, is approved by FDA for MM treatment. In this study, we analyzed the expression of SLAMF7 on seven PEL cell lines. All PEL cells and NK cells showed high expression of SLAMF7. NK cells were enriched from PBMCs of healthy donors by MACS and expanded by co-culturing with MHC-class I negative K562 cells in the presence of IL-2 and IL-15. Expanded NK cells showed direct killing, and Elo demonstrated potent ADCC against PEL in an Effector:Target (E:T) dependent manner. Surface expression of CD107a on NK cells also increased in the process of ADCC. We also examined SLAMF7 expression of NK subpopulations and found that the CD56+CD16+ NK subpopulation demonstrated the highest SLAMF7 expression. Full-length-Elo but not F(ab')2-Elo exerts direct engagement to the expressing SLAMF7 on NK cells, promotes CD107a expression, and further augments NK cytotoxicity toward PEL. Elo enhanced survival of PEL-bearing immunodeficient mice with adoptive transfer of human NK cells. Taken together, our results show that NK cells play roles in PEL killing, and Elo causes ADCC/SLAMF7 ligation to boost NK cytotoxicity against PEL, offering promising preclinical evidence of Elo as a therapeutic monoclonal antibody treatment for PEL.


Subject(s)
Antineoplastic Agents , Lymphoma, Primary Effusion , Multiple Myeloma , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Antineoplastic Agents/therapeutic use , Humans , Killer Cells, Natural , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/metabolism , Mice , Multiple Myeloma/drug therapy
6.
Sci Rep ; 12(1): 1085, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058488

ABSTRACT

Overexpression of antiapoptotic BCL2 family proteins occurs in various hematologic malignancies and contributes to tumorigenesis by inhibiting the apoptotic machinery of the cells. Antagonizing BH3 mimetics provide an option for medication, with venetoclax as the first drug applied for chronic lymphocytic leukemia and for acute myeloid leukemia. To find additional hematologic entities with ectopic expression of BCL2 family members, we performed expression screening of cell lines applying the LL-100 panel. Anaplastic large cell lymphoma (ALCL) and primary effusion lymphoma (PEL), 2/22 entities covered by this panel, stood out by high expression of MCL1 and low expression of BCL2. The MCL1 inhibitor AZD-5991 induced apoptosis in cell lines from both malignancies, suggesting that this BH3 mimetic might be efficient as drug for these diseases. The ALCL cell lines also expressed BCLXL and BCL2A1, both contributing to survival of the cells. The combination of specific BH3 mimetics yielded synergistic effects, pointing to a novel strategy for the treatment of ALCL. The PI3K/mTOR inhibitor BEZ-235 could also efficiently be applied in combination with AZD-5991, offering an alternative to avoid thrombocytopenia which is associated with the use of BCLXL inhibitors.


Subject(s)
Lymphoma, Large-Cell, Anaplastic/metabolism , Lymphoma, Primary Effusion/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Imidazoles/pharmacology , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/genetics , Macrocyclic Compounds/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinolines/pharmacology
7.
Biochim Biophys Acta Mol Cell Res ; 1869(1): 119168, 2022 01.
Article in English | MEDLINE | ID: mdl-34728235

ABSTRACT

The induction of DNA damage together with the interference with DNA repair represents a promising strategy in cancer treatment. Here we show that the PARP-1/2/3 inhibitor AZD2461 in combination with the CHK1 inhibitor UCN-01 altered the DNA damage response and reduced cell proliferation in PEL cells, an aggressive B cell lymphoma highly resistant to chemotherapies. AZD2461/UCN-01 combination activated p53/p21 and downregulated c-Myc in these cells, leading to a reduced expression level of RAD51, molecule involved in DNA repair. The effect of AZD2461/UCN-01 on c-Myc and p53/p21 was inter-dependent and, besides impairing cell proliferation, contributed to the activation of the replicative cycle of KSHV, carried in a latent state in PEL cells. Finally, we found that the pharmacological or genetic inhibition of p21 counteracted the viral lytic cycle activation and further reduced PEL cell proliferation, suggesting that it could induce a double beneficial effect in this setting. This study unveils that, therapeutic approaches, based on the induction of DNA damage and the reduction of DNA repair, could be used to successfully treat this malignant lymphoma.


