Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Genet ; 38(11): 1269-77, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17041602

ABSTRACT

Knockdown of the transcription factor PU.1 (encoded by Sfpi1) leads to acute myeloid leukemia (AML) in mice. We examined the transcriptome of preleukemic hematopoietic stem cells (HSCs) in which PU.1 was knocked down (referred to as 'PU.1-knockdown HSCs') to identify transcriptional changes preceding malignant transformation. Transcription factors c-Jun and JunB were among the top-downregulated targets. Restoration of c-Jun expression in preleukemic cells rescued the PU.1 knockdown-initiated myelomonocytic differentiation block. Lentiviral restoration of JunB at the leukemic stage led to loss of leukemic self-renewal capacity and prevented leukemia in NOD-SCID mice into which leukemic PU.1-knockdown cells were transplanted. Examination of human individuals with AML confirmed the correlation between PU.1 and JunB downregulation. These results delineate a transcriptional pattern that precedes leukemic transformation in PU.1-knockdown HSCs and demonstrate that decreased levels of c-Jun and JunB contribute to the development of PU.1 knockdown-induced AML by blocking differentiation and increasing self-renewal. Therefore, examination of disturbed gene expression in HSCs can identify genes whose dysregulation is essential for leukemic stem cell function and that are targets for therapeutic interventions.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins c-jun/physiology , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Animals , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Down-Regulation , Granulocytes/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Monocytes/cytology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Transduction, Genetic
2.
Blood ; 117(17): 4409-19, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21321360

ABSTRACT

Recurrence of multiple myeloma (MM) after therapy suggests the presence of tumor-initiating subpopulations. In our study, we performed flow cytometry-based Hoechst 33342 staining to evaluate the existence of a MM population with stem-like features known as side population (SP) cells. SP cells exhibit substantial heterogeneity in MM cell lines and primary MM cells; express CD138 antigen in MM cell lines; display higher mRNA expression and functional activity of ABCG2 transporter; and have a higher proliferation index compared with non-SP cells. We observed evidence for clonogenic potential of SP cells, as well as the ability of SP cells to regenerate original population. Moreover, SP cells revealed higher tumorigenicity compared with non-SP cells. Importantly, lenalidomide decreased the percentage and clonogenicity of SP cells, and also induced phosphorylation changes in Akt, GSK-3α/ß, MEK1, c-Jun, p53, and p70S6K in SP cells. Adherence to bone marrow stromal cells (BMSCs) increased the percentage, viability, and proliferation potential of SP cells. Lenalidomide and thalidomide abrogated this stimulatory effect of BMSCs and significantly decreased the percentage of SP cells. Our studies demonstrate a novel mechanism of action for lenalidomide, namely targeting SP fraction, providing the framework for new therapeutic strategies targeting subpopulations of MM cells including presumptive stem cells.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma , Neoplastic Stem Cells/drug effects , Thalidomide/analogs & derivatives , ATP-Binding Cassette Transporters/genetics , Angiogenesis Inhibitors/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Division/drug effects , Cell Fractionation , Cell Line, Tumor , Cell Survival/drug effects , Colony-Forming Units Assay , Drug Resistance, Neoplasm , Humans , Lenalidomide , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Multiple Myeloma/physiopathology , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/physiology , Syndecan-1/metabolism , Thalidomide/pharmacology
3.
Br J Haematol ; 159(3): 340-51, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22970818

