151
|
Zheng C, Huang DR, Bergenbrant S, Sundblad A, Osterborg A, Björkholm M, Holm G, Yi Q. Interleukin 6, tumour necrosis factor alpha, interleukin 1beta and interleukin 1 receptor antagonist promoter or coding gene polymorphisms in multiple myeloma. Br J Haematol 2000; 109:39-45. [PMID: 10848780 DOI: 10.1046/j.1365-2141.2000.01963.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Proinflammatory cytokines such as interleukin 6 (IL-6), tumour necrosis factor alpha (TNF-alpha) and IL-1beta are considered to be involved in the pathogenesis of multiple myeloma (MM). In the present study, we examined a G/C polymorphism at position -174 in the promoter region of IL-6, a biallelic polymorphism at position -308 in the promoter region of TNF-alpha, the TaqI restriction fragment length polymorphism in exon 5 of IL-1beta and a variable number of identical tandem repeat polymorphisms in intron 2 of IL-1 receptor antagonist (IL-1Ra) genes. The alleles of these loci are known to influence the level of production of the cytokines and the IL-1Ra. Seventy-three patients with MM, 27 with monoclonal gammopathy of undetermined significance (MGUS) and 129 healthy individuals were included. No difference was found between patients and healthy controls or between MM and MGUS patients in the distributions of genotypes and frequencies of alleles of the IL-6 (-174), TNF-alpha (-308), IL-1beta TaqI and IL-1Ra gene polymorphisms. No associations between the polymorphisms at the loci under study and clinical factors such as age, sex, clinical stage at onset and M-protein type were observed. Our results indicate that the cytokine (IL-6, TNF-alpha and IL-1beta) and IL-Ra gene polymorphisms do not confer susceptibility to the development of MM.
Collapse
Affiliation(s)
- C Zheng
- Division of Haematology, and Immunological Research Laboratory, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
152
|
Minami R, Muta K, Ilseung C, Abe Y, Nishimura J, Nawata H. Interleukin-6 sensitizes multiple myeloma cell lines for apoptosis induced by interferon-alpha. Exp Hematol 2000; 28:244-55. [PMID: 10720689 DOI: 10.1016/s0301-472x(99)00156-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Interleukin-6 (IL-6) is a multifunctional cytokine affecting growth and survival of normal B cell lineage and multiple myeloma cells. To test the hypothesis that IL-6, as well as other hematopoietic growth factors, may enhance apoptosis of target cells, we investigated the effect of IL-6 on myeloma cells in the presence of IFN-alpha, which is prescribed for patients with multiple myeloma. MATERIALS AND METHODS Four myeloma cell lines, PCM6, NOP-2, U266, RPMI8226 were tested. We determined the induction of apoptosis by flow cytometry, using an FITC-Annexin V. RESULTS IFN-alpha induced apoptosis on myeloma cell lines, and this apoptosis was further enhanced in the presence of IL-6, via activation of caspase 3. During induction of this apoptosis, the expression of c-Myc and Fas increased. The addition of IL-6 further increased the expression of Fas, but not that of c-Myc. Bcl-2, Bcl-x, and p53 were not affected by the addition of IL-6 and/or IFN-alpha. Addition of a PI-3-K inhibitor interfered with the enhancing effect of IL-6 on the apoptosis induced by IFN-alpha. CONCLUSION We propose that IL-6 has the death signal, as well as growth promoting effects, and that PI-3-K may play a key role in the induction of apoptosis by IL-6.
Collapse
Affiliation(s)
- R Minami
- Third Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | |
Collapse
|
153
|
Karadag A, Oyajobi BO, Apperley JF, Russell RG, Croucher PI. Human myeloma cells promote the production of interleukin 6 by primary human osteoblasts. Br J Haematol 2000; 108:383-90. [PMID: 10691869 DOI: 10.1046/j.1365-2141.2000.01845.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Interleukin-6 (IL-6) is an important growth and survival factor for myeloma cells. However, the identity of the cells producing IL-6 in vivo remains unclear. Myeloma cells are found closely associated with sites of active bone turnover, and cells of the osteogenic lineage, including bone marrow osteoprogenitors, osteoblasts and bone lining cells, may therefore be ideally placed to synthesize IL-6. We have examined the possibility that human osteogenic cells may produce IL-6 in response to stimulation by myeloma cells. Primary human osteoblasts (hOBs) were isolated from normal donors, co-cultured with the human myeloma cell lines, JJN-3, RPMI-8226 and NCI-H929, and the amount of IL-6 released was determined by enzyme-linked immunosorbent assay (ELISA). All myeloma cells stimulated a significant increase in the production of IL-6 when cultured with hOBs (P < 0.05). Prior fixation of hOBs completely abrogated release of IL-6 in the co-cultures. In contrast, fixed myeloma cells retained the ability to induce IL-6 production, suggesting that hOBs were the principal source of IL-6. Physical separation of myeloma cells from hOBs using transwell inserts caused a partial inhibition of IL-6 release (P < 0.05), whereas the addition of media conditioned by myeloma cells to cultures of hOBs stimulated a significant increase in IL-6 production (P < 0.05). hOBs secreted greater amounts of IL-6 than human bone marrow stromal cells (hBMSCs) (2.2- to 3.5-fold, P < 0.05), but incubating hBMSCs with dexamethasone to stimulate osteoblastic differentiation resulted in an increase in their ability to produce IL-6 (1.7- to 4. 8-fold, P < 0.05) and to respond to myeloma cells (P < 0.05). These data clearly indicate that cells of the osteoblast lineage release significant amounts of IL-6 in response to stimulation by myeloma cells and may contribute to the IL-6 that promotes the proliferation and survival of myeloma cells in vivo.
Collapse
Affiliation(s)
- A Karadag
- Division of Biochemical and Musculoskeletal Medicine, University of Sheffield Medical School, Sheffield, UK
| | | | | | | | | |
Collapse
|
154
|
Hayashida K, Shimaoka Y, Ochi T, Lipsky PE. Rheumatoid arthritis synovial stromal cells inhibit apoptosis and up-regulate Bcl-xL expression by B cells in a CD49/CD29-CD106-dependent mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:1110-6. [PMID: 10623863 DOI: 10.4049/jimmunol.164.2.1110] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inflammatory sites, such as rheumatoid arthritis (RA) synovial tissue, contain large numbers of activated B cells and plasma cells. However, the mechanisms maintaining B cell viability and promoting their differentiation are not known, but interactions with stromal cells may play a role. To examine this, purified human peripheral B cells were cultured with a stromal cell line (SCL) derived from RA synovial tissue, and the effects on apoptosis and expression of Bcl-2-related proteins were analyzed. As a control, B cells were also cultured with SCL from osteoarthritis synovium or skin fibroblasts. B cells cultured with medium alone underwent spontaneous apoptosis. However, B cells cultured with RA SCL cells exhibited less apoptosis and greater viability. Although SCL from osteoarthritis synovium and skin fibroblasts also rescued B cells from apoptosis, they were less effective than RA SCL. B cell expression of Bcl-xL was markedly increased by RA SCL in a contact-dependent manner, whereas B cell expression of Bcl-2 was unaffected. Protection of B cells from apoptosis and up-regulation of Bcl-xL by RA SCL were both blocked by mAbs to CD106 (VCAM-1), but not CD54 (ICAM-1). Furthermore, cross-linking of CD49d/CD29 (very late Ag-4) on the surface of B cells rescued them from apoptosis and up-regulated Bcl-xL expression. These results indicate that SCL derived from RA synovial tissue play a role in promoting B cell survival by inducing Bcl-xL expression and blocking B cell apoptosis in a CD49d/CD29-CD106-dependent manner.
Collapse
Affiliation(s)
- K Hayashida
- University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | | | | | | |
Collapse
|
155
|
Expression of functional interleukin-15 receptor and autocrine production of interleukin-15 as mechanisms of tumor propagation in multiple myeloma. Blood 2000. [DOI: 10.1182/blood.v95.2.610] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin-15 (IL-15) induces proliferation and promotes cell survival of human T and B lymphocytes, natural killer cells, and neutrophils. Here we report the constitutive expression of a functional IL-15 receptor (IL-15R) in 6 of 6 myeloma cell lines and in CD38high/CD45low plasma cells belonging to 14 of 14 patients with multiple myeloma. Furthermore, we detected IL-15 transcripts in all 6 myeloma cell lines, and IL-15 protein in 4/6 cell lines and also in the primary plasma cells of 8/14 multiple myeloma patients. Our observations confirm the existence of an autocrine IL-15 loop and point to the potential paracrine stimulation of myeloma cells by IL-15 released from the cellular microenvironment. Blocking autocrine IL-15 in cell lines increased the rate of spontaneous apoptosis, and the degree of this effect was comparable to the pro-apoptotic effect of depleting autocrine IL-6 by antibody targeting. IL-15 was also capable of substituting for autocrine IL-6 in order to promote cell survival and vice versa. In short-term cultures of primary myeloma cells, the addition of IL-15 reduced the percentage of tumor cells spontaneously undergoing apoptosis. Furthermore, IL-15 lowered the responsiveness to Fas-induced apoptosis and to cytotoxic treatment with vincristine and doxorubicin but not with dexamethasone. These data add IL-15 to the list of important factors promoting survival of multiple myeloma cells and demonstrate that it can be produced and be functionally active in an autocrine manner.
Collapse
|
156
|
Abstract
Mcl-1 is an anti-apoptotic member of the Bcl-2 family which is tightly regulated during myeloid and B cell differentiation. We have recently reported that Mcl-1 is expressed in human myeloma cells and that Mcl-1 and Bcl-x(L) expression are correlated. In the current study, we demonstrate that IL-6, a survival factor for the human myeloma cell line MDN, rapidly up-regulates Mcl-1 whereas it has no effect on Bcl-2 protein level. In MDN cells, IL-6 induces both extracellular signal-regulated protein kinase (ERK)1,2 and STAT3 activation whereas STAT1 and STAT5 activation remains undetectable. Furthermore, while investigating the IL-6 signaling pathway leading to Mcl-1 up-regulation, we show that a janus kinase (JAK)-2 inhibitor is able to inhibit both STAT3 activation and Mcl-1 up-regulation whereas an MAP/ERK kinase (MEK) inhibitor has no effect. In conclusion, our data suggest the involvement of the JAK / STAT pathway but not of the Ras / mitogen-activated protein (MAP) kinase pathway in IL-6-induced Mcl-1 up-regulation.
