Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
2.
Histopathology ; 53(5): 513-24, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18983461

ABSTRACT

AIMS: Kaposi sarcoma herpesvirus (KSHV) is aetiologically related to Kaposi sarcoma, classical and extracavitary primary effusion lymphoma (PEL; EC-PEL) and multicentric Castleman disease (MCD), entities preferentially occurring in HIV-infected individuals. Characterization of HIV-associated PELs/EC-PELs suggests that the KSHV-infected malignant cells originate from a pre-terminal stage of B-cell differentiation. However, only limited phenotypic studies have been performed on HIV+ MCD, including for PR domain containing 1 with zinc finger domain/B lymphocyte-induced maturation protein 1 (PRDM1/BLIMP1), a key regulator of terminal B-cell differentiation. The aim was to characterize KSHV-infected cells in 17 cases of HIV+ MCD. METHODS AND RESULTS: Double immunohistochemistry and immunohistochemistry-in situ hybridization were used to characterize the KSHV-infected cells in MCD; the results were compared with the phenotypic profiles of 39 PELs/EC-PELs and seven PEL cell lines. Whereas the immunophenotype of KSHV-infected cells in MCD and malignant KSHV+ PEL cells was similar (PAX5, Bcl-6-; PRDM1/BLIMP1, IRF4/MUM1+; Ki67+), the MCD KSHV-infected cells differed, as they expressed OCT2, cytoplasmic lambda immunoglobulin; variably expressed CD27; lacked CD138; and were Epstein-Barr virus negative. CONCLUSIONS: Although both PEL and MCD originate from KSHV-infected pre-terminally differentiated B cells, these findings, with previously reported genetic studies, indicate HIV+ MCD may arise from extrafollicular B cells, whereas PELs may originate from cells that have traversed the germinal centre.


Subject(s)
B-Lymphocytes/virology , Castleman Disease/virology , HIV Infections/complications , Herpesviridae Infections/virology , Herpesvirus 8, Human , Lymphoma, Primary Effusion/virology , Adult , B-Lymphocytes/metabolism , Castleman Disease/immunology , Castleman Disease/metabolism , Cell Differentiation , Female , Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/metabolism , Humans , Immunohistochemistry , Immunophenotyping , In Situ Hybridization , Lymphoma, Primary Effusion/immunology , Male , Middle Aged
3.
Oncogene ; 26(34): 4979-86, 2007 Jul 26.
Article in English | MEDLINE | ID: mdl-17310999

ABSTRACT

Primary effusion lymphoma (PEL) is a rare subtype of non-Hodgkin's lymphoma, which is associated with infection by Kaposi's sarcoma herpesvirus (KSHV)/human herpesvirus-8. The c-Myc transcription factor plays an important role in cellular proliferation, differentiation and apoptosis. Lymphomas frequently have deregulated c-Myc expression owing to chromosomal translocations, amplifications or abnormal stabilization. However, no structural abnormalities were found in the c-myc oncogene in PEL. Given that c-Myc is often involved in lymphomagenesis, we hypothesized that it is deregulated in PEL. We report that PEL cells have abnormally stable c-Myc protein. The turnover of c-Myc protein is stringently regulated by post-transcriptional modifications, including phosphorylation of c-Myc threonine 58 (T58) by glycogen synthase kinase-3beta (GSK-3beta). Our data show that the impaired c-Myc degradation in PEL cells is associated with a significant underphosphorylation of c-Myc T58. The KSHV latency-associated nuclear antigen (LANA) is responsible for this deregulation. Overexpression of LANA in human embryonic kidney 293 or peripheral blood B cells leads to post-transcriptional deregulation of c-Myc protein. Conversely, when LANA is eliminated from PEL cells using RNA interference, GSK-3beta-mediated c-Myc T58 phosphorylation is restored. The presence of c-Myc and LANA in GSK-3beta-containing complexes in PEL cells further confirms the significance of these interactions in naturally KSHV-infected cells.


