Human Herpesvirus 8 K1 Blocks Fas in Cells and Mice by Binding and Blocking Fas.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 515-515
Author(s):  
Hoyoung Maeng ◽  
Suizhao Wang ◽  
Zuzuna Berkova ◽  
Shu Wang ◽  
Jaehoon Jung ◽  
...  

Abstract Background: Kaposi’s sarcoma and primary effusion lymphoma are linked to human herpesvirus 8 (HHV-8 or Kaposi’s sarcoma-associated virus) infection. Key to the pathogenesis of these cancers is the HHV-8 transmembrane oncoprotein K1. This viral protein, which immortalizes cells, produces lymphoproliferation and lymphomas through unknown mechanisms when expressed in mice. Results: We show here that K1 blocks Fas (CD95)-mediated apoptosis by binding the ectodomain of K1 to Fas in hematopoietic cancer cells (BJAB, U937 and THP-1). K1 bound to Fas in K1-transfected BJAB cells. To assess whether K1 and Fas formed complexes in cells, we chemically crosslinked proteins in transfected cells using 3,3′-dithiobissulfosuccinimidyl propionate in the presence of N-ethyl maleimide. Stabled K1-Fas complexes were present in intact cells despite blocking of free reactive cysteines. The selective ability of K1 to antagonize Fas-mediated apoptosis was strictly dependent on having its immunoreceptor tyrosine-based activation motif (ITAM). Using activating anti-Fas (CH-11) and nonactivating anti-Fas (B10) antibodies in a serial manner, we showed that K1 associated with Fas that was not activated, strongly suggesting that K1 prevents Fas from being activated. Mice transfected with K1 were protected from the apoptosis inducing effects of agonistic Jo2 anti-Fas antibody and tissues from these mice contained K1-Fas complexes. K1-transfected compared to vector-transfected mice had fewer terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling positive cells (13 ± 3% vs. 60 ± 7%, P=0.008), fewer activated caspase 3 positive cells (24 ± 5% vs. 88 ± 6%, P=0.006). Tissues had less hemorrhaging, and necrosis. Conclusion: These data support a mechanism of K1 direct binding to and blockage by K1 to Fas in suppression of apoptosis in an ITAM-dependent manner, which may be a key step in lymphoma development.

2002 ◽  
Vol 76 (5) ◽  
pp. 2551-2556 ◽  
Author(s):  
Isabelle Widmer ◽  
Marion Wernli ◽  
Felix Bachmann ◽  
Fred Gudat ◽  
Gieri Cathomas ◽  
...  

ABSTRACT Expression of human herpesvirus 8 viral Bcl-2 protein was demonstrated in spindle cells of late-stage Kaposi's sarcoma lesions but not in primary effusion lymphoma cell lines. In contrast, strong expression of human Bcl-2 was found in stimulated primary effusion lymphoma cells, whereas in Kaposi's sarcoma lesions preferential mononuclear cells, and to a lesser extent spindle cells, stained positive.


1999 ◽  
Vol 73 (7) ◽  
pp. 6177-6181 ◽  
Author(s):  
Johnan A. R. Kaleeba ◽  
Eric P. Bergquam ◽  
Scott W. Wong

ABSTRACT The rhesus rhadinovirus strain 17577 (RRV strain 17577) genome is essentially colinear with human herpesvirus 8 (HHV8)/Kaposi’s sarcoma-associated herpesvirus (KSHV) and encodes several analogous open reading frames (ORFs), including the homologue of cellular interleukin-6 (IL-6). To determine if the RRV IL-6-like ORF (RvIL-6) is biologically functional, it was expressed either transiently in COS-1 cells or purified from bacteria as a glutathioneS-transferase (GST)-RvIL-6 fusion and analyzed by IL-6 bioassays. Utilizing the IL-6-dependent B9 cell line, we found that both forms of RvIL-6 supported cell proliferation in a dose-dependent manner. Moreover, antibodies specific to the IL-6 receptor (IL-6R) or the gp130 subunit were capable of blocking the stimulatory effects of RvIL-6. Reciprocal titrations of GST-RvIL-6 against human recombinant IL-6 produced a more-than-additive stimulatory effect, suggesting that RvIL-6 does not inhibit but may instead potentiate normal cellular IL-6 signaling to B cells. These results demonstrate that RRV encodes an accessory protein with IL-6-like activity.


2001 ◽  
Vol 75 (10) ◽  
pp. 4843-4853 ◽  
Author(s):  
Mini Paulose-Murphy ◽  
Nguyen-Khoi Ha ◽  
Chunsheng Xiang ◽  
Yidong Chen ◽  
Laura Gillim ◽  
...  

ABSTRACT Human herpesvirus 8 (HHV-8), a gammaherpesvirus implicated in Kaposi's sarcoma, primary effusion lymphoma, and Castleman's disease, encodes several pathogenically important cellular homologs. To define the HHV-8 transcription program, RNA obtained from latently infected body cavity-based lymphoma 1 cells induced to undergo lytic replication was used to query a custom HHV-8 DNA microarray containing nearly every known viral open reading frame. The patterns of viral gene expression offer insights into the replication and pathogenic strategies of HHV-8.