Collapse
Affiliation(s)
- D Puthier
- Inserm U463, Institut de biologie, Nantes, France
| | | | | |
Collapse
|
157
|
Abstract
By necessity, this article focuses on only a handful of molecules with demonstrated ability to affect growth of myeloma cells. The heterogeneity in growth-factor responsiveness has made formulation of a uniform hypothesis a daunting challenge. One trait that appears to be consistent among all myeloma patients is the uncoupling of the normally highly integrated relationship between terminal differentiation and loss of growth potential. Thus, the common feature in myeloma may not be the precise cytokines or cell-to-cell interactions that drive tumor-cell growth, but rather the underlying genetic traits that afford the tumor cell the ability to proliferate despite its relatively advanced stage of differentiation. The success of current strategies used to treat myeloma patients has thus far been somewhat limited, and in general, has only modestly prolonged survival. It is clear that successful treatment of this disease will require the development of new therapeutic agents aimed at the biochemical events that sustain the aberrant growth of the tumor cells. The knowledge of cell signaling, gene transcription, and cell growth and differentiation has expanded rapidly, and this information has provided a greater understanding of the cell biology of a variety of malignancies. Application of this information to the study of multiple myeloma, however, has thus far been relatively limited, primarily because the heterogeneity of the disease and the lack of appropriate model systems. Review of the literature, particularly over the last 5 years, reveals a significant number of exciting new findings in this field and the development of new model systems that are certain to yield greater insight into this devastating disease.
Collapse
Affiliation(s)
- D F Jelinek
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
158
|
Sjak-Shie NN, Vescio RA, Berenson JR. The role of human herpesvirus-8 in the pathogenesis of multiple myeloma. Hematol Oncol Clin North Am 1999; 13:1159-67. [PMID: 10626142 DOI: 10.1016/s0889-8588(05)70118-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human herpesvirus-8 has been strongly implicated in the pathogenesis of KS, BCBL, and multicentric Castleman's disease. Evidence for its role in the pathogenesis of multiple myeloma is accumulating. Human herpesvirus-8 is detectable in the nonmalignant bone marrow dendritic cells from most myeloma patients. In addition, HHV-8 is also detected in the peripheral blood of most myeloma patients. In contrast, this virus is rarely detected in close contacts of myeloma patients, healthy individuals, or patients with other malignancies. Furthermore, only about one fourth of patients with MGUS are infected with HHV-8. Sequencing of HHV-8 DNA isolated from myeloma patients shows both minor differences among patients and a conserved deletion unique to myeloma compared with HHV-8 in other malignancies. Consistent expression of both the viral homologues of IRF and IL-8R in myeloma suggests a possible role for these transforming viral genes in the pathogenesis of this disease. Although the described association between multiple myeloma and HHV-8 implies a causal role in the pathogenesis of this disease, no cause-and-effect relationship is yet demonstrated. Evidence may be obtained directly by fulfilling Koch's postulate in an animal model and indirectly through therapeutic interventions with antiviral agents or through extensive epidemiological studies. Such epidemiological studies would be greatly facilitated by the development of antibodies directed against the HHV-8 viral proteins uniquely present in myeloma. A direct or indirect causal effect of HHV-8 has potentially enormous implications for the therapeutic benefit of antiviral agents and preventative strategies using vaccines. There is, indeed, preliminary evidence that antiviral therapy in HIV-infected patients reduces the risk or development of KS. Clinical improvement in patients with KS treated with antiviral agents has also been reported. These observations suggest that future treatment strategies to combat multiple myeloma may include antiviral agents.
Collapse
Affiliation(s)
- N N Sjak-Shie
- Division of Hematology and Oncology, University of California School of Medicine, Los Angeles, USA
| | | | | |
Collapse
|
159
|
Oskeritzian CA, Wang Z, Kochan JP, Grimes M, Du Z, Chang HW, Grant S, Schwartz LB. Recombinant Human (rh)IL-4-Mediated Apoptosis and Recombinant Human IL-6-Mediated Protection of Recombinant Human Stem Cell Factor-Dependent Human Mast Cells Derived from Cord Blood Mononuclear Cell Progenitors. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.9.5105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Although stem cell factor (SCF) appears to be the major growth factor for human mast cells, other factors undoubtedly play important roles in the development, survival, and function of these cells. The current study examined the effects of recombinant human (rh) IL-4 and rhIL-6 on rhSCF-dependent development and survival of human mast cells derived in vitro from cord blood progenitor cells. After 4–8 wk of culture with rhSCF and various amounts of rhIL-4, a dramatic decline in mast cell numbers was observed with rhIL-4, the EC50 being about 0.1 ng/ml. Numbers of other cell types remained high. Mast cells derived from cord blood progenitors after 7 wk of culture with rhSCF alone displayed an MCT phenotype and expressed Kit, FcεRI, and IL-4R on their surface. Mast cells examined after purification by immunomagnetic sorting became apoptotic within hours after exposure to rhIL-4, a phenomenon blocked by anti-IL-4 Ab. Because rhIL-4-dependent apoptosis but not the loss of mitochondrial membrane potential was prevented by the pan-caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp-(Z-VAD)-fluoromethylketone, mitochondrial perturbation most likely preceded caspase activation. Consistent with this conclusion was the observation that both apoptosis and loss of mitochondrial membrane potential (Δψm) were inhibited by cyclosporin A in combination with aristolochic acid. rhIL-6 protected cord blood mast cells from rhIL-4-induced apoptosis. Thus, IL-4 can cause both maturation and apoptosis of human mast cells, the latter effect being abrogated by IL-6.
Collapse
Affiliation(s)
| | | | - Jarema P. Kochan
- ‡Department of Metabolic Diseases, Hoffman-LaRoche Inc., Nutley, NJ 07110
| | - Margaret Grimes
- †Pathology, Virginia Commonwealth University, Richmond, VA 23298; and
| | | | | | | | | |
Collapse
|
160
|
Matsushima AY, Strauchen JA, Lee G, Scigliano E, Hale EE, Weisse MT, Burstein D, Kamel O, Moore PS, Chang Y. Posttransplantation plasmacytic proliferations related to Kaposi's sarcoma-associated herpesvirus. Am J Surg Pathol 1999; 23:1393-400. [PMID: 10555008 DOI: 10.1097/00000478-199911000-00010] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV), which was originally detected in Kaposi's sarcoma, also has been found in primary effusion lymphomas (PELs) and some cases of multicentric Castleman's disease. We describe two transplant recipients who developed Kaposi's sarcoma and a spectrum of non-neoplastic lymphoproliferative disorders that show pronounced plasmacytic and plasmacytoid features. The first patient had recurrent pleural effusions and Castleman's disease-like changes in lymph nodes. The second patient had systemic lymphadenopathy and hepatosplenomegaly secondary to diffuse infiltration by polyclonal plasma cells and plasmacytoid B lymphocytes that clinically mimicked Epstein-Barr virus (EBV)-associated posttransplant lymphoproliferative disease. In both cases, KSHV DNA was detected by polymerase chain reaction and Southern blotting, and KSHV vIL-6 protein expression was identified in affected tissues by immunohistochemical localization. In contrast, no evidence of KSHV coinfection was detected in any of 31 EBV-related posttransplant lymphoproliferative disorders or 112 non-PEL lymphomas tested. The pathologic findings in these two patients were not representative of malignancy by morphologic, immunophenotypic, or molecular criteria. This study underscores the marked propensity for hematolymphoid proliferations associated with KSHV infections to show plasmacytic features. Additionally, this study describes use of an antibody reactive against KSHV vIL-6 that can readily detect a subpopulation of KSHV-infected hematopoietic cells.
Collapse
Affiliation(s)
- A Y Matsushima
- Department of Pathology, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Dalton WS. The tumor microenvironment as a determinant of drug response and resistance. Drug Resist Updat 1999; 2:285-288. [PMID: 11504502 DOI: 10.1054/drup.1999.0097] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Classically, studies of drug resistance in cancer have focused on the molecular biology of single cancer cells. These types of studies have provided important information regarding certain drug resistance mechanisms, including mechanisms that reduce intracellular drug accumulation, alter or repair drug-induced damage, and reduce drug-induced apoptosis. While these cellular mechanisms undoubtedly contribute to the overall phenomenon of drug resistance, it is now evident that the tumor cell microenvironment also influences how a tumor cell behaves and responds to cytotoxic drugs or radiation. Two different forms of tumor cell-environmental interaction may explain how some tumor cells survive initial drug exposure and eventually express classical mechanisms of drug resistance. The first form involves soluble mediators, such as interleukins, that are secreted by non-tumor, stromal cells. Interleukin-6 (IL-6) is a classical example of how a soluble mediator secreted by the tumor microenvironment is capable of enhancing tumor cell survival and perhaps blocking apoptosis. The second form of tumor cell-environment interaction requires direct cell contact and has been given the term cell-adhesion-mediated drug resistance (CAM-DR). In this case, binding extracellular matrix ligands in the tumor microenvironment may activate cell adhesion molecules, such as the integrins, and these interactions result in the activation of signal transduction pathways that block drug-induced apoptosis. Interrupting the tumor cell-environment interactions or the associated signal transduction pathways may represent a new approach for the treatment of cancer. Copyright 1999 Harcourt Publishers Ltd.