Subject(s)
Antigens, Viral/metabolism , Lymphoma, Non-Hodgkin/virology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Line , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Lymphoma, Non-Hodgkin/metabolism , Nuclear Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , Threonine/metabolism
4.
Curr Top Microbiol Immunol ; 312: 263-87, 2007.
Article in English | MEDLINE | ID: mdl-17089801

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV), also called human herpesvirus 8 (HHV-8), is associated with a specific subset of lymphoproliferative disorders. These include two main categories. The first is primary effusion lymphomas and related solid variants. The second is multicentric Castleman disease, from which KSHV-positive plasmablastic lymphomas can arise. KSHV contributes to lymphomagenesis by subverting the host cell molecular signaling machinery to deregulate cell growth and survival. KSHV expresses a selected set of genes in the lymphoma cells, encoding viral proteins that play important roles in KSHV lymphomagenesis. Deregulation of the NF-kappaB pathway is an important strategy used by KSHV to promote lymphoma cell survival, and the viral protein vFLIP is essential for this process. Two other viruses that are well documented to be causally associated with lymphoid neoplasia in humans are Epstein-Barr virus (EBV/HHV-4) and human T-cell lymphotropic virus (HTLV-1). Both of these are similar to KSHV in their use of viral proteins to promote cell survival by deregulating the NF-kappaB pathway. Here we review the basic information and recent developments that have contributed to our knowledge of lymphomas caused by KSHV and other viruses. The understanding of the mechanisms of viral lymphomagenesis should lead to the identification of novel therapeutic targets and to the development of rationally designed therapies.


Subject(s)
Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/pathogenicity , Human T-lymphotropic virus 1/pathogenicity , Lymphoma/virology , Animals , Disease Models, Animal , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/isolation & purification , Humans , Lymphoma/etiology , Lymphoma/therapy , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Viral Proteins/physiology , Virus Latency
5.
Blood ; 98(10): 3050-7, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698290

ABSTRACT

Chronic lymphocytic leukemia (CLL) is an incurable leukemia characterized by the slow but progressive accumulation of cells in a CD5+ B-cell clone. Like the nonmalignant counterparts, B-1 cells, CLL cells often express surface immunoglobulin with the capacity to bind autologous structures. Previously there has been no established link between antigen-receptor binding and inhibition of apoptosis in CLL. In this work, using primary CLL cells from untreated patients with this disease, it is demonstrated that engagement of surface IgM elicits a powerful survival program. The response includes inhibition of caspase activity, activation of NF-kappaB, and expression of mcl-1, bcl-2, and bfl-1 in the tumor cells. Blocking phosphatidylinositol 3-kinase (PI3-K), a critical mediator of signals through the antigen receptor, completely abrogated mcl-1 induction and impaired survival in the stimulated cells. These data support the contention that CLL cell survival is promoted by antigen for which the malignant clone has affinity, and suggest that pharmacologic interference with antigen-receptor-derived signals has potential for therapy in patients with CLL.


Subject(s)
B-Lymphocytes/pathology , Immunoglobulin M/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Proteins/physiology , Neoplastic Stem Cells/pathology , Receptors, Antigen, B-Cell/physiology , Aged , Aged, 80 and over , Apoptosis , B-Lymphocytes/immunology , Blotting, Western , CD40 Antigens/physiology , CD5 Antigens/analysis , Cell Survival , Electrophoretic Mobility Shift Assay , Female , Gene Expression Regulation, Leukemic , Humans , Male , Microscopy, Fluorescence , Middle Aged , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein , NF-kappa B/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplastic Stem Cells/immunology , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , bcl-X Protein
6.
Virology ; 287(1): 62-70, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11504542

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV/HHV-8) encodes a G protein-coupled receptor (vGPCR) in open reading frame (ORF) 74, which is homologous to human chemokine receptors. KSHV vGPCR is constitutively active and induces VEGF-mediated angiogenesis. Previous studies have shown that ORF 74 is transcribed as part of a bicistronic message containing ORF K14 upstream of ORF 74, with an early lytic pattern of expression. We have now extended these studies by analyzing three different KSHV-positive primary effusion lymphoma (PEL) cell lines and three PEL clinical samples. In addition, we have identified another less abundant monocistronic transcript containing only ORF 74. Both transcripts were identified at low but similar levels in two PEL clinical samples. We evaluated the degree of sequence and functional conservation of ORF74 in three additional PELs and two KS clinical specimens, demonstrating complete identity at the amino acid level among all isolates. While it is expressed as an early lytic transcript in PEL cell lines, in primary clinical PEL samples transcription of KSHV vGPCR can be readily detected.