ABSTRACT

Jasmonates, plant stress hormones, have been demonstrated to be effective in killing various types of cancer cells. We therefore tested if methyljasmonate (MJ) has activity against multiple myeloma (MM) in vitro and in vivo. MM cell lines and primary MM tumour cells responded to MJ in vitro at concentrations that did not significantly affect normal haematopoietic cells, without stroma-mediated resistance. Brief MJ exposures of MM cells caused release of Hexokinase 2 (HK2) from mitochondria, rapid ATP depletion, perturbation of major intracellular signalling pathways, and ensuing mainly apoptotic cell death. Sensitivity to MJ correlated with lower cellular glucose consumption and lactate production, as well as lower intracellular protein levels of HK2, phosphorylated Voltage-dependent anion channel 2/3 (pVDAC2/3) and Aldo-keto reductase family 1 member C1 (AKR1C1), which represent potential biomarkers of responsiveness to MJ treatment, especially as AKR1C1 transcript levels also correlate with clinical outcome in bortezomib- or dexamethasone-treated MM patients. Interestingly, MJ synergized with bortezomib in vitro and prolonged survival of immunocompromised mice harbouring diffuse lesions of MM.1S cells compared to vehicle-treated mice (P = 0·0046). These studies indicate that jasmonates represent a new, promising strategy to treat MM.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cyclopentanes/pharmacology , Multiple Myeloma/metabolism , Oxylipins/pharmacology , Plant Growth Regulators/pharmacology , Acetates/administration & dosage , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclopentanes/administration & dosage , Drug Synergism , Humans , Leukocytes, Mononuclear/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Oxylipins/administration & dosage
4.
Br J Haematol ; 157(6): 718-31, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22533681

ABSTRACT

Multiple Myeloma (MM), a malignancy of plasma cells, remains incurable despite the use of conventional and novel therapies. Halofuginone (HF), a synthetic derivative of quinazolinone alkaloid, has recently been shown to have anti-cancer activity in various preclinical settings. This study demonstrated the anti-tumour activity of HF against a panel of human MM cell lines and primary patient-derived MM cells, regardless of their sensitivity to conventional therapy or novel agents. HF showed anti-MM activity in vivo using a myeloma xenograft mouse model. HF suppressed proliferation of myeloma cells alone and when co-cultured with bone marrow stromal cells. Similarly, HF induced apoptosis in MM cells even in the presence of insulin-like growth factor 1 or interleukin 6. Importantly, HF, even at high doses, did not induce cytotoxicity against CD40 activated peripheral blood mononuclear cells from normal donors. HF treatment induced accumulation of cells in the G(0) /G(1) cell cycle and induction of apoptotic cell death associated with depletion of mitochondrial membrane potential; cleavage of poly (ADP-ribose) polymerase and caspases-3, 8 and 9 as well as down-regulation of anti-apoptotic proteins including Mcl-1 and X-IAP. Multiplex analysis of phosphorylation of diverse components of signalling cascades revealed that HF induced changes in P38MAPK activation; increased phosphorylation of c-jun, c-jun NH(2)-terminal kinase (JNK), p53 and Hsp-27. Importantly, HF triggered synergistic cytotoxicity in combination with lenalidomide, melphalan, dexamethasone, and doxorubicin. Taken together, these preclinical studies provide the preclinical framework for future clinical studies of HF in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , G1 Phase/drug effects , Multiple Myeloma/drug therapy , Piperidines/pharmacology , Quinazolinones/pharmacology , Resting Phase, Cell Cycle/drug effects , Animals , Antineoplastic Agents/agonists , Antineoplastic Agents/therapeutic use , Caspases/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, SCID , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/drug effects , Piperidines/agonists , Piperidines/therapeutic use , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinazolinones/agonists , Quinazolinones/therapeutic use , Tumor Suppressor Protein p53/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Haematologica ; 96(8): 1170-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21712538