Collapse
Affiliation(s)
- William S. Dalton
- Department of Interdisciplinary Oncology, H. Lee Moffitt Cancer Center, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
162
|
Abstract
In contrast to classical mutations, DNA methylation is a mechanism of changing the base sequence without altering the coding function of a gene. The interplay between this epigenetic modification and classical mutations plays an important role in tumorigenesis. Global genomic hypomethylation has been associated with the induction of chromosomal instability, which is commonly seen in solid tumors and multiple myeloma. De novo methylation of CpG islands on the promoter region may contribute to the progressive inactivation of growth-inhibitory genes resulting in the clonal selection of cells with growth advantage. Recently, alteration of p16 and p15 solely by hypermethylation has been detected in high frequencies hitherto unreported in multiple myeloma (MM). Hypermethylation of p16 has been shown to be associated with plasmablastic disease (p=0.026) in primary MM and transcriptional silencing of p16 and p15 has also been found to correlate with hypermethylation of these genes in MM-derived cell lines. Our results in studies with cell lines and primary MM support the fact that hypermethylation of p16 and p15 plays an important role in MM tumorigenesis. Because of its high frequency, the presence of hypermethylation of p16 may prove to be a useful tumor marker for the majority of MM patients. Promoters silenced by methylation can be reactivated by treatment with the demethylating agent 5-aza-2'deoxycytidine. The reversibility of this epigenetic inactivation of the p16 and p15 genes in MM may also provide a broad clinical application in the development of new therapeutic interventions in this uniformly fatal form of cancer.
Collapse
Affiliation(s)
- M H Ng
- Department of Anatomical & Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, SAR, China.
| | | | | |
Collapse
|
163
|
Tarte K, Zhang XG, Legouffe E, Hertog C, Mehtali M, Rossi JF, Klein B. Induced Expression of B7-1 on Myeloma Cells Following Retroviral Gene Transfer Results in Tumor-Specific Recognition by Cytotoxic T Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.1.514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The aim of this study was to evaluate whether tumor cells from patients with multiple myeloma activate allogeneic and autologous T cells. Results showed that myeloma cells expressed few B7-2 and no B7-1 in six cell lines and primary cells from 11 patients. They expressed substantial levels of HLA class I, CD40, and a set of adhesion molecules. In accordance with the low density of B7 molecules on these cells, they were poor allogeneic CD8+ T cell stimulators. Neither IFN-γ plus TNF-α nor CD40 stimulation significantly induced B7-1 or up-regulated B7-2 on human myeloma cell line or primary myeloma cells from six of seven patients. However, such induction was found on autologous bone-marrow nontumoral cells and on autologous dendritic cells following CD40 stimulation. High B7-1 expression was stably obtained on human myeloma cell line using transduction with a B7-1 retrovirus, enabling these cells to stimulate allogeneic CD8+, though not CD4+, T cell proliferation. For one patient with advanced disease, B7-1 gene transfer made it possible to amplify autologous cytotoxic T cells that killed autologous myeloma cells in an HLA class I-restricted manner, but not autologous PHA blasts. These results suggest that B7-1 gene transfer could be a promising immunotherapeutic approach in multiple myeloma.
Collapse
Affiliation(s)
- Karin Tarte
- *Institut National de la Santé et de la Recherche Médicale Unite 475, Montpellier, France
| | - Xue-Guang Zhang
- *Institut National de la Santé et de la Recherche Médicale Unite 475, Montpellier, France
- †Immunology Research Unit, Suzhou Medical College, Suzhou, China
| | - Eric Legouffe
- ‡Service des Maladies du Sang B, Centre Hospitalier Universitaire Montpellier, Hôpital Lapeyronie, Montpellier, France
| | - Catherine Hertog
- §Unite de Therapie Cellulaire, Centre Hospitalier Universitaire Montpellier, Hôpital Saint Eloi, Montpellier, France; and
| | | | - Jean-François Rossi
- ‡Service des Maladies du Sang B, Centre Hospitalier Universitaire Montpellier, Hôpital Lapeyronie, Montpellier, France
| | - Bernard Klein
- *Institut National de la Santé et de la Recherche Médicale Unite 475, Montpellier, France
- §Unite de Therapie Cellulaire, Centre Hospitalier Universitaire Montpellier, Hôpital Saint Eloi, Montpellier, France; and
| |
Collapse
|
164
|
Sarid R, Olsen SJ, Moore PS. Kaposi's sarcoma-associated herpesvirus: epidemiology, virology, and molecular biology. Adv Virus Res 1999; 52:139-232. [PMID: 10384236 DOI: 10.1016/s0065-3527(08)60299-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- R Sarid
- Division of Epidemiology, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | |
Collapse
|
165
|
Wierzbowska A, Urbańska-Ryś H, Robak T. Circulating IL-6-type cytokines and sIL-6R in patients with multiple myeloma. Br J Haematol 1999. [PMID: 10233412 DOI: 10.1111/j.1365-2141.1999.01350.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We investigated the serum concentration of interleukin-6 (IL-6) and four IL-6 family cytokines - oncostatin M (OSM), leukaemia inhibitory factor (LIF), interleukin-11 (IL-11) and ciliary neurotrophic factor (CNTF) as well as IL-6 soluble receptor (sIL-6R) - using an enzyme-linked immunosorbent assay (ELISA) in 67 patients with multiple myeloma (MM) and 24 healthy controls, for a possible association between the serum levels of these peptides with disease activity and known prognostic factors. sIL-6R was detectable in all 67 and IL-6 in 65 (97%) patients. Both peptides were measurable in all healthy controls. In contrast, OSM was detectable in 30 (44.8%) MM patients and in only four (16.6%) normal individuals. The serum levels of IL-6, OSM and sIL-6R were significantly higher in MM patients compared with control group (P < 0.001, P < 0.03 and P < 0. 001 respectively). The highest concentrations of these cytokines were found in patients with progressive disease and the lowest in MM patients with stable disease and in healthy persons. LIF was detectable in four (6%), CNTF in 28 (41.8%) and IL-11 in eight (11. 9%) of the patients with MM. In the control group LIF, CNTF and IL-11 were measurable in 8.3%, 33.3% and 8.3% respectively. The serum concentration of these cytokines did not correlate either with clinical stage or with the phase of disease and was similar to those in healthy individuals. We found significant positive correlation between IL-6 levels and OSM (P < 0.001). We also observed positive correlation between beta2-M concentration and serum levels of IL-6 (P < 0.002), sIL-6R (P < 0.02) and OSM (P < 0.04) as well as a positive relationship between CRP and IL-6 (P < 0.001) and OSM (P < 0.002). In conclusion, the serum levels of IL-6, OSM and sIL-6R, but not LIF, IL-11 and CNTF, may be useful markers of MM activity.
Collapse
Affiliation(s)
- A Wierzbowska
- Department of Haematology, Medical University of Lódź, Copernicus Hospital, Lódź, Poland
| | | | | |
Collapse
|
166
|
|
167
|
Affiliation(s)
- D Provan
- Department of Haematology, Southampton General Hospital, Southampton SO16 6UY.
| | | |
Collapse
|
168
|
Bellos F, Goldschmidt H, Dörner M, Ho AD, Moos M. Bone marrow derived dendritic cells from patients with multiple myeloma cultured with three distinct protocols do not bear Kaposi's sarcoma associated herpesvirus DNA. Ann Oncol 1999; 10:323-7. [PMID: 10355577 DOI: 10.1023/a:1008320717531] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND An association between Kaposi's sarcoma associated herpesvirus (KSHV) and the pathogenesis of multiple myeloma (MM) was postulated recently. The dendritic cells of patients with MM were proposed to be infected with the virus. PATIENTS AND METHODS Bone marrow mononuclear cells (MNC) of 23 patients, 22 with MM and one with MGUS, were cultured according to three distinct protocols for the generation of dendritic cells. One was essentially the stromal cell culture protocol described by Rettig et al. (Science 1997; 276: 1851-4), while the two other protocols comprised growth factors. Cultured cells were characterised by FACS analysis and assessed for the presence of KSHV DNA with a highly sensitive and specific nested PCR assay detecting the KS 330233 sequence of the virus genome followed by hybridisation with a KSHV specific oligonucleotide. RESULTS FACS analysis of the cells with the specific markers CD1a, CD86 and HLA-DR, characteristic for dendritic cells, revealed differences in the expression pattern depending on the protocol used. The proportion of CD1a+ cells was very low in the stromal cell cultures (median 0.4%), while a higher percentage of CD14+ cells could be observed (median 37.8%). Growth factor containing cultures revealed a distinctly higher median percentage of CD1a+ cells of 32.5%. The proportion of CD86+ cells varied between 10.4% and 78.5% and HLA-DR+ cells between 26% and 94.4%. Examination of those cells with PCR did not reveal positivity for KSHV in any of the 34 samples assessed. Amplification of seven samples revealed PCR products of approximately the size of the KS 330(233), which, however, could not be confirmed as KSHV specific after hybridisation. CONCLUSION We have no evidence that bone marrow derived dendritic cells from patients with MM are infected with KSHV.
Collapse
Affiliation(s)
- F Bellos
- Department of Internal Medicine V, University of Heidelberg, Germany
| | | | | | | | | |
Collapse
|
169
|
Ishioka S, van Haaften-Day C, Sagae S, Kudo R, Hacker NF. Interleukin-6 (IL-6) does not change the expression of Bcl-2 protein in the prevention of cisplatin-induced apoptosis in ovarian cancer cell lines. J Obstet Gynaecol Res 1999; 25:23-7. [PMID: 10067009 DOI: 10.1111/j.1447-0756.1999.tb01117.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To study whether interleukin-6 (IL-6) changes the expression of the anti-apoptic Bcl-2 protein in the prevention of cis-diaminedichloroplatinum (II) (CDDP)-induced apoptosis of human ovarian cancer cells. METHODS Comparative studies were performed on 3 ovarian cancer cell lines after 48 hours of exposure to 0.5-50 ng/ml IL-6, 2 micrograms/ml anti-IL-6 monoclonal antibody (anti-IL-6 mAb), 10 microM CDDP, 10 microM CDDP + 0.5-50 ng/ml IL-6, and 10 microM CDDP + 2 micrograms/ml anti-IL-6 mAb. Apoptosis was measured morphologically and by a DNA fragmentation assay. Bcl-2 protein levels were measured by an ELISA. RESULTS An increase in apoptosis was observed for each cell line after 48 hours of exposure to 10 microM CDDP. Although high doses of IL-6 decreased the percentage of apoptotic cells, this cytokine did not change the expression of the Bcl-2 protein. CONCLUSION CDDP-induced apoptosis was negatively controlled by IL-6. However, the anti-apoptic Bcl-2 protein level was not changed by IL-6 in the process of apoptosis in the ovarian cancer cell lines.