Subject(s)
Herpesvirus 8, Human/genetics , Lymphoma/virology , Receptors, Chemokine/biosynthesis , Amino Acid Sequence , Base Sequence , Castleman Disease/genetics , Castleman Disease/virology , Humans , Lymphoma/genetics , Molecular Sequence Data , Open Reading Frames , Receptors, Chemokine/genetics , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Hum Pathol ; 32(3): 288-91, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11274637

ABSTRACT

We studied 15 bone marrow biopsy specimens from patients with human immunodeficiency virus infection for detection of Kaposi sarcoma herpesvirus (KSHV/HHV-8) DNA sequences by a very sensitive and specific polymerase chain reaction (PCR) assay (with 3 different sets of primers). In addition, we used immunohistochemistry with antiviral interleukin-6 (vIL-6) and anti-latent nuclear antigen-1 (LNA-1) antibodies to localize the infected cells on tissue sections. Among the 15 samples, 6 had positive PCR results with the 3 sets of primers (orf26, orf72, orf75). Interestingly, in 2 of these 6 patients (both with Kaposi sarcoma) vIL-6 and LNA-1 were detected in mononuclear lymphoid cells but not in stromal cells of the bone marrow. The detection of vIL-6--positive lymphoid cells in bone marrow suggests a homing for HHV-8--infected elements in this tissue. The local release of vIL-6 may play some role in the plasmacytosis observed in bone marrow in the acquired immunodeficiency syndrome. HUM PATHOL 32:288-291.


Subject(s)
Bone Marrow/virology , HIV Infections/virology , Herpesvirus 8, Human/isolation & purification , Phosphoproteins , Adult , Aged , Biopsy , Bone Marrow/chemistry , DNA, Viral/analysis , Female , Herpesvirus 8, Human/genetics , Humans , Immunohistochemistry , Interleukin-6/analysis , Lymphocytes/chemistry , Lymphocytes/virology , Male , Middle Aged , Nuclear Proteins/analysis , Polymerase Chain Reaction , Sarcoma, Kaposi/virology , Stromal Cells/chemistry , Stromal Cells/virology
8.
J Virol ; 75(5): 2435-43, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11160746

ABSTRACT

Previous studies have shown the presence of Kaposi's sarcoma-associated herpesvirus (KSHV/HHV8) DNA in endothelial cells, in keratinocytes in the basal layer of the epidermis overlying plaque-stage nodular lesions of cutaneous Kaposi's sarcoma (KS), and in the epithelial cells of eccrine glands within KS lesions. We infected primary cell cultures of human keratinocytes with KSHV/HHV8. At 6 days post infection, transcription of viral genes was detected by reverse transcriptase PCR (RT-PCR), and protein expression was documented by an immunofluorescence assay with an anti-LANA monoclonal antibody. To determine whether the viral lytic cycle was inducible by chemical treatment, KSHV/HHV8-infected keratinocytes were treated with 12-O-tetradecanoylphorbol-13-acetate (TPA) and RT-PCR was performed to confirm the transcription of lytic genes such as open reading frame 26, (which encodes a capsid protein). Finally, to assess infectious viral production, other primary human cells (human umbilical vein endothelial cells), were infected with concentrated supernatant of KSHV-infected, TPA-induced keratinocytes and the presence of viral transcripts was confirmed by RT-PCR. The uninfected keratinocytes senesced 3 to 5 weeks after mock infection, while the KSHV/HHV8-infected keratinocytes continued to proliferate and to date are still in culture. However, 8 weeks after infection, viral genomes were no longer detectable by nested PCR. Although the previously KSHV/HHV8-infected keratinocytes still expressed epithelial markers, they acquired new characteristics such as contact inhibition loss, telomerase activity, anchorage-independent growth, and changes in cytokine production. These results show that KSHV/HHV8, like other herpesviruses, can infect and replicate in epithelial cells in vitro and suggest that in vivo these cells may play a significant role in the establishment of KSHV/HHV8 infection and viral transmission.