ABSTRACT

BACKGROUND: Isothiocyanates, a family of phytochemicals found in cruciferous vegetables, have cytotoxic effects against several types of tumor cells. Multiple myeloma is a fatal disease characterized by clonal proliferation of plasma cells in the bone marrow. The growing body of preclinical information on the anti-cancer activity of isothiocyanates led us to investigate their anti-myeloma properties. DESIGN AND METHODS: We evaluated the anti-myeloma activity of the isothiocyanates, sulforaphane and phenethyl isothiocyanate, on a panel of human myeloma cell lines as well as primary myeloma tumor cells. Cell viability, apoptosis, cell cycle alterations and cell proliferation were then analyzed in vitro and in a xenograft mouse model in vivo. The molecular sequelae of isothiocyanate treatment in multiple myeloma cells were evaluated by multiplex analyses using bead arrays and western blotting. RESULTS: We observed that sulforaphane and phenylethyl isothiocyanate have activity against myeloma cell lines and patients' myeloma cells both in vitro and in vivo using a myeloma xenograft mouse model. Isothiocyanates induced apoptotic death of myeloma cells; depletion of mitochondrial membrane potential; cleavage of PARP and caspases-3 and -9; as well as down-regulation of anti-apoptotic proteins including Mcl-1, X-IAP, c-IAP and survivin. Isothiocyanates induced G(2)/M cell cycle arrest accompanied by mitotic phosphorylation of histone H3. Multiplex analysis of phosphorylation of diverse components of signaling cascades revealed changes in MAPK activation; increased phosphorylation of c-jun and HSP27; as well as changes in the phosphorylation of Akt, and GSK3α/ß and p53. Isothiocyanates suppressed proliferation of myeloma cells alone and when co-cultured with HS-5 stromal cells. Sulforaphane and phenylethyl isothiocyanate enhanced the in vitro anti-myeloma activity of several conventional and novel therapies used in multiple myeloma. CONCLUSIONS: Our study shows that isothiocyanates have potent anti-myeloma activities and may enhance the activity of other anti-multiple myeloma agents. These results indicate that isothiocyanates may have therapeutic potential in multiple myeloma and provide the preclinical framework for future clinical studies of isothiocyanates in multiple myeloma.


Subject(s)
Antineoplastic Agents/pharmacology , Isothiocyanates/pharmacology , Multiple Myeloma/drug therapy , Signal Transduction/drug effects , Thiocyanates/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Isothiocyanates/therapeutic use , Isothiocyanates/toxicity , Mice , Mice, SCID , Multiple Myeloma/metabolism , Stromal Cells/drug effects , Sulfoxides , Thiocyanates/therapeutic use , Thiocyanates/toxicity , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Semin Hematol ; 46(2): 166-75, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19389500

ABSTRACT

The successful clinical development of thalidomide, bortezomib, and lenalidomide not only transformed the therapeutic management of multiple myeloma (MM) but also catalyzed a renewed interest in the development of additional classes of novel agents for this disease. This review focuses on a series of new therapeutics that have shown promising preclinical results, as well as encouraging safety profiles and early evidence of anti-MM activity in clinical studies, either alone or in combination with other, conventional or novel, anti-MM treatments. These agents include second-generation proteasome inhibitors and immunomodulatory agents, as well as members of other therapeutic classes, such as histone deacetylase inhibitors (HDAC), heat shock protein 90 (Hsp90) inhibitors, and the alkylphospholipid Akt inhibitor perifosine.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Multiple Myeloma/drug therapy , Animals , Boronic Acids/therapeutic use , Bortezomib , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Immunologic Factors/therapeutic use , Multiple Myeloma/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/therapeutic use
7.
Br J Haematol ; 147(5): 672-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19751238

ABSTRACT

This study characterized the preclinical anti-myeloma activity of VE465, a low molecular weight pan-Aurora kinase inhibitor. After 96-h drug exposure, several multiple myeloma (MM) cell lines were more sensitive to VE465 compared to non-malignant cells. The anti-MM activity of VE465 was maintained in the presence of interleukin-6 and, interestingly, enhanced by co-culture with stromal cells. However, primary MM cells were less responsive than cell lines. Combinations with dexamethasone (Dex), doxorubicin (Doxo) and bortezomib showed no antagonism. Our study highlights the potential role of the tumour microenvironment in modulating the activity of this drug class.