Collapse
Affiliation(s)
- S Ishioka
- Department of Gynecological Oncology, Royal Hospital for Women, Sydney, NSW, Australia
| | | | | | | | | |
Collapse
|
170
|
Witzig TE, Timm M, Larson D, Therneau T, Greipp PR. Measurement of apoptosis and proliferation of bone marrow plasma cells in patients with plasma cell proliferative disorders. Br J Haematol 1999; 104:131-7. [PMID: 10027725 DOI: 10.1046/j.1365-2141.1999.01136.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The proliferative rate of malignant plasma cells, as measured by the plasma cell labelling index (PCLI), is an important prognostic factor in multiple myeloma (MM); however, the PCLI alone is probably Inadequate to describe tumour growth because it ignores the idea that myeloma cells may have a reduced rate of apoptosis. The aims of this study were to develop a flow cytometric method to measure the apoptosis index of fresh marrow plasma cells and develop a plasma cell growth index (PCGI) that related both proliferation and apoptosis to disease activity. Marrow aspirates were obtained from 91 patients with plasma cell disorders and the plasma cells in apoptosis were identified by either 7-amino actinomycin-D (7-AAD) or annexin V-FITC three-colour flow cytometry. The median plasma cell apoptotic index (PCAI) for patients with monoclonal gammopathy of undetermined significance (MGUS), smouldering or indolent myeloma (SMM/IMM), and new multiple myeloma (MM) was 5.2, 3.4 and 2.4, respectively (P=0.03, MGUS v MM). The median PCLI for these same patient groups was 0.0, 0.2 and 0.6, respectively (P<0.001, MGUS v MM). The paired PCLI and PCAI for each sample were used to derive the PCGI=2 + [PCLI-(O.1)(PCAI)]. The median PCGI for patients with inactive disease (MGUS, SMM/IMM or amyloidosis) was 1.8 compared to 2.4 for those with active disease (new or relapsed MM) (P<0.001). These results suggest that a decrease in the PCAI may be a factor in the progression from MGUS to SMM to overt MM.
Collapse
Affiliation(s)
- T E Witzig
- Department of Internal Medicine, Mayo Clinic and Mayo Foundation, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
171
|
Catlett-Falcone R, Landowski TH, Oshiro MM, Turkson J, Levitzki A, Savino R, Ciliberto G, Moscinski L, Fernández-Luna JL, Nuñez G, Dalton WS, Jove R. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity 1999; 10:105-15. [PMID: 10023775 DOI: 10.1016/s1074-7613(00)80011-4] [Citation(s) in RCA: 1287] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Interleukin 6 (IL-6) is the major survival factor for myeloma tumor cells and induces signaling through the STAT proteins. We report that one STAT family member, Stat3, is constitutively activated in bone marrow mononuclear cells from patients with multiple myeloma and in the IL-6-dependent human myeloma cell line U266. Moreover, U266 cells are inherently resistant to Fas-mediated apoptosis and express high levels of the antiapoptotic protein Bcl-xL. Blocking IL-6 receptor signaling from Janus kinases to the Stat3 protein inhibits Bcl-xL expression and induces apoptosis, demonstrating that Stat3 signaling is essential for the survival of myeloma tumor cells. These findings provide evidence that constitutively activated Stat3 signaling contributes to the pathogenesis of multiple myeloma by preventing apoptosis.
Collapse
Affiliation(s)
- R Catlett-Falcone
- H. Lee Moffitt Cancer Center and Research Institute, University of South Florida College of Medicine, Tampa 33612, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Bellos F, Cremer FW, Ehrbrecht E, Goldschmidt H, Moos M. Leukapheresis cells of patients with multiple myeloma collected after mobilization with chemotherapy and G-CSF do not bear Kaposi's sarcoma associated herpesvirus DNA. Br J Haematol 1998; 103:1192-7. [PMID: 9886341 DOI: 10.1046/j.1365-2141.1998.01148.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The presence of Kaposi's sarcoma associated herpesvirus (KSHV) in bone marrow dendritic cells, in bone marrow biopsies and in dendritic cells of peripheral blood from patients with multiple myeloma (MM) has been reported. These data suggested an association between infection with KSHV and the development of MM. The mobilization of infected cells into leukapheresis products (LP) has also been described. We assessed the LP of 35 patients with MM for the presence of KSHV using a sensitive and specific nested PCR assay, capable of detecting one copy of the virus genome. None of the samples tested revealed positivity for KSHV after amplification and subsequent hybridization with a KSHV-specific probe. Amplification products of approximately the same size as the positive control seen in eight samples did not hybridize with the specific oligonucleotide. No homologies of these products to the KSHV genome could be discovered after sequencing. Therefore we have no evidence that LP of patients with MM bear KSHV, and they can therefore be used as a source for dendritic cells for immunotherapy.
Collapse
Affiliation(s)
- F Bellos
- Department of Internal Medicine V, University of Heidelberg, Germany
| | | | | | | | | |
Collapse
|
173
|
Frassanito MA, Silvestris F, Silvestris N, Cafforio P, Camarda G, Iodice G, Dammacco F. Fas/Fas ligand (FasL)-deregulated apoptosis and IL-6 insensitivity in highly malignant myeloma cells. Clin Exp Immunol 1998; 114:179-88. [PMID: 9822274 PMCID: PMC1905107 DOI: 10.1046/j.1365-2249.1998.00711.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
IL-6 is a growth factor which interferes in the apoptosis of malignant plasma cells. Here we explore its role in the spontaneous and Fas/FasL-regulated apoptosis of seven myeloma cell clones (MCC). MCC-2 and -7 were constitutively defective in Fas antigen in the presence of large membrane exposure of FasL, and showed a high rate of cell proliferation irrespective of the presence of IL-6. Cytofluorimetric analysis following propidium iodide (PI) staining revealed a minimal extent of spontaneous apoptosis, as in other IL-6-insensitive, though Fas-positive MCC, namely MCC-3 and -5. By contrast, a regular amplitude of apoptosis occurred in the remaining IL-6-dependent clones. Their propensity to cell death, as well as their FasL membrane expression, were promptly down-modulated by the cytokine, whereas no substantial effect was detected in IL-6-independent MCC. Furthermore, we investigated the quantitative secretion of FasL. Both [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] (MTT) cytotoxicity assay and PI staining of WC8 lymphoblasts from a Fas-transfected mouse lymphoma, incubated with supernatants from MCC, showed a variable cytocidal property, thus confirming the cellular release of FasL. However, a significant elevation of FasL secretion occurred in both Fas- MCC, whereas molecular cloning and sequencing of Fas revealed the presence of a splicing variant, namely Fas Exo4,6Del, in the cDNA from both MCC-3 and -5, which were previously demonstrated to be unresponsive to Fas stimulation. Taken together, these data provide evidence that concurrence of IL-6 insensitivity and deregulation of apoptosis in myeloma cells reflects a high malignancy grade. It is suggested that the secretion of Fas splicing variants in Fas+ plasma cells, as well as the over-production of FasL in Fas- myelomas, are differential mechanisms by which myeloma cells escape host immune surveillance.
Collapse
Affiliation(s)
- M A Frassanito
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Italy
| | | | | | | | | | | | | |
Collapse
|
174
|
Ferlin-Bezombes M, Jourdan M, Liautard J, Brochier J, Rossi JF, Klein B. IFN-α Is a Survival Factor for Human Myeloma Cells and Reduces Dexamethasone-Induced Apoptosis. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.6.2692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
IFN-α is used as a maintenance therapy in patients with multiple myeloma, but its benefit is a matter of controversy. In vitro studies show that IFN-α can both stimulate and inhibit myeloma cell proliferation. We have tested the effect of IFN-α on the survival of myeloma cell lines and primary plasma cells. IFN-α significantly reduced the apoptosis induced by removal of IL-6 in four IL-6-dependent myeloma cell lines. It also reduced the level of apoptosis induced by dexamethasone in these cell lines as well as in purified primary myeloma cells from seven patients. IFN-α promoted the survival of myeloma cells, which, following removal of IL-6, were blocked in G1 and died. However, unlike IL-6, IFN-α-treated cells remained mainly blocked in the G1 phase of the cycle. While the effects of IL-6 are mediated through stimulation of its gp130 receptor subunit, the IFN-α-induced survival of myeloma cells was independent of gp130 transducer activation (as demonstrated using a neutralizing anti-gp130 Ab). However, the signal transduction cascades activated by these two cytokines share at least some common elements, since stimulation with either IFN-α or IL-6 resulted in STAT3 phosphorylation. These results indicate that IFN-α promotes the survival, but not the proliferation, of myeloma cells, preventing the apoptosis induced by removal of IL-6 or addition of dexamethasone. This survival factor activity may explain the conflicting reports on the effects of IFN-α on myeloma cell proliferation.