Subject(s)
Cell Proliferation , DNA Damage , Lymphoma, Primary Effusion/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/metabolism , Virus Replication , Cell Line , Cells, Cultured , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/metabolism , Herpesvirus 8, Human/physiology , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , Phthalazines/pharmacology , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/toxicity , Protein Kinase Inhibitors/toxicity , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
8.
Am J Surg Pathol ; 46(3): 353-362, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34560683

ABSTRACT

Primary effusion lymphoma (PEL) is associated with human herpesvirus 8 and frequently with Epstein-Barr virus (EBV). We report here a single-center series of 19 human immunodeficiency virus-associated PELs, including 14 EBV+ and 5 EBV- PELs. The objectives were to describe the clinicopathologic features of PELs, with a focus on programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) expression, to search for genetic alterations by targeted deep sequencing analysis, and to compare the features between EBV+ and EBV- cases. All the patients were male, and the median age at diagnosis was 47 years old (interquartile range: 40 to 56 y). Reflecting the terminal B-cell differentiation, immunophenotypic profiles showed low expression levels of B-cell markers, including CD19 (0/19), CD20 (1/19), CD79a (0/19), PAX5 (1/19), BOB1 (3/19), and OCT2 (4/19), contrasting with a common expression of CD38 (10/19), CD138 (7/19), and IRF4/MUM1 (18/19). We observed a frequent aberrant expression of T-cell markers, especially CD3 (10/19), and less frequently CD2 (2/19), CD4 (3/19), CD5 (1/19), and CD8 (0/19). Only 2 cases were PD-L1 positive on tumor cells and none PD-1 positive. With respect to immune cells, 3 samples tested positive for PD-L1 and 5 for PD-1. Our 36-gene lymphopanel revealed 7 distinct variants in 5/10 PELs, with either a single or 2 mutations per sample: B2M (n=2), CD58 (n=1), EP300 (n=1), TNFAIP3 (n=1), ARID1A (n=1), and TP53 (n=1). Finally, we did not observe any major clinical, pathologic, or immunohistochemical differences between EBV+ and EBV- PELs and the outcome was similar (2-y overall survival probability of 61.9% [95% confidence interval, 31.2-82.1] vs. 60.0% [95% confidence interval, 12.6-88.2], respectively, P=0.62).


Subject(s)
HIV Infections/complications , Lymphoma, Primary Effusion/diagnosis , Adult , Biomarkers, Tumor/metabolism , Humans , Immunohistochemistry , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Lymphoma, Primary Effusion/virology , Male , Middle Aged
9.
Mol Carcinog ; 60(8): 567-581, 2021 08.
Article in English | MEDLINE | ID: mdl-34101920

ABSTRACT

The sesquiterpene lactone parthenolide is a major component of the feverfew medicinal plant, Tanacetum parthenium. Parthenolide has been extensively studied for its anti-inflammatory and anticancer properties in several tumor models. Parthenolide's antitumor activities depend on several mechanisms but it is mainly known as an inhibitor of the nuclear factor-κB (NF-κB) pathway. This pathway is constitutively activated and induces cell survival in primary effusion lymphoma (PEL), a rare aggressive AIDS-related lymphoproliferative disorder that is commonly caused by the human herpesvirus 8 (HHV-8) infection. The aim of this study is to evaluate the targeted effect of Parthenolide both in vitro and in vivo. Herein, parthenolide significantly inhibited cell growth, induced G0 /G1 cell cycle arrest, and induced massive apoptosis in PEL cells and ascites. In addition, parthenolide inhibited the NF-ĸB pathway suppressing IĸB phosphorylation and p65 nuclear translocation. It also reduced the expression of the DNA methylase inhibitor (DNMT1). Parthenolide induced HHV-8 lytic gene expression without inhibiting latent viral gene expression. Importantly, DMAPT, the more soluble parthenolide prodrug, promoted delay in ascites development and prolonged the survival of PEL xenograft mice. This study supports the therapeutic use of parthenolide in PEL and encourages its further clinical development.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Lymphoma, Primary Effusion/drug therapy , Sesquiterpenes/pharmacology , Animals , Apoptosis/drug effects , Biomarkers , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Disease Models, Animal , Disease Susceptibility , Drug Evaluation, Preclinical , Humans , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
IUBMB Life ; 73(7): 968-977, 2021 07.
Article in English | MEDLINE | ID: mdl-33987937