Subject(s)
Herpesviridae Infections/virology , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/pathogenicity , Keratinocytes/virology , Phosphoproteins , Antigens, Viral/genetics , Antigens, Viral/metabolism , Cells, Cultured , Cytokines/biosynthesis , Endothelial Growth Factors/genetics , Fibroblast Growth Factor 2/genetics , Herpesvirus 8, Human/genetics , Humans , Immunohistochemistry , Keratinocytes/physiology , Lymphokines/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Virus Replication
9.
Hum Pathol ; 32(1): 95-100, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11172301

ABSTRACT

Human herpesvirus-8 (HHV-8) also called Kaposi's sarcoma-associated herpesvirus infects spindle cells in Kaposi's sarcoma (KS) and lymphoid cells in multicentric Castleman's disease (MCD). In KS cells, HHV-8 is mainly latent with the expression of latent nuclear antigen-1 (LNA-1), whereas in MCD both lytic and latent antigens are produced by lymphoid cells. We show by immunohistochemical labeling that in KS viral interleukin-6 (vIL-6) is expressed in rare spindle cells, whereas in MCD, vIL-6 is detectable in lymphoid cells around lymphoid follicles but also within the follicular dendritic reticulum cell network. The staining of apoptotic bodies with anti IL-6 antibody suggests the achievement of a complete lytic cycle in a subset of lymphoid cells. Interestingly, in MCD, some areas contained vascular spindle cells latently infected by HHV-8 on the basis of LNA-1 expression. This finding might imply that in MCD, both vascular and lymphoid cells proliferate in response to the viral infection. Double immunostaining with anti LNA-1 and anti vIL-6 in MCD and KS identifies 2 subsets of HHV-8 infected (vascular and lymphoid) cells, some with exclusive expression of LNA-1 and some with coexpression of vIL-6 and LNA-1. This suggests that in vivo the regulation of the expression vIL-6 and LNA-1 protein varies with the cell type. In addition, the detection of infected endothelial cells in MCD may indicate that these cells belong to the reservoir for HHV-8.


Subject(s)
Castleman Disease/pathology , Herpesvirus 8, Human , Interleukin-6/analysis , Nuclear Proteins/analysis , Phosphoproteins , Sarcoma, Kaposi/pathology , Apoptosis , Castleman Disease/metabolism , Castleman Disease/virology , DNA, Viral/genetics , Endothelium, Vascular/chemistry , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Herpesvirus 8, Human/chemistry , Herpesvirus 8, Human/genetics , Humans , Immunohistochemistry , Lymphocytes/chemistry , Lymphocytes/pathology , Lymphocytes/virology , Polymerase Chain Reaction , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/virology
10.
J Mol Diagn ; 3(1): 32-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11227070