Collapse
Affiliation(s)
| | - Michel Jourdan
- *Institut National de la Santé et de la Recherche Médicale, Unit 475,
| | - Janny Liautard
- *Institut National de la Santé et de la Recherche Médicale, Unit 475,
| | - Jean Brochier
- *Institut National de la Santé et de la Recherche Médicale, Unit 475,
| | - Jean-François Rossi
- †Service des Maladies du Sang B, Centre Hospitalier Universitaire Montpellier, and
| | - Bernard Klein
- *Institut National de la Santé et de la Recherche Médicale, Unit 475,
- ‡Unit for Cellular Therapy, Centre Hospitalier Universitaire Montpellier, Hôpital Saint Eloi, Montpellier, France
| |
Collapse
|
175
|
Abstract
Multiple counterregulatory mechanisms have been identified in B-cell precursors that operate to regulate cell survival and growth, thereby ensuring the orderly development and differentiation of B-cells. Inappropriate apoptosis may underlie the pathogenesis of immunodeficiencies, as well as pathogenesis and drug/radiation resistance of human leukemias and lymphomas, which makes control of apoptosis an important potential target for therapeutic interventions. Therefore, identification of the molecular regulators of apoptosis is an area of intense investigation. Bruton's tyrosine kinase (BTK) is the first tyrosine kinase to be identified as a dual-function regulator of apoptosis, which promotes radiation-induced apoptosis but inhibits Fas-activated apoptosis in B-cells. BTK functions in a pro-apoptotic manner when B-cells are exposed to reactive oxygen intermediates, at least in part, by down-regulating the anti-apoptotic activity of STAT-3 transcription factor. In contrast, BTK associates with the death receptor Fas and impairs its interaction with Fas-associated protein with death domain (FADD), which is essential for the recruitment and activation of FLICE by Fas during the apoptotic signal, thereby preventing the assembly of a pro-apoptotic death inducing signaling complex (DISC) after Fas-ligation. The identification of BTK as a dual-function regulator of apoptosis will significantly increase our understanding of both the biological processes involved in programmed cell death and the diseases associated with dysregulation of apoptosis. New agents with BTK-modulatory activity may have clinical potential in the treatment of B-cell malignancies (in particular acute lymphoblastic leukemia, the most common form of childhood cancer), as well as B-cell immunodeficiencies.
Collapse
Affiliation(s)
- F M Uckun
- Wayne Hughes Institute, St. Paul, MN 55113, USA
| |
Collapse
|
176
|
Abstract
Abstract
Previous studies have suggested that multiple myeloma (MM) cells express estrogen receptors (ER). In the present study, we characterized the effects of estrogen agonists and antagonists (anti-estrogens [AE]) on growth of MM cell lines and MM patient cells. In addition to antagonizing estrogen binding to ER, AE can trigger apoptosis. Hence, we also determined whether estrogens or AE altered MM cell survival. Immunoblotting showed that ER- is expressed in 4 of 5 MM cell lines (ARH-77, RPMI 8226, S6B45, and U266, but not OCI-My-5 cells), as well as in freshly isolated MM cells from 3 of 3 patients. 17β-estradiol (E2) did not significantly alter proliferation of MM cell lines or MM patient cells. In contrast, two structurally distinct AE, tamoxifen (TAM) and ICI 182,780 (ICI), significantly inhibited the proliferation of all 5 MM cell lines and MM cells from 2 of 2 patients (IC50, 2 to 4 μmol/L). Proliferation of these cell lines was also inhibited by the hydroxylated TAM derivative, 4-hydroxytamoxifen (4HTAM), although this derivative was less potent than TAM (IC50, 3 to 25 μmol/L). In contrast, the dehalogenated TAM derivative toremifene (TOR) did not inhibit MM cell proliferation. We next examined the effects of these agents on MM cell survival. TAM, ICI, and, to a lesser extent, 4HTAM and TOR triggered apoptosis in both ER-–positive as well as ER-–negative MM cell lines and patient MM cells, evidenced both by fluorescence-activated cell sorting (FACS) analysis using propidium iodide staining and the TUNEL assay. TAM-induced growth inhibition and apoptosis of ER-–positive S6B45 MM cells was not blocked by coculture with excess E2. TAM-induced apoptosis of S6B45 MM cells was also unaffected by addition of exogenous interleukin-6. Importantly, both the inhibition of MM cell proliferation and the induction of MM cell apoptosis were achieved at concentrations of TAM (0.5 and 5.0 μmol/L) that did not significantly alter in vitro growth of normal hematopoietic progenitor cells. Similar plasma levels of TAM have been achieved using high-dose oral TAM therapy, with an acceptable toxicity profile. These studies therefore provide the rationale for trials to define the utility of AE therapy in MM.
© 1998 by The American Society of Hematology.
Collapse
|
177
|
Abstract
Previous studies have suggested that multiple myeloma (MM) cells express estrogen receptors (ER). In the present study, we characterized the effects of estrogen agonists and antagonists (anti-estrogens [AE]) on growth of MM cell lines and MM patient cells. In addition to antagonizing estrogen binding to ER, AE can trigger apoptosis. Hence, we also determined whether estrogens or AE altered MM cell survival. Immunoblotting showed that ER- is expressed in 4 of 5 MM cell lines (ARH-77, RPMI 8226, S6B45, and U266, but not OCI-My-5 cells), as well as in freshly isolated MM cells from 3 of 3 patients. 17β-estradiol (E2) did not significantly alter proliferation of MM cell lines or MM patient cells. In contrast, two structurally distinct AE, tamoxifen (TAM) and ICI 182,780 (ICI), significantly inhibited the proliferation of all 5 MM cell lines and MM cells from 2 of 2 patients (IC50, 2 to 4 μmol/L). Proliferation of these cell lines was also inhibited by the hydroxylated TAM derivative, 4-hydroxytamoxifen (4HTAM), although this derivative was less potent than TAM (IC50, 3 to 25 μmol/L). In contrast, the dehalogenated TAM derivative toremifene (TOR) did not inhibit MM cell proliferation. We next examined the effects of these agents on MM cell survival. TAM, ICI, and, to a lesser extent, 4HTAM and TOR triggered apoptosis in both ER-–positive as well as ER-–negative MM cell lines and patient MM cells, evidenced both by fluorescence-activated cell sorting (FACS) analysis using propidium iodide staining and the TUNEL assay. TAM-induced growth inhibition and apoptosis of ER-–positive S6B45 MM cells was not blocked by coculture with excess E2. TAM-induced apoptosis of S6B45 MM cells was also unaffected by addition of exogenous interleukin-6. Importantly, both the inhibition of MM cell proliferation and the induction of MM cell apoptosis were achieved at concentrations of TAM (0.5 and 5.0 μmol/L) that did not significantly alter in vitro growth of normal hematopoietic progenitor cells. Similar plasma levels of TAM have been achieved using high-dose oral TAM therapy, with an acceptable toxicity profile. These studies therefore provide the rationale for trials to define the utility of AE therapy in MM.
© 1998 by The American Society of Hematology.
Collapse
|
178
|
Interleukin-6–Induced Inhibition of Multiple Myeloma Cell Apoptosis: Support for the Hypothesis That Protection Is Mediated Via Inhibition of the JNK/SAPK Pathway. Blood 1998. [DOI: 10.1182/blood.v92.1.241.413k28_241_251] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The mechanism by which interleukin-6 (IL-6) protects multiple myeloma (MM) plasma cells from apoptosis induced by anti-fas antibodies and dexamethasone was studied. Anti-apoptotic concentrations of IL-6 had no effect on cell-cycle distribution or activation of RAF-1 or ERK in dexamethasone- or anti–fas-treated 8226 and UCLA #1 MM cell lines. However, IL-6–dependent protection of viability correlated with an inhibition of dexamethasone- and anti–fas-induced activation ofjun kinase (JNK) and AP-1 transactivation. To test the hypothesis that cytokine-induced protection was mediated through inhibition of JNK/c-jun, we also inhibited c-junfunction in 8226 cells via introduction of a mutant dominant negative c-jun construct. Mutant c-jun–containing MM cells were also resistant to anti–fas-induced apoptosis but were significantly more sensitive to dexamethasone-induced apoptosis. These results support the notion that IL-6 protects MM cells against anti-fas through its inhibitory effects on JNK/c-junbut indicate protection against dexamethasone occurs through separate, yet unknown pathways.
Collapse
|
179
|
Sati HI, Apperley JF, Greaves M, Lawry J, Gooding R, Russell RG, Croucher PI. Interleukin-6 is expressed by plasma cells from patients with multiple myeloma and monoclonal gammopathy of undetermined significance. Br J Haematol 1998; 101:287-95. [PMID: 9609524 DOI: 10.1046/j.1365-2141.1998.00687.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interleukin-6 (IL-6) is an important growth factor for human myeloma cells in vitro and in vivo. However, the identity of the cells producing IL-6 in vivo in patients with multiple myeloma (MM) remains the subject of debate. We have developed a sensitive dual-colour fluorescence in situ hybridization (FISH) technique to investigate the expression of IL-6 mRNA by individual bone marrow plasma cells from patients with multiple myeloma, monoclonal gammopathy of undetermined significance (MGUS) and healthy subjects. IL-6 mRNA could be identified in all immunoglobulin light chain (IgLC) expressing cells from all patients with MM and MGUS. The IL-6 protein could also be detected by direct immunofluorescence in all plasma cells (cytoplasmic light chain positive) from all patients with MM and MGUS. Furthermore, it was also possible to demonstrate cytoplasmic IL-6 staining of plasma cells from patients with MM by flow cytometric analysis. In contrast, neither the IL-6 mRNA or protein could be detected in normal plasma cells from healthy bone marrow donors. These data demonstrate that plasma cells from patients with MM and MGUS express the IL-6 mRNA and synthesize the IL-6 protein and support the hypothesis that autocrine synthesis of IL-6 is of importance in patients with MM.