ABSTRACT

Statins are inhibitors of the mevalonate pathway that besides being cholesterol lowering agents, display anti-cancer properties. This is because cholesterol is an essential component of cell membranes but also because the mevalonate pathway controls protein farnesylation and geranylation, processes essential for the activity of GTPase family proteins. In this study, we found that Lovastatin exerted a dose- and time-dependent cytotoxic effect against PEL cells, an aggressive B cell lymphoma strictly associated with the gammaherpesvirus KSHV and characterized by a poor response to conventional chemotherapies. At molecular level, Lovastatin by dephosphorylating STAT3, induced ERK1/2 activation that inhibited autophagy and phosphorylated p53ser15 that in turn maintained ERK1/2 activated and up-regulated p21. However, p21 played a pro-survival role in this setting, as its inhibition by UC2288 further reduced cell survival in PEL cells undergoing Lovastatin treatment. In conclusion, this study suggests that Lovastatin may represent a valid therapeutic alternative against PEL cells, especially if used in combination with p21 inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Lovastatin/pharmacology , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Tyrosine/metabolism
11.
PLoS Pathog ; 17(1): e1009179, 2021 01.
Article in English | MEDLINE | ID: mdl-33471866

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive B cell lymphoma that is etiologically linked to Kaposi's sarcoma-associated herpesvirus (KSHV). Despite standard multi-chemotherapy treatment, PEL continues to cause high mortality. Thus, new strategies to control PEL are needed urgently. Here, we show that a phosphodegron motif within the KSHV protein, latency-associated nuclear antigen (LANA), specifically interacts with E3 ubiquitin ligase FBW7, thereby competitively inhibiting the binding of the anti-apoptotic protein MCL-1 to FBW7. Consequently, LANA-FBW7 interaction enhances the stability of MCL-1 by preventing its proteasome-mediated degradation, which inhibits caspase-3-mediated apoptosis in PEL cells. Importantly, MCL-1 inhibitors markedly suppress colony formation on soft agar and tumor growth of KSHV+PEL/BCBL-1 in a xenograft mouse model. These results strongly support the conclusion that high levels of MCL-1 expression enable the oncogenesis of PEL cells and thus, MCL-1 could be a potential drug target for KSHV-associated PEL. This work also unravels a mechanism by which an oncogenic virus perturbs a key component of the ubiquitination pathway to induce tumorigenesis.


Subject(s)
Antigens, Viral/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism , Herpesvirus 8, Human/physiology , Lymphoma, Primary Effusion/virology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nuclear Proteins/metabolism , Sarcoma, Kaposi/virology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , Apoptosis , Cell Proliferation , F-Box-WD Repeat-Containing Protein 7/genetics , Female , Humans , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Nuclear Proteins/genetics , Phosphorylation , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , Tumor Cells, Cultured , Ubiquitination , Xenograft Model Antitumor Assays
12.
Oncogene ; 40(3): 536-550, 2021 01.
Article in English | MEDLINE | ID: mdl-33188297

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV, HHV-8) is a gamma herpesvirus associated with several human malignancies. Transposable elements (TEs) are ubiquitous in eukaryotic genomes, occupying about 45% of the human genome. TEs have been linked with a variety of disorders and malignancies, though the precise nature of their contribution to many of them has yet to be elucidated. Global transcriptome analysis for differentially expressed TEs in KSHV-associated primary effusion lymphoma (PEL) cells (BCBL1 and BC3) revealed large number of differentially expressed TEs. These differentially expressed TEs include LTR transposons, long interspersed nuclear elements (LINEs), and short interspersed nuclear elements (SINEs). Further analysis of LINE-1 (L1) elements revealed expression upregulation, hypo-methylation, and transition into open chromatin in PEL. In agreement with high L1 expression, PEL cells express ORF1 protein and possess high reverse transcriptase (RT)-activity. Interestingly, inhibition of this RT-activity suppressed PEL cell growth. Collectively, we identified high expression of TEs, and specifically of L1 as a critical component in the proliferation of PEL cells. This observation is relevant for the treatment of KSHV-associated malignancies since they often develop in AIDS patients that are treated with RT inhibitors with potent inhibition for both HIV and L1 RT activity.