Subject(s)
Antineoplastic Agents/pharmacology , Multiple Myeloma/pathology , Piperazines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Aurora Kinases , Cell Communication/drug effects , Cell Cycle/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Hematopoietic Stem Cells/cytology , Humans , Tumor Cells, Cultured
9.
Hematol Oncol Clin North Am ; 21(6): 1007-34, vii-viii, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17996586

ABSTRACT

Multiple myeloma (MM) is viewed as a prototypic disease state for the study of how neoplastic cells interact with their local bone marrow (BM) microenvironment. This interaction reflects not only the osteo-tropic clinical behavior of MM and the clinical impact of the lytic bone lesions caused by its tumor cells but also underlines the broadly accepted notion that nonneoplastic cells of the BM can attenuate the activity of cytotoxic chemotherapy and glucocorticoids. This article summarizes the recent progress in characterization, at the molecular and cellular levels, of how the BM milieu interacts with MM cells and modifies their biologic behavior.


Subject(s)
Bone Marrow/physiology , Cell Communication/physiology , Multiple Myeloma/physiopathology , Humans , Multiple Myeloma/therapy , Osteoclasts/physiology , Stromal Cells/physiology
10.
Cancer Chemother Pharmacol ; 71(5): 1357-68, 2013 May.
Article in English | MEDLINE | ID: mdl-23589314

ABSTRACT

PURPOSE: Bortezomib is an important agent in multiple myeloma treatment, but resistance in cell lines and patients has been described. The main mechanisms of resistance described in cancer fall into one of two categories, pharmacokinetic resistance (PK), e.g. over expression of drug efflux pumps and pharmacodynamic resistance, e.g. apoptosis resistance or altered survival pathways, where the agent reaches an appropriate concentration, but this fails to propagate an appropriate cell death response. Of the known pump mechanisms, P-glycoprotein (P-gp) is the best studied and considered to be the most important in contributing to general PK drug resistance. Resistance to bortezomib is multifactorial and there are conflicting indications that cellular overexpression of P-gp may contribute to resistance agent. Hence, better characterization of the interactions of this drug with classical resistance mechanisms should identify improved treatment applications. METHODS: Cell lines with different P-gp expression levels were used to determine the relationship between bortezomib and P-gp. Coculture system with stromal cells was used to determine the effect of the local microenvironment on the bortezomib-elacridar combination. To further assess P-gp function, intracellular accumulation of P-gp probe rhodamine-123 was utilised. RESULTS: In the present study, we show that bortezomib is a substrate for P-gp, but not for the other drug efflux transporters. Bortezomib activity is affected by P-gp expression and conversely, the expression of P-gp affect bortezomib's ability to act as a P-gp substrate. The local microenvironment did not alter the cellular response to bortezomib. We also demonstrate that bortezomib directly affects the expression and function of P-gp. CONCLUSIONS: Our findings strongly support a role for P-gp in bortezomib resistance and, therefore, suggest that combination of a P-gp inhibitor and bortezomib in P-gp positive myeloma would be a reasonable treatment combination to extend efficacy of this important drug.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Drug Resistance, Neoplasm , Multiple Myeloma/drug therapy , Pyrazines/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Biological Transport , Bortezomib , Cell Line , Cell Line, Tumor , Cellular Microenvironment , Coculture Techniques , Fluorescent Dyes/pharmacokinetics , Gene Expression Regulation/drug effects , Humans , Multiple Myeloma/pathology , Rhodamine 123/pharmacokinetics , Stromal Cells/metabolism
11.
PLoS One ; 6(7): e20226, 2011.
Article in English | MEDLINE | ID: mdl-21750699