Collapse
Affiliation(s)
- H I Sati
- Department of Human Metabolism and Clinical Biochemistry, University of Sheffield Medical School
| | | | | | | | | | | | | |
Collapse
|
180
|
Siegel DS, Zhang X, Feinman R, Teitz T, Zelenetz A, Richon VM, Rifkind RA, Marks PA, Michaeli J. Hexamethylene bisacetamide induces programmed cell death (apoptosis) and down-regulates BCL-2 expression in human myeloma cells. Proc Natl Acad Sci U S A 1998; 95:162-6. [PMID: 9419346 PMCID: PMC18160 DOI: 10.1073/pnas.95.1.162] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma (MM) is a B cell malignancy characterized by the expansion of monoclonal Ig-secreting plasma cells with low proliferative activity. It is postulated that inhibition of physiologic cell death is an underlying factor in the pathophysiology of MM. The development of chemoresistance is a common feature in patients with MM. In the present studies, hexamethylene bisacetamide (HMBA), a hybrid polar compound that is a potent inducer of terminal differentiation of various transformed cells, is shown to inhibit the growth of several human myeloma cell lines (ARP-1, U266, and RPMI 8226), including doxorubicin-resistant RPMI 8226 variants that overexpress the multidrug-resistance gene, MDR-1, and its product, p-glycoprotein. In addition to growth arrest and suppression of clonogenicity, HMBA induces apoptosis both in freshly isolated human myeloma cells and in cell lines, as determined by morphologic alterations, cell cycle distribution and endonucleosomal DNA fragmentation. Further, HMBA decreases BCL-2 protein expression in myeloma cells within 12-48 hr. Overexpression of BCL-2 protein in ARP-1 cells confers resistance to HMBA-induced apoptosis. Taken together, these data suggest that HMBA is a potent inducer of apoptosis in human myeloma cells, which may act through suppressing the anti-apoptotic function of the bcl-2 gene. HMBA, and related hybrid polar compounds, may prove useful in the management of this presently incurable disease.
Collapse
Affiliation(s)
- D S Siegel
- Program of Cell Biology, The University of Arkansas, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
|
182
|
|
183
|
Bloem AC, Lamme T, de Smet M, Kok H, Vooijs W, Wijdenes J, Boom SE, Lokhorst HM. Long-term bone marrow cultured stromal cells regulate myeloma tumour growth in vitro: studies with primary tumour cells and LTBMC-dependent cell lines. Br J Haematol 1998; 100:166-75. [PMID: 9450806 DOI: 10.1046/j.1365-2141.1998.00517.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Long-term bone marrow cultured stromal cells (LTBMC) produce IL-6 after contact with tumour cells from multiple myeloma patients. We found that LTBMC could substitute for exogenous IL-6 in the stimulation of bone marrow plasma cells from myeloma patients with active disease in short-term cultures. In addition, tumour cells of some patients with inactive disease, which were unresponsive to exogenous IL-6, were induced to IL-6-dependent growth after LTBMC co-culture. To study the role of LTBMC in myeloma tumour growth in vitro, plasma cell lines UM-2 and UM-3 were selected. UM-2 and UM-3 grew in contact with LTBMC and proliferation was blocked by antibodies against IL-6, IL-6 receptor (IL-6R, gp80, CD126) or the common signal transducing unit, gp130 (CD130). Culture with IL-6 alone or combined with GM-CSF resulted in cell death via apoptosis. The combination of IL-6 with soluble gp80, however, maintained in vitro proliferation of UM-2 and UM-3 cells. These data imply that LTBMC regulate myeloma growth in vitro via production of IL-6, possibly via induction of a functional IL-6 receptor on the tumour cells.
Collapse
Affiliation(s)
- A C Bloem
- Department of Immunology, University Hospital Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
184
|
Siegel DS, Terry JA, Koury J, Barlogie B, Epstein J, Feinman R. Myc/max family of transcription factors and bcl-2 are involved in drug-induced apoptosis of myeloma cells. Curr Top Microbiol Immunol 1997; 224:257-60. [PMID: 9308249 DOI: 10.1007/978-3-642-60801-8_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- D S Siegel
- Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, USA
| | | | | | | | | | | |
Collapse
|
185
|
Molden J, Chang Y, You Y, Moore PS, Goldsmith MA. A Kaposi's sarcoma-associated herpesvirus-encoded cytokine homolog (vIL-6) activates signaling through the shared gp130 receptor subunit. J Biol Chem 1997; 272:19625-31. [PMID: 9235971 DOI: 10.1074/jbc.272.31.19625] [Citation(s) in RCA: 218] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The present studies analyzed the biologic activity of a gene product (vIL-6) encoded by the recently discovered Kaposi's sarcoma-associated herpesvirus (KSHV) bearing 24.8% amino acid identity with human interleukin-6 (huIL-6). Based on this similarity, we hypothesized that this viral homolog might trigger the JAK/STAT pathway, which typically is engaged by IL-6 and other cytokines. Activation of receptor-associated Janus tyrosine kinases (JAKs) results in the subsequent phosphorylation of signal transducers and activators of transcription (STATs) leading to nuclear entry and transcriptional regulation of target genes. Treatment of HepG2 cells with culture medium containing recombinant KSHV-encoded vIL-6 led to rapid induction of JAK1 phosphorylation and a nuclear DNA-binding activity found to contain STAT1 and STAT3. An antibody to the IL-6 receptor (IL-6R) alpha subunit effectively neutralized the response to huIL-6 but failed to block STAT activation by vIL-6. In contrast, an antibody reactive with the gp130 subunit of IL-6R abrogated signaling of both responses. Moreover, a transfected cell line expressing human gp130 without IL-6Ralpha exhibited a robust response to vIL-6 but not to huIL-6. These results demonstrate that KSHV encodes a cytokine that activates specific JAK/STAT signaling via interactions with the gp130 signal transducing subunit independently of the IL-6Ralpha chain. This activity may have an impact on gp130-mediated signaling in response to native cytokines and thereby influence disease pathogenesis upon KSHV infection.
Collapse
Affiliation(s)
- J Molden
- Gladstone Institute of Virology and Immunology, San Francisco, California 94141-9100, USA
| | | | | | | | | |
Collapse
|
186
|
Abstract
Abstract
Prior in vitro studies have suggested a role of adhesion molecules, bone marrow stromal cells (BMSCs), and cytokines in the regulation of human multiple myeloma (MM) cell growth and survival. Although in vivo models have been developed in severe combined immunodeficient (SCID) mice that support the growth of human MM within the murine BM microenvironment, these xenograft models do not permit a study of the role of adhesion proteins in human MM cell-human BMSC interactions. We therefore established an in vivo model of human MM using SCID mice implanted with bilateral human fetal bone grafts (SCID-hu mice). For the initial tumor innoculum, human MM derived cell lines (1 × 104 or 5 × 104 ARH-77, OCI-My5, U-266, or RPMI-8226 cells) were injected directly into the BM cavity of the left bone implants in irradiated SCID-hu mice. MM cells engrafted and proliferated in the left human fetal bone implants within SCID-hu mice as early as 4 weeks after injection of as few as 1 × 104 MM cells. To determine whether homing of tumor cells occurred, animals were observed for up to 12 weeks after injection and killed to examine for tumor in the right bone implants. Of great interest, metastases to the right bone implants were observed at 12 weeks after the injection of 5 × 104 MM cells, without spread of human MM cells to murine BM. Human MM cells were identified on the basis of characteristic histology and monoclonal human Ig. Importantly, monoclonal human Ig and human interleukin-6 (IL-6), but not human IL-1β or tumor necrosis factor-α, were detectable in sera of SCID-hu mice injected with MM cells. In addition, specific monoclonal Ig light chain deposition was evident within renal tubules. This in vivo model of human MM provides for the first time a means for identifying adhesion molecules that are responsible for specific homing of human MM cells to the human, as opposed to murine, BM microenvironment. Moreover, induction of human IL-6 suggests the possibility that regulation of MM cell growth by this cytokine might also be investigated using this in vivo model.
Collapse
|
187
|
Abstract
Prior in vitro studies have suggested a role of adhesion molecules, bone marrow stromal cells (BMSCs), and cytokines in the regulation of human multiple myeloma (MM) cell growth and survival. Although in vivo models have been developed in severe combined immunodeficient (SCID) mice that support the growth of human MM within the murine BM microenvironment, these xenograft models do not permit a study of the role of adhesion proteins in human MM cell-human BMSC interactions. We therefore established an in vivo model of human MM using SCID mice implanted with bilateral human fetal bone grafts (SCID-hu mice). For the initial tumor innoculum, human MM derived cell lines (1 × 104 or 5 × 104 ARH-77, OCI-My5, U-266, or RPMI-8226 cells) were injected directly into the BM cavity of the left bone implants in irradiated SCID-hu mice. MM cells engrafted and proliferated in the left human fetal bone implants within SCID-hu mice as early as 4 weeks after injection of as few as 1 × 104 MM cells. To determine whether homing of tumor cells occurred, animals were observed for up to 12 weeks after injection and killed to examine for tumor in the right bone implants. Of great interest, metastases to the right bone implants were observed at 12 weeks after the injection of 5 × 104 MM cells, without spread of human MM cells to murine BM. Human MM cells were identified on the basis of characteristic histology and monoclonal human Ig. Importantly, monoclonal human Ig and human interleukin-6 (IL-6), but not human IL-1β or tumor necrosis factor-α, were detectable in sera of SCID-hu mice injected with MM cells. In addition, specific monoclonal Ig light chain deposition was evident within renal tubules. This in vivo model of human MM provides for the first time a means for identifying adhesion molecules that are responsible for specific homing of human MM cells to the human, as opposed to murine, BM microenvironment. Moreover, induction of human IL-6 suggests the possibility that regulation of MM cell growth by this cytokine might also be investigated using this in vivo model.