ABSTRACT

Since its discovery in 1994, KSHV (also called human herpesvirus-8 or HHV8) has been implicated in a variety of disorders. Although the association of KSHV with Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease has been well established, its presence in some other diseases, such as multiple myeloma, remains controversial. Because most KSHV studies are based on polymerase chain reaction (PCR) analysis, the conflicting data may be attributable to variations in the methods, primer sets, and target sequences selected. To establish an efficient and reliable PCR approach for KSHV detection we designed eight sets of primers to six regions (ORFK1, ORFK2, ORFK9, ORK26, ORF72, and ORF74) of the KSHV genome using appropriate database and software. The detection sensitivity of these primers was carefully assessed and their reliability was strictly validated in a series of positive (15 KS and PEL samples) and negative (16 lymphoid tissues) controls. We found that primer sets to the ORFK9 region showed the highest sensitivity, whereas primer sets to ORFK1 and ORF74 showed the lowest sensitivity. Primer sets to ORFK9, ORF26 and ORF72 regions detected all of the positive cases, whereas other primer sets showed varying detection rates or nonspecific bands. All 16 negative controls were negative with all primer sets. However, six of 16 negative controls became positive when we used nested PCR targeting ORF26. Therefore, multiple target KSHV sequences increase the detection efficiency, while nested PCR protocols are likely to introduce false positivity. Using ORFK9, ORF26 and ORF72 primer sets, we screened bone marrow biopsies from 18 cases of multiple myeloma, and failed to detect any KSHV sequences. This finding supports the conclusion that KSHV is not associated with multiple myeloma. Indeed, our results further confirm that although KSHV is universally present in Kaposi's sarcoma and primary effusion lymphoma, it is not ubiquitious.


Subject(s)
Herpesvirus 8, Human/isolation & purification , Multiple Myeloma/virology , Polymerase Chain Reaction/methods , Sarcoma, Kaposi/virology , Archives , DNA Primers , Herpesvirus 8, Human/genetics , Humans , Lymphoma/virology , Sensitivity and Specificity , Tissue Banks
11.
Blood ; 97(1): 270-6, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11133771

ABSTRACT

The organization and expression of the BCL-6 gene in normal and neoplastic thymic T cells has not been fully determined. We examined 8 precursor T-cell lymphoblastic lymphomas (T-LBLs) and found significant BCL-6 expression in 4 cases. Three of the BCL-6(+) cases expressed a common thymocyte phenotype (CD4(+), CD8(+)), and one expressed a precursor thymocyte phenotype (CD4(-), CD8(-)). In 6 cases evaluated, including those expressing BCL-6, molecular analyses demonstrated a germline configuration of the BCL-6 gene and a wild-type BCL-6 gene first exon-intron boundary region. We also evaluated 12 normal prenatal and postnatal thymuses for BCL-6 protein. BCL-6 was expressed by most cortical thymocytes and by scattered medullary thymocytes. BCL-6(+) cortical and medullary thymocytes also expressed CD2, CD3, CD4, CD5, CD7, or CD8. We further analyzed the pattern of BCL-2 and BCL-X(L) expression and their coexpression with BCL-6 in normal thymus and T-LBL and compared it to that of follicle centers of reactive lymph nodes and follicular lymphoma. BCL-6(+) cortical thymocytes coexpressed BCL-X(L) but not BCL-2. All 4 BCL-6(+) T-LBLs and 4 BCL-6(-) T-LBLs coexpressed BCL-2 and BCL-X(L). Conceivably, T-LBLs may arise through clonal expansion of cortical thymocytes normally expressing the BCL-6 protein. The pattern of BCL-6, BCL-2, and BCL-X(L) expression in cortical thymocytes is highly reminiscent of germinal centers, and the abnormal coexpression of BCL-2, BCL-X(L), and BCL-6 in T-LBL is analogous to coexpression in follicle center cell lymphomas, suggesting that coexpression of these anti-apoptotic genes may contribute to the pathogenesis of T-LBL.


Subject(s)
DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Thymus Gland/embryology , Transcription Factors/metabolism , Adolescent , Adult , Apoptosis/drug effects , Child , Child, Preschool , Female , Fetus/chemistry , Fetus/cytology , Humans , Immunophenotyping , Infant , Infant, Newborn , Lymphoma, Follicular/etiology , Lymphoma, Follicular/metabolism , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-6 , Thymus Gland/chemistry , Thymus Gland/pathology , bcl-X Protein
12.
Methods Mol Med ; 49: 331-6, 2001.
Article in English | MEDLINE | ID: mdl-21370151

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV), also called human herpesvirus-8 (HHV-8), is the most recently identified human herpesvirus (1). It has been found to be invariably present in Kaposi's sarcoma (KS) lesions, whether these are associated with AIDS (epidemic KS), therapeutic immunosuppression (iatrogenic KS), or high-incidence regions in Africa (endemic KS), or in its "classic" form (sporadic KS) (for reviews see refs. 2 and 3). By contrast, with few reported exceptions, it has not been found to be present in a variety of other vascular tumors and reactive conditions. A seroepidemiologic association of this virus and KS has been well documented, and it is currently accepted that KSHV plays a necessary, although not sufficient, role in the development of KS. Although diagnosis of KS is usually not difficult based on clinical and histologic features, some cases may have unusual morphology, with features overlapping those of other vascular and spindle cells proliferations. In these instances, molecular detection is useful to confirm or rule out a diagnosis of KS.