Subject(s)
Gene Expression Regulation, Neoplastic , Herpesviridae Infections/metabolism , Herpesvirus 8, Human/metabolism , Long Interspersed Nucleotide Elements , Lymphoma, Primary Effusion/metabolism , Cell Line, Tumor , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Herpesvirus 8, Human/genetics , Humans , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/pathology , Lymphoma, Primary Effusion/virology
13.
Proc Natl Acad Sci U S A ; 117(35): 21618-21627, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817485

ABSTRACT

Enhancers play indispensable roles in cell proliferation and survival through spatiotemporally regulating gene transcription. Active enhancers and superenhancers often produce noncoding enhancer RNAs (eRNAs) that precisely control RNA polymerase II activity. Kaposi's sarcoma-associated herpesvirus (KSHV) is a human oncogenic gamma-2 herpesvirus that causes Kaposi's sarcoma and primary effusion lymphoma (PEL). It is well characterized that KSHV utilizes host epigenetic machineries to control the switch between two lifecycles, latency and lytic replication. However, how KSHV impacts host epigenome at different stages of viral lifecycle is not well understood. Using global run-on sequencing (GRO-seq) and chromatin-immunoprecipitation sequencing (ChIP-seq), we profiled the dynamics of host transcriptional regulatory elements during latency and lytic replication of KSHV-infected PEL cells. This revealed that a number of critical host genes for KSHV latency, including MYC proto-oncogene, were under the control of superenhancers whose activities were globally repressed upon viral reactivation. The eRNA-expressing MYC superenhancers were located downstream of the MYC gene in KSHV-infected PELs and played a key role in MYC expression. RNAi-mediated depletion or dCas9-KRAB CRISPR inhibition of eRNA expression significantly reduced MYC mRNA level in PELs, as did the treatment of an epigenomic drug that globally blocks superenhancer function. Finally, while cellular IRF4 acted upon eRNA expression and superenhancer function for MYC expression during latency, KSHV viral IRF4 repressed cellular IRF4 expression, decreasing MYC expression and thereby, facilitating lytic replication. These results indicate that KSHV acts as an epigenomic driver that modifies host epigenomic status upon reactivation by effectively regulating host enhancer function.


Subject(s)
Gene Expression Regulation, Viral/genetics , Herpesvirus 8, Human/genetics , Lymphoma, Primary Effusion/genetics , Cell Line , Epigenomics/methods , Genes, myc/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Immediate-Early Proteins/genetics , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Nuclear Proteins/metabolism , Proto-Oncogene Mas , RNA/metabolism , Sarcoma, Kaposi/virology , Trans-Activators/metabolism , Transcription, Genetic/genetics , Viral Proteins/metabolism , Virus Activation/genetics , Virus Latency/genetics , Virus Replication/genetics
14.
Blood ; 136(19): 2175-2187, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32518949

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive malignancy with poor prognosis even under chemotherapy. Kaposi sarcoma-associated herpesvirus (KSHV), one of the human oncogenic viruses, is the principal causative agent. Currently, there is no specific treatment for PEL; therefore, developing new therapies is of great importance. Sphingolipid metabolism plays an important role in determining the fate of tumor cells. Our previous studies have demonstrated that there is a correlation between sphingolipid metabolism and KSHV+ tumor cell survival. To further develop sphingolipid metabolism-targeted therapy, after screening a series of newly synthesized ceramide analogs, here, we have identified compounds with effective anti-PEL activity. These compounds induce significant PEL apoptosis, cell-cycle arrest, and intracellular ceramide production through regulation of ceramide synthesizing or ceramide metabolizing enzymes and dramatically suppress tumor progression without visible toxicity in vivo. These new compounds also increase viral lytic gene expression in PEL cells. Our comparative transcriptomic analysis revealed their mechanisms of action for inducing PEL cell death and identified a subset of novel cellular genes, including AURKA and CDCA3, controlled by sphingolipid metabolism, and required for PEL survival with functional validation. These data provide the framework for the development of promising sphingolipid-based therapies against this virus-associated malignancy.


Subject(s)
Aurora Kinase A/metabolism , Ceramides/pharmacology , Herpesvirus 8, Human/pathogenicity , Lymphoma, Primary Effusion/drug therapy , Sarcoma, Kaposi/complications , Sphingolipids/pharmacology , Animals , Apoptosis , Aurora Kinase A/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Survival , Ceramides/chemistry , Female , Gene Expression Profiling , Humans , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Sarcoma, Kaposi/virology , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
15.
Nat Commun ; 11(1): 1786, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286300

ABSTRACT

Protein-protein interactions featuring intricate binding epitopes remain challenging targets for synthetic inhibitors. Interactions of NEMO, a scaffolding protein central to NF-κB signaling, exemplify this challenge. Various regulators are known to interact with different coiled coil regions of NEMO, but the topological complexity of this protein has limited inhibitor design. We undertook a comprehensive effort to block the interaction between vFLIP, a Kaposi's sarcoma herpesviral oncoprotein, and NEMO using small molecule screening and rational design. Our efforts reveal that a tertiary protein structure mimic of NEMO is necessary for potent inhibition. The rationally designed mimic engages vFLIP directly causing complex disruption, protein degradation and suppression of NF-κB signaling in primary effusion lymphoma (PEL). NEMO mimic treatment induces cell death and delays tumor growth in a PEL xenograft model. Our studies with this inhibitor reveal the critical nexus of signaling complex stability in the regulation of NF-κB by a viral oncoprotein.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Lymphoma, Primary Effusion/metabolism , NF-kappa B/metabolism , Animals , Cell Line , Circular Dichroism , Herpesvirus 8, Human/metabolism , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Lymphoma, Primary Effusion/genetics , Male , Mice , Microscopy, Confocal , Models, Biological , Signal Transduction/genetics , Signal Transduction/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Xenograft Model Antitumor Assays
16.
J Virol ; 94(10)2020 05 04.
Article in English | MEDLINE | ID: mdl-32161170

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV) is necessary but not sufficient for primary effusion lymphoma (PEL) development. Alterations in cellular signaling pathways are also a characteristic of PEL. Other B cell lymphomas have acquired an oncogenic mutation in the myeloid differentiation primary response 88 (MYD88) gene. The MYD88 L265P mutant results in the activation of interleukin-1 receptor associated kinase (IRAK). To probe IRAK/MYD88 signaling in PEL, we employed CRISPR/Cas9 technology to generate stable deletion clones in BCBL-1Cas9 and BC-1Cas9 cells. To look for off-target effects, we determined the complete exome of the BCBL-1Cas9 and BC-1Cas9 cells. Deletion of either MYD88, IRAK4, or IRAK1 abolished interleukin-1 beta (IL-1ß) signaling; however, we were able to grow stable subclones from each population. Transcriptome sequencing (RNA-seq) analysis of IRAK4 knockout cell lines (IRAK4 KOs) showed that the IRAK pathway induced cellular signals constitutively, independent of IL-1ß stimulation, which was abrogated by deletion of IRAK4. Transient complementation with IRAK1 increased NF-κB activity in MYD88 KO, IRAK1 KO, and IRAK4 KO cells even in the absence of IL-1ß. IL-10, a hallmark of PEL, was dependent on the IRAK pathway, as IRAK4 KOs showed reduced IL-10 levels. We surmise that, unlike B cell receptor (BCR) signaling, MYD88/IRAK signaling is constitutively active in PEL, but that under cell culture conditions, PEL rapidly became independent of this pathway.IMPORTANCE One hundred percent of primary effusion lymphoma (PEL) cases are associated with Kaposi sarcoma-associated herpesvirus (KSHV). PEL cell lines, such as BCBL-1, are the workhorse for understanding this human oncovirus and the host pathways that KSHV dysregulates. Understanding their function is important for developing new therapies as well as identifying high-risk patient groups. The myeloid differentiation primary response 88 (MYD88)/interleukin-1 receptor associated kinase (IRAK) pathway, which has progrowth functions in other B cell lymphomas, has not been fully explored in PEL. By performing CRISPR/Cas9 knockout (KO) studies targeting the IRAK pathway in PEL, we were able to determine that established PEL cell lines can circumvent the loss of IRAK1, IRAK4, and MYD88; however, the deletion clones are deficient in interleukin-10 (IL-10) production. Since IL-10 suppresses T cell function, this suggests that the IRAK pathway may serve a function in vivo and during early-stage development of PEL.