Collapse
|
188
|
Interleukin-6 Overcomes p21WAF1 Upregulation and G1 Growth Arrest Induced by Dexamethasone and Interferon-γ in Multiple Myeloma Cells. Blood 1997. [DOI: 10.1182/blood.v90.1.279] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractInterleukin-6 (IL-6) is a growth factor for multiple myeloma (MM) cells and can inhibit MM cell apoptosis. Our recent studies show that IL-6 facilitates MM cell growth via phosphorylation of retinoblastoma protein (pRB); however, the effects of IL-6 on those cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors (CDIs) that are known to regulate phosphorylation of pRB have not been defined in MM cells. In the present report, we cultured MM cell lines and patient cells with IL-6 and/or dexamethasone (Dex) and characterized changes in cell cycle; expression and association of cyclins, CDKs, and CDIs; and phosphorylation of pRB. Dex induced G1 growth arrest in MM cells, whereas IL-6 facilitated G1 to S phase transition; moreover, the effect of Dex was blocked by IL-6. p21WAF1 (p21) protein was constitutively expressed in the majority of MM cells independent of the status of p53. Its expression was upregulated by Dex and downregulated by IL-6; again, IL-6 inhibited the increase in p21 triggered by Dex. These alterations in p21 expression in MM cells were associated with changes in p21 binding to CDK2, CDK4, and CDK6; CDK2, CDK4, and CDK6 kinase activities; and phosphorylation of pRB. In contrast, expression of G1 cell cycle regulatory proteins, including p27KIP1, cyclin D2, and cyclin E, was not altered in MM cells cultured with Dex and/or IL-6. Finally, interferon-γ (IFN-γ) also induced G1 growth arrest and upregulated p21 protein expression; as with Dex, affects of IFN-γ were inhibited by IL-6. Our results therefore show that changes in cell cycle distribution in MM cells triggered by Dex, IL-6, and IFN-γ correlate with changes in p21 protein expression and implicate p21 in the coupling of Dex-, IL-6–, and IFN-γ–related signals to G1 cell cycle regulation in MM cells.
Collapse
|
189
|
Interleukin-6 Overcomes p21WAF1 Upregulation and G1 Growth Arrest Induced by Dexamethasone and Interferon-γ in Multiple Myeloma Cells. Blood 1997. [DOI: 10.1182/blood.v90.1.279.279_279_289] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin-6 (IL-6) is a growth factor for multiple myeloma (MM) cells and can inhibit MM cell apoptosis. Our recent studies show that IL-6 facilitates MM cell growth via phosphorylation of retinoblastoma protein (pRB); however, the effects of IL-6 on those cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors (CDIs) that are known to regulate phosphorylation of pRB have not been defined in MM cells. In the present report, we cultured MM cell lines and patient cells with IL-6 and/or dexamethasone (Dex) and characterized changes in cell cycle; expression and association of cyclins, CDKs, and CDIs; and phosphorylation of pRB. Dex induced G1 growth arrest in MM cells, whereas IL-6 facilitated G1 to S phase transition; moreover, the effect of Dex was blocked by IL-6. p21WAF1 (p21) protein was constitutively expressed in the majority of MM cells independent of the status of p53. Its expression was upregulated by Dex and downregulated by IL-6; again, IL-6 inhibited the increase in p21 triggered by Dex. These alterations in p21 expression in MM cells were associated with changes in p21 binding to CDK2, CDK4, and CDK6; CDK2, CDK4, and CDK6 kinase activities; and phosphorylation of pRB. In contrast, expression of G1 cell cycle regulatory proteins, including p27KIP1, cyclin D2, and cyclin E, was not altered in MM cells cultured with Dex and/or IL-6. Finally, interferon-γ (IFN-γ) also induced G1 growth arrest and upregulated p21 protein expression; as with Dex, affects of IFN-γ were inhibited by IL-6. Our results therefore show that changes in cell cycle distribution in MM cells triggered by Dex, IL-6, and IFN-γ correlate with changes in p21 protein expression and implicate p21 in the coupling of Dex-, IL-6–, and IFN-γ–related signals to G1 cell cycle regulation in MM cells.
Collapse
|
190
|
Rettig MB, Ma HJ, Vescio RA, Põld M, Schiller G, Belson D, Savage A, Nishikubo C, Wu C, Fraser J, Said JW, Berenson JR. Kaposi's sarcoma-associated herpesvirus infection of bone marrow dendritic cells from multiple myeloma patients. Science 1997; 276:1851-4. [PMID: 9188529 DOI: 10.1126/science.276.5320.1851] [Citation(s) in RCA: 346] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) was found in the bone marrow dendritic cells of multiple myeloma patients but not in malignant plasma cells or bone marrow dendritic cells from normal individuals or patients with other malignancies. In addition the virus was detected in the bone marrow dendritic cells from two out of eight patients with monoclonal gammopathy of undetermined significance (MGUS), a precursor to myeloma. Viral interleukin-6, the human homolog of which is a growth factor for myeloma, was found to be transcribed in the myeloma bone marrow dendritic cells. KSHV may be required for transformation from MGUS to myeloma and perpetuate the growth of malignant plasma cells.
Collapse
Affiliation(s)
- M B Rettig
- Division of Hematology and Oncology, Veterans Affairs West Los Angeles Medical Center, Los Angeles, CA 90073, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Teoh G, Anderson KC. Interaction of tumor and host cells with adhesion and extracellular matrix molecules in the development of multiple myeloma. Hematol Oncol Clin North Am 1997; 11:27-42. [PMID: 9081202 DOI: 10.1016/s0889-8588(05)70413-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Adhesion molecules play an important role in the growth regulation and migration of multiple myeloma (MM) cells. They mediate homing of MM cells to the bone marrow and MM cell to bone marrow stromal cell adhesion, with resultant interleukin-6 related autocrine and paracine growth and antiapoptotic affects. Their pattern of expression on tumor cells correlates with the development of plasma cell leukemia or extramedullary disease. Clinically, expression of adhesion molecules on tumor cells or in the serum has already shown prognostic utility. Finally, since adhesion molecules are involved at multiple steps in the pathogenesis of MM, therapeutic studies may target these molecules.
Collapse
Affiliation(s)
- G Teoh
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | |
Collapse
|
192
|
Feinman R, Sawyer J, Hardin J, Tricot G. Cytogenetics and molecular genetics in multiple myeloma. Hematol Oncol Clin North Am 1997; 11:1-25. [PMID: 9081201 DOI: 10.1016/s0889-8588(05)70412-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Specific cytogenetic abnormalities have been identified in multiple myeloma that confer a poor prognosis, even with intensive chemotherapy and autotransplants. The identification and characterization of potential genes involved in these different chromosomal changes and their interplay with oncogenes and tumor suppressor genes controlling cellular growth and apoptosis is the major focus of this review.
Collapse
Affiliation(s)
- R Feinman
- Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, USA
| | | | | | | |
Collapse
|
193
|
Abstract
The phenotypic heterogeneity of myeloma cells in fact delineates a differentiation process that appears to be an integral part of the disease process. Immature myeloma cells interact with their microenvironment differently than do the more mature cells. As a result of this interaction, the immature cells display different responses to chemotherapy than do the mature cells. Addressing this issue by tailoring treatment to target immature as well as mature myeloma cells may change dramatically the outcome of treatment. The ability to define the myeloma clone by molecular genetic techniques has markedly increased the ability to detect clonal cells. This technique provides a most sensitive tool for monitoring elimination of tumor cells; however, the role of the early clonal B cells identified through the use of ASO-PCR in the disease process needs to be clarified. Currently, a great deal of effort is directed towards development of treatment protocol that will eliminate all clonal cells, and a method of purging clonal cells from harvested mobilized peripheral stem cells. Understanding the biologic significance of early clonal B cells in myeloma will allow for a more rational approach to curative treatment.
Collapse
Affiliation(s)
- J Epstein
- Division of Hematology/Oncology, Arkansas Cancer Research Center, University of Arkansas for Medical Sciences, Little Rock, USA
| |
Collapse
|
194
|
Interleukin-6 Inhibits Fas-Induced Apoptosis and Stress-Activated Protein Kinase Activation in Multiple Myeloma Cells. Blood 1997. [DOI: 10.1182/blood.v89.1.227.227_227_234] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fas belongs to the family of type-1 membrane proteins that transduce apoptotic signals. In the present studies, we characterized signaling during Fas-induced apoptosis in RPMI-8226 and IM-9 multiple myeloma (MM) derived cell lines as well as patient plasma cell leukemia cells. Treatment with anti-Fas (7C11) monoclonal antibody (MoAb) induced apoptosis, evidenced by internucleosomal DNA fragmentation and propidium iodide staining, and was associated with increased expression of c-jun early response gene. We also show that anti-Fas MoAb treatment is associated with activation of stress-activated protein kinase (SAPK) and p38 mitogen-activated protein kinase (MAPK); however, no detectable increase in extracellular signal-regulated kinases (ERK1 and ERK2) activity was observed. Because interleukin-6 (IL-6) is a growth factor for MM cells and inhibits apoptosis induced by dexamethasone and serum starvation, we examined whether IL-6 affects anti-Fas MoAb-induced apoptosis and activation of SAPK or p38 MAPK in MM cells. Culture of MM cells with IL-6 before treatment with anti-Fas MoAb significantly reduced both DNA fragmentation and activation of SAPK, without altering induction of p38 MAPK activity. These results therefore suggest that anti-Fas MoAb-induced apoptosis in MM cells is associated with activation of SAPK, and that IL-6 may both inhibit apoptosis and modulate SAPK activity.
Collapse
|
195
|
Dixon SC, Soriano BJ, Lush RM, Borner MM, Figg WD. Apoptosis: its role in the development of malignancies and its potential as a novel therapeutic target. Ann Pharmacother 1997; 31:76-82. [PMID: 8997471 DOI: 10.1177/106002809703100113] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE To review the current literature regarding the role of apoptosis in the development of malignant cells and how the induction of this pathway could be used in cancer therapy. DATA SOURCE A MEDLINE search of basic science articles pertinent to the understanding of the normal physiologic process of apoptosis was conducted. STUDY SELECTION Because of the rapidly growing literature regarding apoptosis, only articles describing key processes in the biology of the cell and the genetic control of apoptosis were included. DATA SYNTHESIS Apoptosis is imperative for host survival since it discards unwanted, damaged, and atypical cells. The process is therefore implicated in the continuous regulation of development, differentiation, and homeostasis. Furthermore, apoptosis is a response to physiologic and pathologic stresses that disrupt the balanced rates of cell generation and elimination. In a disease such as cancer, there is a lack of equilibrium between the rates of cell division and cell death; agents that promote or suppress apoptosis can manipulate these rates, influencing the anomalous accumulation of neoplastic cells. Pharmacologic manipulation of apoptosis can manipulate these rates, influencing the anomalous accumulation represents a novel approach in targeting malignant cells and has far-reaching implications for new directions in cancer therapy. CONCLUSIONS Apoptosis is a highly organized physiologic mechanism of destroying injured and abnormal cells as well as maintaining homeostasis in multicellular organisms. Both the activation and inhibition of apoptosis are tightly controlled. Pharmacologic manipulation of this pathway is a novel therapeutic target in cancer therapy.
Collapse
Affiliation(s)
- S C Dixon
- Clinical Pharmacokinetics Section, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
196
|
Interleukin-6 Inhibits Fas-Induced Apoptosis and Stress-Activated Protein Kinase Activation in Multiple Myeloma Cells. Blood 1997. [DOI: 10.1182/blood.v89.1.227] [Citation(s) in RCA: 203] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractFas belongs to the family of type-1 membrane proteins that transduce apoptotic signals. In the present studies, we characterized signaling during Fas-induced apoptosis in RPMI-8226 and IM-9 multiple myeloma (MM) derived cell lines as well as patient plasma cell leukemia cells. Treatment with anti-Fas (7C11) monoclonal antibody (MoAb) induced apoptosis, evidenced by internucleosomal DNA fragmentation and propidium iodide staining, and was associated with increased expression of c-jun early response gene. We also show that anti-Fas MoAb treatment is associated with activation of stress-activated protein kinase (SAPK) and p38 mitogen-activated protein kinase (MAPK); however, no detectable increase in extracellular signal-regulated kinases (ERK1 and ERK2) activity was observed. Because interleukin-6 (IL-6) is a growth factor for MM cells and inhibits apoptosis induced by dexamethasone and serum starvation, we examined whether IL-6 affects anti-Fas MoAb-induced apoptosis and activation of SAPK or p38 MAPK in MM cells. Culture of MM cells with IL-6 before treatment with anti-Fas MoAb significantly reduced both DNA fragmentation and activation of SAPK, without altering induction of p38 MAPK activity. These results therefore suggest that anti-Fas MoAb-induced apoptosis in MM cells is associated with activation of SAPK, and that IL-6 may both inhibit apoptosis and modulate SAPK activity.
Collapse
|
197
|
Moore PS, Boshoff C, Weiss RA, Chang Y. Molecular mimicry of human cytokine and cytokine response pathway genes by KSHV. Science 1996; 274:1739-44. [PMID: 8939871 DOI: 10.1126/science.274.5293.1739] [Citation(s) in RCA: 700] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Four virus proteins similar to two human macrophage inflammatory protein (MIP) chemokines, interleukin-6 (IL-6), and interferon regulatory factor (IRF) are encoded by the Kaposi's sarcoma-associated herpesvirus (KSHV) genome. vIL-6 was functional in B9 proliferation assays and primarily expressed in KSHV-infected hematopoietic cells rather than KS lesions. HIV-1 transmission studies showed that vMIP-I is similar to human MIP chemokines in its ability to inhibit replication of HIV-1 strains dependent on the CCR5 co-receptor. These viral genes may form part of the response to host defenses contributing to virus-induced neoplasia and may have relevance to KSHV and HIV-I interactions.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cell Division
- Cell Line
- Chemokine CCL4
- Gene Expression
- Genes, Viral
- HIV-1/physiology
- Herpesvirus 4, Human/physiology
- Herpesvirus 8, Human/genetics
- Herpesvirus 8, Human/physiology
- Humans
- Interleukin-6/chemistry
- Interleukin-6/genetics
- Lymph Nodes/virology
- Lymphoma, B-Cell/virology
- Macrophage Inflammatory Proteins/chemistry
- Macrophage Inflammatory Proteins/genetics
- Mice
- Molecular Mimicry
- Molecular Sequence Data
- Receptors, CCR5
- Receptors, Cytokine/metabolism
- Receptors, HIV/metabolism
- Sarcoma, Kaposi/virology
- Sequence Alignment
- Signal Transduction
- Tetradecanoylphorbol Acetate/pharmacology
- Tumor Cells, Cultured
- Viral Nonstructural Proteins/chemistry
- Viral Nonstructural Proteins/genetics
- Viral Nonstructural Proteins/physiology
- Virus Replication
Collapse
Affiliation(s)
- P S Moore
- School of Public Health, Columbia University, New York, NY 10032, USA
| | | | | | | |
Collapse
|
198
|
Morris SM, Domon OE, McGarrity LJ, Chen JJ, Manjanatha MG, Andrews AM, Aidoo A, Casciano DA. A role for apoptosis in the toxicity and mutagenicity of bleomycin in AHH-1 tk+/- human lymphoblastoid cells. Mutat Res 1996; 357:143-65. [PMID: 8876690 DOI: 10.1016/0027-5107(96)00095-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The chromosomal mutagen, bleomycin, is also noted for its toxic properties, although the mechanism of cell death is not fully understood. In order to determine if cell death occurred by apoptosis or necrosis, AHH-1 tk+/- cells were exposed to bleomycin and the percentage of viable, apoptotic and necrotic cells quantified by flow cytometry. Logistic regression analysis indicated that the primary manner of cell death was through the apoptosis pathways, that apoptosis was delayed, and that apoptosis was accompanied by an arrest in the G2 phase of the cell cycle. Once apoptosis was established as a mechanism for cell death, the efficiency with which these pathways removed damaged cells from the population was evaluated with the use of specific-locus mutation assays (tk and hprt) as indicators of cells with DNA damage that maintained viability and clonogenicity. Linear regression analysis detected a significant, concentration-dependent increase in the numbers of TFTr clones with the slow-growth phenotype. This suggests that a proportion of cells with bleomycin-induced DNA damage did not undergo cell death by apoptosis and that apoptosis, a mechanism for the destruction of damaged cells, is not fully efficient in the AHH-1 tk +/- cell line.
Collapse
Affiliation(s)
- S M Morris
- Department of Health and Human Services, Food and Drug Administration, Jefferson, AR 72079, USA.
| | | | | | | | | | | | | | | |
Collapse
|
199
|
To LB, Horvath N, Dyson P, Henry J, Sykes P, Brisco M, Morley A, Bennetts B. Myeloma stem cells in autografting in multiple myeloma. JOURNAL OF HEMATOTHERAPY 1996; 5:557-9. [PMID: 8938529 DOI: 10.1089/scd.1.1996.5.557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Autologous transplantation has been used increasingly over the last 10 years for the treatment of multiple myeloma. As is the case in other cancers treated by high-dose therapy and stem cell rescue, the contribution of occult tumor cells in the graft to relapse posttransplant remains to be resolved. In this report, we review the biology and differentiation of plasma cells in the context of their significance as an origin of relapse in multiple myeloma.
Collapse
Affiliation(s)
- L B To
- Division of Haematology, Hanson Centre for Cancer Research, Adelaide, S. Australia
| | | | | | | | | | | | | | | |
Collapse
|
200
|
Lai CF, Ripperger J, Morella KK, Jurlander J, Hawley TS, Carson WE, Kordula T, Caligiuri MA, Hawley RG, Fey GH, Baumann H. Receptors for interleukin (IL)-10 and IL-6-type cytokines use similar signaling mechanisms for inducing transcription through IL-6 response elements. J Biol Chem 1996; 271:13968-75. [PMID: 8662928 DOI: 10.1074/jbc.271.24.13968] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The cytoplasmic domain of the receptor for interleukin 10 (IL-10R) contains two box 3 sequence motifs that have been identified in the signal-transducing receptor subunits for IL-6-type cytokines and noted to be required for activating STAT3 and inducing transcription through IL-6-responsive elements. To determine whether the IL-10R has signaling functions similar to IL-6R in cells normally expressing these receptors, leukocytes of the B-, T-, and NK-cell lineages were treated with either cytokine. Both cytokines activated factors that bound to the sis-inducible element and included STAT1 and STAT3. The cell response to IL-10 characteristically differed from that to IL-2/IL-15, IL-4, and interferon gamma. The signaling capabilities of the IL-10R for activating specific STAT proteins and inducing gene transcription were defined by reconstitution of receptor functions in transfected tissue culture cells. COS-1 cells, co-expressing the human IL-10R and individual STAT proteins, confirmed a preference of the IL-10R for STAT3 and STAT1. Unlike many hematopoietin receptors, the IL-10R did not detectably activate STAT5. The IL-10R, together with reporter gene constructs containing different IL-6-responsive gene elements, reconstituted in hepatoma cells an induction of transcription by IL-10 that was comparable to that by IL-6. This regulation could not be appreciably modified by enhanced expression of STAT proteins. The similar actions of IL-10R and IL-6R on the induction of endogenous IL-6-responsive genes were demonstrated in hepatoma cells stably expressing the IL-10R. These receptor functions required the presence of the box 3 motifs, as shown by the analysis of the mouse IL-10R constructs containing progressively truncated cytoplasmic domains. The data demonstrate that the IL-10R, unlike other members of the interferon receptor family, is highly effective in recruiting the signaling pathways of IL-6-type cytokine receptors.
Collapse
MESH Headings
- Acute-Phase Proteins/metabolism
- Amino Acid Sequence
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/physiology
- Base Sequence
- Binding Sites
- Carcinoma, Hepatocellular
- Cell Line
- Chlorocebus aethiops
- DNA-Binding Proteins/metabolism
- Humans
- Interleukin-10/pharmacology
- Interleukin-6/pharmacology
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukocytes/metabolism
- Liver Neoplasms
- Mice
- Molecular Sequence Data
- Oligodeoxyribonucleotides
- Receptors, Interleukin/biosynthesis
- Receptors, Interleukin/physiology
- Receptors, Interleukin-10
- Receptors, Interleukin-6
- Recombinant Proteins/metabolism
- STAT3 Transcription Factor
- Signal Transduction/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/physiology
- Trans-Activators/metabolism
- Transcription, Genetic/drug effects
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- C F Lai
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|