2002 ◽  
Vol 15 (3) ◽  
pp. 439-464 ◽  
Author(s):  
Dharam V. Ablashi ◽  
Louise G. Chatlynne ◽  
James E. Whitman, ◽  
Ethel Cesarman

SUMMARY Human herpesvirus 8 (HHV-8), also known as Kaposi's sarcoma-associated herpesvirus (KSHV), discovered in 1994, is a human rhadinovirus (gamma-2 herpesvirus). Unlike other human herpesviruses (herpes simplex virus, Epstein-Barr virus, varicella-zoster virus, cytomegalovirus, HHV-6, and HHV-7), it is not widespread in the general population and has many unique proteins. HHV-8 is strongly associated with all subtypes of Kaposi's sarcoma (KS), multicentric Castleman's disease, and a rare form of B-cell lymphoma, primary effusion lymphoma. In addition, HHV-8 DNA sequences have been found in association with other diseases, but the role of the virus in these diseases is largely unconfirmed and remains controversial. The seroprevalence of HHV-8, based on detection of latent and lytic proteins, is 2 to 5% in healthy donors except in certain geographic areas where the virus is endemic, 80 to 95% in classic KS patients, and 40 to 50% in HIV-1 patients without KS. This virus can be transmitted both sexually and through body fluids (e.g., saliva and blood). HHV-8 is a transforming virus, as evidenced by its presence in human malignancies, by the in vitro transforming properties of several of its viral genes, and by its ability to transform some primary cells in culture. It is not, however, sufficient for transformation, and other cofactors such as immunosuppressive cytokines are involved in the development of HHV-8-associated malignancies. In this article, we review the biology, molecular virology, epidemiology, transmission, detection methods, pathogenesis, and antiviral therapy of this newly discovered human herpesvirus.


2003 ◽  
Vol 77 (17) ◽  
pp. 9399-9411 ◽  
Author(s):  
Wei Liao ◽  
Yong Tang ◽  
Yu-liang Kuo ◽  
Bao-Ying Liu ◽  
Chi-Jie Xu ◽  
...  

ABSTRACT Kaposi's sarcoma associated herpesvirus (KSHV)/human herpesvirus 8 (HHV-8) encodes an immediate early transcriptional activator, Rta, which mediates viral reactivation from latency and lytic viral replication. Here we report the purification and characterizations of HHV-8 Rta and its interaction with Rta-responsive DNA elements. The Rta response element (RtaRE) in the promoter of the KSHV/HHV-8 K8 open reading frame was mapped to a 47-bp sequence (RtaRE1) and a 60-bp sequence (RtaRE2) upstream of the TATA motif. A comparison of the K8 RtaREs with other viral RtaREs revealed a pattern of multiple A/T triplets spaced with a periodicity of 10 or 20 bp. Substitutions of the in-phase A/T trinucleotides of the RtaRE1 with G/C bases greatly diminished Rta responsiveness and Rta binding. By contrast, base substitutions in an out-of-phase A/T-trinucleotide sequence had no effect. Importantly, multimers of (A/T)3N7 and N5(A/T)5N6(A/T)4 motifs supported a strong Rta response in a copy number-dependent manner. No specific sequence motifs in the spacer regions could be discerned. Potent Rta response, however, was obtained with phased A/T trinucleotides with 7-bp spacers of arbitrary sequences with high G/C content. Lengthening of the phased A/T motifs or lowering of the G/C content of the spacers resulted in a reduction in Rta response. Finally, Escherichia coli-derived Rta is an oligomer of 440 kDa in molecular size and binds RtaRE as an oligomer. These results support a model of Rta transactivation wherein the subunits of the Rta oligomer make multiple contacts with a tandem array of phased A/T triplets in the configuration of (A/T)3(G/C)7 repeats.


2001 ◽  
Vol 75 (22) ◽  
pp. 11261-11261
Author(s):  
Paola Rimessi ◽  
Angela Bonaccorsi ◽  
Michael Stürzl ◽  
Marina Fabris ◽  
Egidio Brocca-Cofano ◽  
...  

2001 ◽  
Vol 75 (15) ◽  
pp. 7161-7174 ◽  
Author(s):  
Paola Rimessi ◽  
Angela Bonaccorsi ◽  
Michael Stürzl ◽  
Marina Fabris ◽  
Egidio Brocca-Cofano ◽  
...  

ABSTRACT Human herpesvirus 8 (HHV-8) is found in immunoblastic B cells of patients with multicentric Castleman's disease (MCD) and, predominantly in a latent form, in primary effusion lymphoma (PEL) cells and Kaposi's sarcoma (KS) spindle cells. Recent studies have shown that upon reactivation, HHV-8 expresses factors that downregulate major histocompatibility class I proteins and coactivation molecules and that may enable productively infected cells to escape cytotoxic T lymphocytes and natural killer cell responses. One of these viral factors is encoded by open reading frame (ORF) K3. Here we show that in PEL cells, ORF K3 is expressed through viral transcripts that are induced very early upon virus reactivation, including bicistronic RNA molecules containing coding sequences from viral ORFs K3 and 70. Specifically, we found that a bicistronic transcript was expressed in the absence of de novo protein synthesis, thereby identifying a novel HHV-8 immediate-early gene product. Several features of the RNA molecules encoding the K3 product, including multiple transcriptional start sites, multiple donor splicing sites, and potential alternative ATG usage, suggest that there exists a finely tuned modulation of ORF K3 expression. By contrast, ORF K3 transcripts are not detected in the majority of cells present in KS lesions that are latently infected by the virus, suggesting that there are other, as-yet-unknown mechanisms of immune evasion for infected KS spindle cells. Nevertheless, because HHV-8 viremia precedes the development of KS lesions and is associated with the recrudescence of MCD symptoms, the prompt expression of ORF K3 in productively infected circulating cells may be important for virus pathogenesis. Thus, molecules targeting host or viral factors that activate ORF K3 expression or inactivate the biological functions of the K3 product should be exploited for the prevention or treatment of HHV-8-associated diseases in at-risk individuals.


Sign in / Sign up

Export Citation Format

Share Document