Subject(s)
Herpesvirus 8, Human/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/genetics , B-Lymphocytes , CRISPR-Cas Systems , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Herpesvirus 8, Human/physiology , Humans , Interleukin-10/metabolism , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Sequence Analysis , Transcriptome
17.
Toxicol In Vitro ; 65: 104753, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31857147

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive neoplasm correlated with human herpesvirus 8 (HHV8). Metabolic reprogramming is a hallmark of cancers. The alterations in cellular metabolism are important to the survival of HHV8 latently infected cells. Pyruvate dehydrogenase (PDH) controls the flux of metabolites between glycolysis and the tricarboxylic acid cycle (TCA cycle) and is a key enzyme in cancer metabolic reprogramming. Glutaminolysis is required for the survival of PEL cells. Glutamate dehydrogenase 1 (GDH1) converts glutamate into α-ketoglutarate supplying the TCA cycle with intermediates to support anaplerosis. Previously we have observed that epigallocatechin-3-gallate (EGCG) can induce PEL cell death and N-acetyl cysteine (NAC) attenuates EGCG induced PEL cell death. In this study, results showed that EGCG upregulated the expression of glucose transporter GLUT3, and reduced the expression of pyruvate dehydrogenase E1-alpha (PDHA1), the major regulator of PDH, and GDH1. NAC could partially reverse the effects of EGCG in PEL cells. Overexpression of PDHA1 in PEL cells or supplement of α-ketoglutarate attenuated EGCG induced cell death. EGCG also reduced the levels of oncometabolite D-2-hydroxyglutarate (D2HG). These results suggest that EGCG may modulate the metabolism of PEL cells leading to cell death.


Subject(s)
Catechin/analogs & derivatives , Herpesvirus 8, Human , Lymphoma, Primary Effusion/metabolism , Metabolic Networks and Pathways/drug effects , Pyruvate Dehydrogenase (Lipoamide)/genetics , Catechin/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/drug effects , Glutarates/metabolism , Humans , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology
18.
Diagn Cytopathol ; 48(4): 380-385, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31846233

ABSTRACT

Primary effusion lymphoma (PEL) is a rare non-Hodgkin's lymphoma most commonly occurring in the context of human immune deficiency (HIV) infection. Herpes virus 8 (HHV-8) has been associated with PEL and considered to be the etiologic agent. In addition, most cases (60%-90%) also show evidence of Epstein-Barr virus (EBV) infection. We describe here an elderly man who was HIV seronegative and immunocompetent, and presented with worsening weakness and ascites. The diagnosis of PEL was rendered cytologically and supported by the results of flow cytometry. The presence of HHV-8 was demonstrated by immunohistochemistry, whereas EBV-associated genetic material was absent by EBER ISH. No lymphadenopathy or organ involvement with lymphoma was found. Systemic chemotherapy with lenalidomide was started given the poor prognosis and commodities of severe coronary artery disease; however, the patient did not respond and succumbed to his disease in 4 months. We present detailed cytologic and clinical findings of this very rare occurrence, and review literature of all reported PEL cases of HIV-negative, nontransplant, immunocompetent patients.


Subject(s)
HIV Seronegativity , Herpesviridae Infections , Herpesvirus 8, Human/metabolism , Lenalidomide/administration & dosage , Lymphoma, Primary Effusion , Aged , Fatal Outcome , Herpesviridae Infections/diagnosis , Herpesviridae Infections/drug therapy , Herpesviridae Infections/metabolism , Humans , Lymphoma, Primary Effusion/diagnosis , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Male
19.
Blood Adv ; 3(14): 2105-2117, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31300418

ABSTRACT

Genome-wide CRISPR/Cas9 screens represent a powerful approach to studying mechanisms of drug action and resistance. Cereblon modulating agents (CMs) have recently emerged as candidates for therapeutic intervention in primary effusion lymphoma (PEL), a highly aggressive cancer caused by Kaposi's sarcoma-associated herpesvirus. CMs bind to cereblon (CRBN), the substrate receptor of the cullin-RING type E3 ubiquitin ligase CRL4CRBN, and thereby trigger the acquisition and proteasomal degradation of neosubstrates. Downstream mechanisms of CM toxicity are incompletely understood, however. To identify novel CM effectors and mechanisms of CM resistance, we performed positive selection CRISPR screens using 3 CMs with increasing toxicity in PEL: lenalidomide (LEN), pomalidomide (POM), and CC-122. Results identified several novel modulators of the activity of CRL4CRBN The number of genes whose inactivation confers resistance decreases with increasing CM efficacy. Only inactivation of CRBN conferred complete resistance to CC-122. Inactivation of the E2 ubiquitin conjugating enzyme UBE2G1 also conferred robust resistance against LEN and POM. Inactivation of additional genes, including the Nedd8-specific protease SENP8, conferred resistance to only LEN. SENP8 inactivation indirectly increased levels of unneddylated CUL4A/B, which limits CRL4CRBN activity in a dominant negative manner. Accordingly, sensitivity of SENP8-inactivated cells to LEN is restored by overexpression of CRBN. In sum, our screens identify several novel players in CRL4CRBN function and define pathways to CM resistance in PEL. These results provide rationale for increasing CM efficacy on patient relapse from a less-efficient CM. Identified genes could finally be developed as biomarkers to predict CM efficacy in PEL and other cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Lymphoma, Primary Effusion/etiology , Adaptor Proteins, Signal Transducing/metabolism , Cullin Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Endopeptidases/genetics , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Lenalidomide/adverse effects , Lenalidomide/pharmacology , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Models, Biological , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Ubiquitin-Protein Ligases
20.
J Virol ; 93(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31118261

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma and is associated with two B cell malignancies, primary effusion lymphoma (PEL) and the plasmablastic variant of multicentric Castleman's disease. On several adherent cell types, EphA2 functions as a cellular receptor for the gH/gL glycoprotein complex of KSHV. KSHV gH/gL also has previously been found to interact weakly with other members of the Eph family of receptor tyrosine kinases (Ephs), and other A-type Ephs have been shown to be able to compensate for the absence of EphA2 using overexpression systems. However, whether these interactions are of functional consequence at endogenous protein levels has remained unclear so far. Here, we demonstrate for the first time that endogenously expressed EphA7 in BJAB B cells is critical for the cell-to-cell transmission of KSHV from producer iSLK cells to BJAB target cells. The BJAB lymphoblastoid cell line often serves as a model for B cell infection and expresses only low levels of all Eph family receptors other than EphA7. Endogenous EphA7 could be precipitated from the cellular lysate of BJAB cells using recombinant gH/gL, and knockout of EphA7 significantly reduced transmission of KSHV into BJAB target cells. Knockout of EphA5, the second most expressed A-type Eph in BJAB cells, had a similar, although less pronounced, effect on KSHV infection. Receptor function of EphA7 was conserved for cell-free infection by the related rhesus monkey rhadinovirus (RRV), which is relatively even more dependent on EphA7 for infection of BJAB cells.IMPORTANCE Infection of B cells is relevant for two KSHV-associated malignancies, the plasmablastic variant of multicentric Castleman's disease and PEL. Therefore, elucidating the process of B cell infection is important for the understanding of KSHV pathogenesis. While the high-affinity receptor for the gH/gL glycoprotein complex, EphA2, has been shown to function as an entry receptor for various types of adherent cells, the gH/gL complex can also interact with other Eph receptor tyrosine kinases with lower avidity. We analyzed the Eph interactions required for infection of BJAB cells, a model for B cell infection by KSHV. We identified EphA7 as the principal Eph receptor for infection of BJAB cells by KSHV and the related rhesus monkey rhadinovirus. While two analyzed PEL cell lines exhibited high EphA2 and low EphA7 expression, a third PEL cell line, BCBL-1, showed high EphA7 and low EphA2 expression, indicating a possible relevance for KSHV pathology.


Subject(s)
B-Lymphocytes/metabolism , Receptor, EphA7/metabolism , Receptors, Virus/metabolism , Rhadinovirus/physiology , Virus Internalization , Animals , B-Lymphocytes/pathology , B-Lymphocytes/virology , Cell Line, Tumor , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Herpesvirus 8, Human/physiology , Humans , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Macaca mulatta , Receptor, EphA7/genetics , Receptors, Virus/genetics , Rhadinovirus/genetics , Rhadinovirus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...