13.
Blood ; 96(7): 2537-42, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11001908

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV), or human herpervirus 8 (HHV-8), is a gamma-herpesvirus that infects human lymphocytes and is associated with primary effusion lymphoma (PEL). Currently, the role of viral infection in the transformation of PEL cells is unknown. One possibility is that KSHV, like the lymphotropic viruses Epstein-Barr virus (EBV) and human T-cell leukemia virus I (HTLV-I), activates the transcription factor NF-kappaB to promote survival and proliferation of infected lymphocytes. To examine this possibility, we assessed NF-kappaB activity in KSHV-infected PEL cell lines and primary tumor specimens by electrophoretic mobility shift assay (EMSA). We observed that NF-kappaB is constitutively activated in all KSHV-infected lymphomas, and consists of 2 predominant complexes, p65/p50 heterodimers and p50/p50 homodimers. Inhibition experiments demonstrated that Bay 11-7082, an irreversible inhibitor of IkappaBalpha phosphorylation, completely and specifically abrogated the NF-kappaB/DNA binding in PEL cells. PEL cells treated with Bay 11 demonstrated down-regulation of the NF-kappaB inducible cytokine interleukin 6 (IL-6), and apoptosis. These results suggest that NF-kappaB activity is necessary for survival of KSHV-infected lymphoma cells, and that pharmacologic inhibition of NF-kappaB may be an effective treatment for PEL.


Subject(s)
Apoptosis , Herpesvirus 8, Human , Lymphoma/pathology , Lymphoma/virology , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Apoptosis/drug effects , Aspirin/pharmacology , Cell Survival , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Kaposi , Sodium Salicylate/pharmacology , Tumor Cells, Cultured
14.
Semin Oncol ; 27(4): 409-19, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10950367

ABSTRACT

Kaposi's sarcoma-associated herpes virus (KSHV), also known as human herpes virus-8 (HHV-8), is a recently described gamma-herpes virus that has been etiologically linked to two different acquired immunodeficiency syndrome (AIDS)-related malignancies by strong epidemiologic and pathologic evidence. Infection been shown to precede and predict the development of Kapasi's sarcoma (KS) in human immunodeficiency virus (HIV)-infected patients, and viral DNA has been found in KS lesions of all types and stages. Furthermore, KSHV is a lymphotropic virus and is present in nearly all cases of primary effusion lymphoma, a rare malignancy disproportionately affecting HIV-infected individuals. KSHV is also thought to dramatically affect the incidence, type, and course of multicentric Castleman's disease, another lymphoproliferative disorder over-represented in people with AIDS. KSHV encodes many potentially oncogenic products, including several apparently pirated from the human genome. These include various chemokines, cell cycle regulatory proteins, and survival and proliferation factors. Knowledge is rapidly accumulating concerning the viral pathogenic mechanisms and host cofactors necessary for KSHV-mediated disease.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Herpesviridae Infections/complications , Herpesvirus 8, Human/physiology , Lymphoma, AIDS-Related/virology , Sarcoma, Kaposi/complications , Acquired Immunodeficiency Syndrome/virology , Castleman Disease/complications , Castleman Disease/virology , Herpesvirus 8, Human/pathogenicity , Humans , Sarcoma, Kaposi/etiology
15.
Cancer ; 89(2): 445-52, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10918178