ABSTRACT

Polo-like kinases (PLKs) play an important role in cell cycle progression, checkpoint control and mitosis. The high mitotic index and chromosomal instability of advanced cancers suggest that PLK inhibitors may be an attractive therapeutic option for presently incurable advanced neoplasias with systemic involvement, such as multiple myeloma (MM). We studied the PLK 1, 2, 3 inhibitor BI 2536 and observed potent (IC50<40 nM) and rapid (commitment to cell death <24 hrs) in vitro activity against MM cells in isolation, as well as in vivo activity against a traditional subcutaneous xenograft mouse model. Tumor cells in MM patients, however, don't exist in isolation, but reside in and interact with the bone microenvironment. Therefore conventional in vitro and in vivo preclinical assays don't take into account how interactions between MM cells and the bone microenvironment can potentially confer drug resistance. To probe this question, we performed tumor cell compartment-specific bioluminescence imaging assays to compare the preclinical anti-MM activity of BI 2536 in vitro in the presence vs. absence of stromal cells or osteoclasts. We observed that the presence of these bone marrow non-malignant cells led to decreased anti-MM activity of BI 2536. We further validated these results in an orthotopic in vivo mouse model of diffuse MM bone lesions where tumor cells interact with non-malignant cells of the bone microenvironment. We again observed that BI 2536 had decreased activity in this in vivo model of tumor-bone microenvironment interactions highlighting that, despite BI 2536's promising activity in conventional assays, its lack of activity in microenvironmental models raises concerns for its clinical development for MM. More broadly, preclinical drug testing in the absence of relevant tumor microenvironment interactions may overestimate potential clinical activity, thus explaining at least in part the gap between preclinical vs. clinical efficacy in MM and other cancers.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Xenograft Model Antitumor Assays , Blotting, Western , Bone and Bones/drug effects , Bone and Bones/pathology , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Osteoclasts/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Stromal Cells/drug effects , Time Factors , Tumor Burden/drug effects , Tumor Microenvironment/drug effects , Polo-Like Kinase 1
12.
Nat Med ; 16(4): 483-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20228816

ABSTRACT

Conventional anticancer drug screening is typically performed in the absence of accessory cells of the tumor microenvironment, which can profoundly alter antitumor drug activity. To address this limitation, we developed the tumor cell-specific in vitro bioluminescence imaging (CS-BLI) assay. Tumor cells (for example, myeloma, leukemia and solid tumors) stably expressing luciferase are cultured with nonmalignant accessory cells (for example, stromal cells) for selective quantification of tumor cell viability, in presence versus absence of stromal cells or drug treatment. CS-BLI is high-throughput scalable and identifies stroma-induced chemoresistance in diverse malignancies, including imatinib resistance in leukemic cells. A stroma-induced signature in tumor cells correlates with adverse clinical prognosis and includes signatures for activated Akt, Ras, NF-kappaB, HIF-1alpha, myc, hTERT and IRF4; for biological aggressiveness; and for self-renewal. Unlike conventional screening, CS-BLI can also identify agents with increased activity against tumor cells interacting with stroma. One such compound, reversine, shows more potent activity in an orthotopic model of diffuse myeloma bone lesions than in conventional subcutaneous xenografts. Use of CS-BLI, therefore, enables refined screening of candidate anticancer agents to enrich preclinical pipelines with potential therapeutics that overcome stroma-mediated drug resistance and can act in a synthetic lethal manner in the context of tumor-stroma interactions.


Subject(s)
Drug Screening Assays, Antitumor/methods , Luminescent Measurements/methods , Stromal Cells/metabolism , Tumor Cells, Cultured/drug effects , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Humans , Morpholines/pharmacology , Purines/pharmacology , Stromal Cells/drug effects
13.
Pathol Biol (Paris) ; 52(5): 267-74, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15217712

ABSTRACT

Polycythemia vera, essential thrombocythemia, idiopathic myelofibrosis and chronic myelogenous leukemia have been collectively termed the myeloproliferative disorders due to similarities in their clinical presentation. With the exception of chronic myelogenous leukemia, which is characterized by the presence of the Philadelphia chromosome, the myeloproliferative disorders display no consistent cytogenetic abnormalities. Hence, the diagnosis of Polycythemia vera, essential thrombocythemia and idiopathic myelofibrosis to date relies on clinical criteria. However, several molecular aberrations have been described, which can be used as molecular markers for the diagnosis of these clinical entities. This review outlines the diagnostic assays developed and highlights the advantages and disadvantages of the following markers: (1). Endogenous Erythroid Colonies, (2). Clonality, (3). Reduced c-Mpl protein expression and (4). PRV-1 mRNA over expression.