ABSTRACT

BACKGROUND: Studies have shown telomerase activity to be present in some B-cell non-Hodgkin lymphomas (B-NHLs). However, no large studies have assayed telomerase activity in a systematic and quantitative manner. Furthermore, the relation between telomerase and proliferation suggested by in vitro studies has not been adequately tested in B-NHLs in vivo. This information is necessary to understand the relation between proliferation and telomerase and to predict the efficacy of antitelomerase drugs currently in development. METHODS: Eighteen benign biopsies and 111 B-NHLs of varying types were classified according to the revised European-American classification of lymphoid neoplasms (REAL classification) and assayed for telomerase activity and proliferation index (PI). RESULTS: All B-NHLs contained telomerase activity except for low grade marginal zone B-cell lymphomas (MZBCLs) (96 of 111, 86%) (chi(2) 95.90, P < 0.001). Telomerase activity correlated with PI (r = 0.7536, r(2) = 0.5678, t = 10.51, P < 0.001) and showed a threshold whereby telomerase activity was not present below a PI of 9.2% (t = 4.875, P < 0.001). CONCLUSIONS: The level of telomerase activity fell within characteristic ranges and generally correlated with the clinical aggressiveness of each B-NHL category. Low grade MZBCLs of extranodal, nodal, and splenic types were unique among the categories of B-NHL in lacking or containing very little telomerase activity. The association between telomerase activity and PI is evidence that telomerase is controlled in vivo along with the cell cycle and is not constitutively active in B-NHL. These data provide evidence that antitelomerase drugs may be efficacious in most types of B-NHL.


Subject(s)
Lymphoma, B-Cell/enzymology , Telomerase/metabolism , Cell Division/physiology , Humans , Lymphoma, B-Cell/classification , Lymphoma, B-Cell/pathology
16.
Arch Pathol Lab Med ; 124(2): 304-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10656746

ABSTRACT

We report a case of aggressive natural killer cell lymphoma presenting as an anterior mediastinal mass in an African-American man with acquired immunodeficiency syndrome. Histologically, the anterior mediastinal mass showed a diffuse dense infiltrate of atypical intermediate-sized and large lymphoid cells, as well as scattered immunoblasts with angiocentric and angiodestructive growth and extensive zonal necrosis. Similar lymphoid infiltrates were present in the patient's lungs, spleen, and bone marrow, accompanied by extensive lymphophagocytosis. Electron microscopic and cytologic examinations showed the presence of dense cytoplasmic granules. Immunophenotyping by flow cytometry and by immunohistochemistry yielded surface markers consistent with a natural killer cell lymphoma. The Epstein-Barr virus genome and monoclonality were detected by in situ hybridization and Southern blot analysis. Polymerase chain reaction confirmed the presence of type A Epstein-Barr virus. T-cell receptor gene rearrangement could not be identified by Southern blot analysis or polymerase chain reaction. To the best of our knowledge, this is the first reported case of designated natural killer cell lymphoma from the mediastinum, as well as the first reported case of natural killer cell lymphoma in a patient with acquired immunodeficiency syndrome. This tumor disseminated early and pursued a highly aggressive course. Epstein-Barr virus may play a role in the pathogenesis of this disease.


Subject(s)
Killer Cells, Natural/pathology , Lymphoma, AIDS-Related/diagnosis , Mediastinal Neoplasms/diagnosis , Adult , Antigens, Neoplasm/analysis , Antigens, Surface/analysis , Blotting, Southern , Cell Separation , Cytoplasmic Granules/ultrastructure , DNA, Viral/analysis , Diagnosis, Differential , Fatal Outcome , Flow Cytometry , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Humans , Immunocompromised Host , In Situ Hybridization , Lymphoma, AIDS-Related/virology , Male , Mediastinal Neoplasms/virology , Polymerase Chain Reaction
18.
Curr Opin Oncol ; 11(5): 322-32, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10505767

ABSTRACT

Epstein-Barr virus, Kaposi's sarcoma-associated herpesvirus/human herpesvirus 8, and human T-cell lymphotrophic virus are viruses that are implicated in lymphoid neoplasia in humans. Their association with specific subsets of lymphomas suggests that they play an important, although not sufficient, etiologic role in their development. Current knowledge suggests that these viruses contribute to lymphomagenesis by subverting the host-cell molecular machinery to deregulate cell growth and survival. In this article, the basic information and recent developments that have contributed to our understanding of viral lymphomagenesis are reviewed.


Subject(s)
Gammaherpesvirinae , Human T-lymphotropic virus 1 , Lymphoma/virology , Herpesvirus 4, Human , Herpesvirus 8, Human , Humans
19.
J Infect Dis ; 180(3): 824-8, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10438372

ABSTRACT

In order to characterize the expression of the viral interleukin-6 (vIL-6) homologue in various human herpesvirus 8 (HHV-8)-associated diseases, in situ hybridization and immunohistochemistry were applied to formalin-fixed specimens. These assays showed consistent expression of vIL-6 in primary effusion lymphomas and in a case of human immunodeficiency virus (HIV)-associated lymphadenopathy with a Castleman's disease-like appearance. In contrast, Kaposi's sarcoma specimens showed marked differences among specimens. In a consecutive series of specimens from the Johns Hopkins archives, vIL-6 expression was demonstrated in one of 13 cases. However, among 7 specimens selected from the AIDS Malignancy Bank because of their high levels of the T1.1 lytic transcript and virion production, vIL-6 expression was consistently demonstrated in infiltrating mononuclear cells and occasional spindle-shaped cells. Thus vIL-6 expression in clinical specimens correlates with other measures of the lytic viral cycle. Both assays generally give congruent results and are consistent with the possibility that vIL-6 expression plays a role in the pathogenesis of a variety of HHV-8-associated diseases.


Subject(s)
Herpesvirus 8, Human/immunology , Interleukin-6/genetics , Lymphoma, AIDS-Related/virology , Sarcoma, Kaposi/virology , Viral Proteins/genetics , Adult , Aged , DNA, Viral/analysis , Female , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/isolation & purification , Humans , Immunohistochemistry , In Situ Hybridization , Interleukin-6/analysis , Lymphoma , Lymphoma, AIDS-Related/immunology , Lymphoma, AIDS-Related/pathology , Male , Middle Aged , Open Reading Frames , Retrospective Studies , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/pathology , Tumor Cells, Cultured , Viral Proteins/analysis
20.
Hum Pathol ; 30(8): 988-92, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10452515

ABSTRACT

Although follicle center cell lymphoma and mantle cell lymphoma are both B cell non-Hodgkin's lymphomas (NHL), they are regarded as separate entities with distinct clinical, morphological, immunophenotypic and molecular characteristics. To our knowledge, the coexistence of these 2 lymphomas in the same patient has never been reported. We describe a 70-year-old woman with a long-standing history of follicle center cell lymphoma, cytological grade I, who subsequently developed a composite lymphoma consisting of well-demarcated foci of persistent follicle center cell lymphoma surrounded by mantle cell lymphoma. This morphological interpretation was supported by the presence of both bcl-1 and bcl-2 gene rearrangements, which are molecular genetic hallmarks of mantle cell lymphoma and follicle center cell lymphoma, respectively. Polymerase chain reaction (PCR) analysis for rearranged immunoglobulin heavy chain (IgH) genes showed a dominant band identical in size in microdissected tumor cells of the follicle center cell and mantle cell lymphomas. Cloning and sequence analysis of the PCR products revealed a common clone-specific IgH gene rearrangement in these 2 lymphomas. These findings suggest that this composite lymphoma represents the unusual evolution of a malignant B-cell clone that resulted in the development of 2 morphologically distinct but clonally related B-cell NHLs. These findings also show the importance of integrating morphological, immunophenotypic, and molecular data to enhance our understanding of the complex pathogenic interrelationships in lymphomagenesis.


Subject(s)
Lymphoma, Follicular/pathology , Lymphoma, Non-Hodgkin/pathology , Aged , Antigens, CD/metabolism , Blotting, Southern , Female , Humans , Immunophenotyping , Lymphoma, Follicular/genetics , Lymphoma, Follicular/metabolism , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...