Subject(s)
Genetic Markers , Myeloproliferative Disorders/genetics , Philadelphia Chromosome , Humans , Myeloproliferative Disorders/classification
14.
Blood ; 100(7): 2441-8, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12239154

ABSTRACT

The cDNA for polycythemia rubra vera 1 (PRV-1), a novel hematopoietic receptor, was recently cloned by virtue of its overexpression in patients with polycythemia vera. PRV-1 is a member of the uPAR/CD59/Ly6 family of cell surface receptors, which share a common cysteine-rich domain and are tethered to the cell surface via a glycosylphosphatidylinositol (GPI) link. We have determined the intron-exon structure of the PRV1 gene and show that the locus is structurally intact in patients with polycythemia vera. Thus, PRV-1 overexpression in these patients is not due to rearrangement or structural alteration of the gene. Northern blot analysis detects multiple PRV-1 transcripts. Here we show that these transcripts arise from alternative polyadenylation and encode the same protein. Biochemical analysis reveals that PRV-1 is N-glycosylated and embedded in the cell membrane by a lipid anchor, like other members of this family. Moreover, PRV-1 is shed from the cell surface because soluble protein can be detected in cell supernatants. Fluorescence-activated cell sorting analysis of stably transfected cells revealed that PRV-1 is recognized by antibodies directed against the neutrophil antigen NB1/CD177. Flow cytometry of bone marrow and peripheral blood of both healthy donors and patients with polycythemia vera showed that PRV-1 protein is expressed on myeloid cells of the granulocytic lineage. However, unlike the significant difference in PRV-1 expression observed on the mRNA level, the amount of PRV-1 protein on the cell surface is not consistently elevated in patients with polycythemia vera compared with healthy controls. Therefore, quantification of PRV-1 surface expression cannot be used for the diagnosis of polycythemia vera.


Subject(s)
Hematopoietic Stem Cells/physiology , Polycythemia Vera/blood , Polycythemia Vera/genetics , Receptors, Cell Surface/genetics , 3' Untranslated Regions/genetics , Blotting, Southern , Cell Membrane/physiology , DNA Primers , GPI-Linked Proteins , Glycoside Hydrolases , Glycosylphosphatidylinositols/metabolism , Humans , Isoantigens , Membrane Glycoproteins , RNA, Messenger/genetics , Receptors, Cell Surface/metabolism , Restriction Mapping , Transcription, Genetic
15.
Blood ; 102(10): 3569-74, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12893745

ABSTRACT

To date, the diagnosis of polycythemia vera (PV) relies on clinical criteria. We have recently described the overexpression of a hematopoietic receptor, polycythemia rubra vera-1 (PRV-1), in patients with PV. Here, we report a quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) assay for the measurement of PRV-1 mRNA levels. We have determined PRV-1 expression in 71 patients with PV, 11 patients with secondary erythrocytosis (SE), as well as in 80 healthy controls. PV patients express significantly higher amounts of PRV-1 than healthy controls or patients with SE (P <.0001). Because there is no overlap between the PRV-1 expression in PV patients versus healthy controls or SE patients, the assay has a very high sensitivity and specificity for the diagnosis of PV in our population. In patients with erythrocytosis, the quantitative RT-PCR assay described here therefore provides a rapid, highly specific and sensitive tool for the diagnosis of PV.


Subject(s)
Polycythemia Vera/diagnosis , Polycythemia/diagnosis , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Diagnosis, Differential , Female , GPI-Linked Proteins , Humans , Isoantigens , Male , Membrane Glycoproteins , Middle Aged , Molecular Diagnostic Techniques , Polycythemia/etiology , Polycythemia Vera/chemically induced , RNA, Messenger/analysis , Receptors, Cell Surface/analysis , Receptors, Cell Surface/genetics , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/standards , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL