Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Biol Macromol ; 269(Pt 2): 132141, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723809

ABSTRACT

To enhance the flame retardancy and mechanical performance of PLA, a polyelectrolyte complex predicated on lignin was obtained by electrostatic mutual adsorption of ammonium polyphosphate (APP), polyethyleneimine (PEI), and copper ions as raw materials. The FT-IR spectra and EDX analysis confirmed the successful synthesis of a lignin-based flame retardant hybrid (APL-Cu2+) containing copper, phosphorus, and nitrogen elements. The combustion test results showed that the peak heat release rate and total heat release of the PLA composite containing 12 wt% APL-Cu2+ were decreased by 15.1 % and 18.2 %, respectively, as compared to those of pure PLA. The char residue morphology observation revealed that the addition of APL-Cu2+ could promote the formation of a highly dense and stable graphitized char layer, while TG-MS detected the emission of refractory gases such as ammonia gas, carbon dioxide, and water during combustion. The strong hydrogen bonding between APL-Cu2+ and the PLA matrix kept the composite maintaining good strength and toughness. The tensile strength and impact strength of PLA/6APL-Cu2+ increased by 4.73 % and 65.71 %, respectively, due to its high crystallinity and good interfacial compatibility. This work provides a feasible method to develop biobased flame retardant hybrids for PLA composites with better fire safety and improved mechanical properties.


Subject(s)
Copper , Flame Retardants , Lignin , Polyesters , Lignin/chemistry , Copper/chemistry , Polyesters/chemistry , Fires/prevention & control , Mechanical Phenomena , Tensile Strength , Spectroscopy, Fourier Transform Infrared , Polyethyleneimine/chemistry
2.
Cancer Res ; 75(20): 4384-4397, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26249174

ABSTRACT

Despite recent therapeutic advances that have doubled the median survival time of patients with multiple myeloma, intratumor genetic heterogeneity contributes to disease progression and emergence of drug resistance. miRNAs are noncoding small RNAs that play important roles in the regulation of gene expression and have been implicated in cancer progression and drug resistance. We investigated the role of the miR-221-222 family in dexamethasone-induced drug resistance in multiple myeloma using the isogenic cell lines MM1R and MM1S, which represent models of resistance and sensitivity, respectively. Analysis of array comparative genome hybridization data revealed gain of chromosome X regions at band p11.3, wherein the miR-221-222 resides, in resistant MM1R cells but not in sensitive MM1S cells. DNA copy number gains in MM1R cells were associated with increased miR-221-222 expression and downregulation of p53-upregulated modulator of apoptosis (PUMA) as a likely proapoptotic target. We confirmed PUMA mRNA as a direct target of miR-221-222 in MM1S and MM1R cells by both gain-of-function and loss-of-function studies. In addition, miR-221-222 treatment rendered MM1S cells resistant to dexamethasone, whereas anti-miR-221-222 partially restored the dexamethasone sensitivity of MM1R cells. These studies have uncovered a role for miR-221-222 in multiple myeloma drug resistance and suggest a potential therapeutic role for inhibitors of miR-221-222 binding to PUMA mRNA as a means of overcoming dexamethasone resistance in patients. The clinical utility of this approach is predicated on the ability of antisense miR-221-222 to increase survival while reducing tumor burden and is strongly supported by the metastatic propensity of MM1R cells in preclinical mouse xenograft models of multiple myeloma. Moreover, our observation of increased levels of miR-221-222 with decreased PUMA expression in multiple myeloma cells from patients at relapse versus untreated controls suggests an even broader role for miR-221-222 in drug resistance and provides a rationale for the targeting of miR-221-222 as a means of improving patient outcomes.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Dexamethasone/pharmacology , Drug Resistance, Neoplasm/genetics , MicroRNAs/genetics , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Proto-Oncogene Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , 3' Untranslated Regions , Animals , Apoptosis Regulatory Proteins/genetics , Binding Sites , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Neoplasm Metastasis , Proto-Oncogene Proteins/genetics , RNA Interference , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
3.
Br J Haematol ; 166(5): 677-89, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24801987

ABSTRACT

In Waldenström macroglobulinaemia (WM), the mechanism(s) responsible for repression of B-cell differentiation remains unknown. We found that expression of SPIB and ID2 were significantly increased and decreased, respectively, in WM lymphoplasmacytic cells (LPC). Ectopic expression of SPIB in healthy donor CD19(+) cells inhibited plasmacytic differentiation in conjunction with decreased transcription of IRF4 and XBP1 spliced form. In primary WM LPC, knock-down of SPIB induced plasmacytic differentiation in conjunction with increased transcription of PRDM1, XBP1 spliced form, IRF4 and ID2. Knock-down of SPIB also led to decreased BCL2 expression. Given that SPIB is a direct target of POU2AF1 (OBF1) in complex with POU2F2 or POU2F1, we next examined their expression in WM LPC. POU2F2 transcription, as well as POU2F2 and POU2AF1 protein expression was higher in WM LPC. Ectopic expression of POU2F2 in healthy donor CD19(+) cells induced transcription of SPIB and suppressed transcription of PRDM1 and IRF4. Chromatin immunoprecipitation analysis in BCWM.1 WM cells confirmed binding of POU2F2 and POU2AF1 in SPIB and ID2 promoters. These findings establish a molecular hierarchy among POU2F2, SPIB and ID2 during B-cell differentiation, and suggest that aberrant expression of these transcription factors plays an important role in arresting plasmacytic differentiation in WM.


Subject(s)
DNA-Binding Proteins/biosynthesis , Plasma Cells/physiology , Transcription Factors/biosynthesis , Waldenstrom Macroglobulinemia/blood , Waldenstrom Macroglobulinemia/genetics , Aged , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Female , Gene Knockdown Techniques , Humans , Inhibitor of Differentiation Protein 2/biosynthesis , Inhibitor of Differentiation Protein 2/genetics , Male , Middle Aged , Octamer Transcription Factor-2/biosynthesis , Octamer Transcription Factor-2/genetics , Plasma Cells/cytology , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors/genetics
4.
Blood ; 123(11): 1637-46, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24366360

ABSTRACT

The genetic basis for Waldenström macroglobulinemia (WM) remains to be clarified. Although 6q losses are commonly present, recurring gene losses in this region remain to be defined. We therefore performed whole genome sequencing (WGS) in 30 WM patients, which included germline/tumor sequencing for 10 patients. Validated somatic mutations occurring in >10% of patients included MYD88, CXCR4, and ARID1A that were present in 90%, 27%, and 17% of patients, respectively, and included the activating mutation L265P in MYD88 and warts, hypogammaglobulinemia, infection, and myelokathexis-syndrome-like mutations in CXCR4 that previously have only been described in the germline. WGS also delineated copy number alterations (CNAs) and structural variants in the 10 paired patients. The CXCR4 and CNA findings were validated in independent expansion cohorts of 147 and 30 WM patients, respectively. Validated gene losses due to CNAs involved PRDM2 (93%), BTG1 (87%), HIVEP2 (77%), MKLN1 (77%), PLEKHG1 (70%), LYN (60%), ARID1B (50%), and FOXP1 (37%). Losses in PLEKHG1, HIVEP2, ARID1B, and BCLAF1 constituted the most common deletions within chromosome 6. Although no recurrent translocations were observed, in 2 patients deletions in 6q corresponded with translocation events. These studies evidence highly recurring somatic events, and provide a genomic basis for understanding the pathogenesis of WM.


Subject(s)
Gene Deletion , Genomics , Immunologic Deficiency Syndromes/genetics , Lymphoma, B-Cell/genetics , Mutation/genetics , Myeloid Differentiation Factor 88/genetics , Receptors, CXCR4/genetics , Waldenstrom Macroglobulinemia/genetics , Warts/genetics , Amino Acid Sequence , Bone Marrow/pathology , Chromosome Mapping , Chromosomes, Human, Pair 6/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , DNA-Binding Proteins , Humans , In Situ Hybridization, Fluorescence , Lymphoma, B-Cell/pathology , Molecular Sequence Data , Nuclear Proteins/genetics , Primary Immunodeficiency Diseases , Signal Transduction , Transcription Factors/genetics , Translocation, Genetic , Waldenstrom Macroglobulinemia/pathology
5.
Blood ; 122(7): 1222-32, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23836557

ABSTRACT

Myeloid differentiation factor 88 (MYD88) L265P somatic mutation is highly prevalent in Waldenström macroglobulinemia (WM) and supports malignant growth through nuclear factor κB (NF-κB). The signaling cascade(s) by which MYD88 L265P promotes NF-κB activation in WM remain unclear. By lentiviral knockdown or use of a MYD88 inhibitor, decreased phosphorylation of the NF-κB gatekeeper IκBα and survival occurred in MYD88 L265P-expressing WM cells. Conversely, WM cells engineered to overexpress MYD88 L265P showed enhanced survival. Coimmunoprecipitation studies identified Bruton tyrosine kinase (BTK) complexed to MYD88 in L265P-expressing WM cells, with preferential binding of MYD88 to phosphorylated BTK (pBTK). Increased pBTK was also observed in WM cells transduced to overexpress L265P vs wild-type MYD88. Importantly, MYD88 binding to BTK was abrogated following treatment of MYD88 L265P-expressing cells with a BTK kinase inhibitor. Inhibition of BTK or interleukin-1 receptor-associated kinase 1 and 4 (IRAK-1 and -4) kinase activity induced apoptosis of WM cells, and their combination resulted in more robust inhibition of NF-κB signaling and synergistic WM cell killing. The results establish BTK as a downstream target of MYD88 L265P signaling, and provide a framework for the study of BTK inhibitors alone, and in combination with IRAK inhibitors for the treatment of WM.


Subject(s)
Apoptosis , Cell Proliferation , Lymphocytes/pathology , Mutation/genetics , Myeloid Differentiation Factor 88/metabolism , Protein-Tyrosine Kinases/metabolism , Waldenstrom Macroglobulinemia/pathology , Agammaglobulinaemia Tyrosine Kinase , Blotting, Western , Case-Control Studies , Cells, Cultured , Flow Cytometry , Humans , Immunoprecipitation , Interleukin-1 Receptor-Associated Kinases/metabolism , Lentivirus/genetics , Luciferases/metabolism , Lymphocytes/metabolism , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Signal Transduction , Waldenstrom Macroglobulinemia/genetics , Waldenstrom Macroglobulinemia/metabolism
6.
Blood ; 121(11): 2051-8, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23321251

ABSTRACT

By whole-genome and/or Sanger sequencing, we recently identified a somatic mutation (MYD88 L265P) that stimulates nuclear factor κB activity and is present in >90% of Waldenström macroglobulinemia (WM) patients. MYD88 L265P was absent in 90% of immunoglobulin M (IgM) monoclonal gammopathy of undetermined significance (MGUS) patients. We therefore developed conventional and real-time allele-specific polymerase chain reaction (AS-PCR) assays for more sensitive detection and quantification of MYD88 L265P. Using either assay, MYD88 L265P was detected in 97 of 104 (93%) WM and 13 of 24 (54%) IgM MGUS patients and was either absent or rarely expressed in samples from splenic marginal zone lymphoma (2/20; 10%), CLL (1/26; 4%), multiple myeloma (including IgM cases, 0/14), and immunoglobulin G MGUS (0/9) patients as well as healthy donors (0/40; P < 1.5 × 10(-5) for WM vs other cohorts). Real-time AS-PCR identified IgM MGUS patients progressing to WM and showed a high rate of concordance between MYD88 L265P ΔCT and BM disease involvement (r = 0.89, P = .008) in WM patients undergoing treatment. These studies identify MYD88 L265P as a widely present mutation in WM and IgM MGUS patients using highly sensitive and specific AS-PCR assays with potential use in diagnostic discrimination and/or response assessment. The finding of this mutation in many IgM MGUS patients suggests that MYD88 L265P may be an early oncogenic event in WM pathogenesis.


Subject(s)
B-Lymphocytes , Immunoglobulin M , Lymphoproliferative Disorders/genetics , Monoclonal Gammopathy of Undetermined Significance/genetics , Myeloid Differentiation Factor 88/genetics , Polymerase Chain Reaction/methods , Waldenstrom Macroglobulinemia/genetics , Adult , Aged , Alleles , Amino Acid Substitution/physiology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Base Sequence , Case-Control Studies , Cell Transformation, Neoplastic/genetics , DNA Mutational Analysis , Humans , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Leucine/genetics , Lymphoproliferative Disorders/immunology , Male , Middle Aged , Molecular Sequence Data , Monoclonal Gammopathy of Undetermined Significance/immunology , Monoclonal Gammopathy of Undetermined Significance/metabolism , Polymorphism, Single Nucleotide/physiology , Proline/genetics
7.
N Engl J Med ; 367(9): 826-33, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22931316

ABSTRACT

BACKGROUND: Waldenström's macroglobulinemia is an incurable, IgM-secreting lymphoplasmacytic lymphoma (LPL). The underlying mutation in this disorder has not been delineated. METHODS: We performed whole-genome sequencing of bone marrow LPL cells in 30 patients with Waldenström's macroglobulinemia, with paired normal-tissue and tumor-tissue sequencing in 10 patients. Sanger sequencing was used to validate the findings in samples from an expanded cohort of patients with LPL, those with other B-cell disorders that have some of the same features as LPL, and healthy donors. RESULTS: Among the patients with Waldenström's macroglobulinemia, a somatic variant (T→C) in LPL cells was identified at position 38182641 at 3p22.2 in the samples from all 10 patients with paired tissue samples and in 17 of 20 samples from patients with unpaired samples. This variant predicted an amino acid change (L265P) in MYD88, a mutation that triggers IRAK-mediated NF-κB signaling. Sanger sequencing identified MYD88 L265P in tumor samples from 49 of 54 patients with Waldenström's macroglobulinemia and in 3 of 3 patients with non-IgM-secreting LPL (91% of all patients with LPL). MYD88 L265P was absent in paired normal tissue samples from patients with Waldenström's macroglobulinemia or non-IgM LPL and in B cells from healthy donors and was absent or rarely expressed in samples from patients with multiple myeloma, marginal-zone lymphoma, or IgM monoclonal gammopathy of unknown significance. Inhibition of MYD88 signaling reduced IκBα and NF-κB p65 phosphorylation, as well as NF-κB nuclear staining, in Waldenström's macroglobulinemia cells expressing MYD88 L265P. Somatic variants in ARID1A in 5 of 30 patients (17%), leading to a premature stop or frameshift, were also identified and were associated with an increased disease burden. In addition, 2 of 3 patients with Waldenström's macroglobulinemia who had wild-type MYD88 had somatic variants in MLL2. CONCLUSIONS: MYD88 L265P is a commonly recurring mutation in patients with Waldenström's macroglobulinemia that can be useful in differentiating Waldenström's macroglobulinemia and non-IgM LPL from B-cell disorders that have some of the same features. (Funded by the Peter and Helen Bing Foundation and others.).


Subject(s)
Mutation , Myeloid Differentiation Factor 88/genetics , Waldenstrom Macroglobulinemia/genetics , Diagnosis, Differential , Disease Progression , Gene Expression , Genome, Human , Humans , Immunoglobulin M/analysis , Paraproteinemias/diagnosis , Paraproteinemias/immunology , Sequence Analysis, DNA , Waldenstrom Macroglobulinemia/diagnosis , Waldenstrom Macroglobulinemia/immunology
8.
J Virol ; 85(23): 12605-13, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21937657

ABSTRACT

West Nile virus (WNV) is similar to other RNA viruses in that it forms genetically complex populations within hosts. The virus is maintained in nature in mosquitoes and birds, with each host type exerting distinct influences on virus populations. We previously observed that prolonged replication in mosquitoes led to increases in WNV genetic diversity and diminished pathogenesis in mice without remarkable changes to the consensus genome sequence. We therefore sought to evaluate the relationships between individual and group phenotypes in WNV and to discover novel viral determinants of pathogenesis in mice and fitness in mosquitoes and birds. Individual plaque size variants were isolated from a genetically complex population, and mutations conferring a small-plaque and mouse-attenuated phenotype were localized to the RNA helicase domain of the NS3 protein by reverse genetics. The mutation, an Asp deletion, did not alter type I interferon production in the host but rendered mutant viruses more susceptible to interferon compared to wild type (WT) WNV. Finally, we used an in vivo fitness assay in Culex quinquefasciatus mosquitoes and chickens to determine whether the mutation in NS3 influenced fitness. The fitness of the NS3 mutant was dramatically lower in chickens and moderately lower in mosquitoes, indicating that RNA helicase is a major fitness determinant of WNV and that the effect on fitness is host specific. Overall, this work highlights the complex relationships that exist between individual and group phenotypes in RNA viruses and identifies RNA helicase as an attenuation and fitness determinant in WNV.


Subject(s)
Chickens/virology , Culicidae/virology , Genome, Viral , West Nile Fever/pathology , West Nile Fever/parasitology , West Nile virus/genetics , West Nile virus/pathogenicity , Animals , Cells, Cultured , Chickens/genetics , Chlorocebus aethiops , Cricetinae , Culicidae/genetics , Culicidae/pathogenicity , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Genetic Variation , Interferons/metabolism , Kidney/cytology , Kidney/metabolism , Kidney/virology , Mice , Mice, Inbred C3H , Mutation/genetics , RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Viral/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication , West Nile Fever/virology
9.
Clin Lymphoma Myeloma Leuk ; 11(1): 172-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21856553

ABSTRACT

Soluble CD27 (sCD27) is produced by Waldenström's macroglobulinemia (WM) cells, with high levels found in WM patients which may facilitate disease expansion. Matrix metalloproteinases (MMP) may facilitate sCD27 release by cleavage of CD27. By gene expression analysis, we observed significantly higher transcription levels of MMP-8 and MMP-9, with 58.5 and 16.7 fold increase in mean transcription levels in WM cells relative to healthy donor peripheral blood B cells (P = .04, and .05, respectively). We developed a model for study of sCD27 release by transfecting BCWM.1 WM cells and BL2126 lymphoblastic B cells, both of which express MMP-8 and MMP-9 with a vector expressing FLAG-tagged CD27 (pFLAG-CD27) which in the presence of phorbol myristate acetate resulted in ≥ 10-fold increase in sCD27 release. MMP inhibitors against MMP-8, but not MMP 2, 3, or 9 blocked release of sCD27. The results suggest that MMP-8 may play a role in the pathogenesis of WM, and that its inhibition may be of therapeutic value in WM.


Subject(s)
Enzyme Inhibitors/pharmacology , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase Inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Waldenstrom Macroglobulinemia/enzymology , Waldenstrom Macroglobulinemia/genetics , Antigens, CD19/metabolism , Bone Marrow Cells/metabolism , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation , Humans , Matrix Metalloproteinase 9/genetics , Transfection , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
10.
Leuk Lymphoma ; 52(9): 1777-86, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21657958

ABSTRACT

Histone deacetylases (HDACs) are aberrantly expressed, and inhibitors of HDACs induce apoptosis in lymphoplasmacytic cells (LPCs) in Waldenström macroglobulinemia (WM). The molecular profile by which these agents induce apoptosis in WM LPCs remains to be delineated. We examined the activity of the histone deacetylase inhibitor, vorinostat, and dissected its pro-apoptotic pathways in WM LPCs. Vorinostat induced apoptosis in WM cells through activating specific caspases at varying times. Inhibitors of apoptosis (IAPs) were down-regulated after vorinostat treatment. Cellular stress induced in vorinostat-treated WM cells was reflected by changes in the mitogen activated protein kinase (MAPK) pathways. Activated phospho-p38 MAPK was up-regulated at 12 h, while phospho-extracellular signal-regulated kinase (Erk) abruptly decreased at 24 h. Bortezomib did not augment vorinostat induced primary WM cell killing as reported in other B-cell disorders. These studies support that stress induced apoptosis in vorinostat-treated WM LPCs is mediated through disrupting the activity of the Erk and p38 MAPK pathways.


Subject(s)
Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Stress, Physiological/drug effects , Waldenstrom Macroglobulinemia/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Boronic Acids/pharmacology , Boronic Acids/toxicity , Bortezomib , Caspases/metabolism , Cell Line , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Histone Deacetylase Inhibitors/toxicity , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/toxicity , Pyrazines/pharmacology , Pyrazines/toxicity , Vorinostat
11.
Clin Lymphoma Myeloma Leuk ; 11(1): 152-6, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21454220

ABSTRACT

We studied the role of histone deacetylase inhibitors in Waldenstrom's macroglobulinemia (WM). Gene expression profiling of bone marrow CD19+ cells from 30 patients and 10 healthy donors showed overexpression of HDAC4, HDAC9, and Sirt5, with validation of HDAC9 overexpression by q-PCR in primary and BCWM.1 cells. Suberoylanilide hydroxamic acid, trichostatin A, panobinostat, and sirtinol demonstrated dose-dependent killing of BCWM.1 cells. TSA showed the greatest potency with IC50 of 70 nM. Importantly, HDAC9 activity was decreased following TSA treatment suggesting an essential role for this HDAC in WM therapy. The combination of bortezomib plus HDAC inhibitors resulted in at least additive tumor cell killing in BCWM.1 cells. TSA and bortezomib-induced apoptosis depended on a similar set of caspase activation, whereas their effect on cell cycle regulators was distinctly different. These results provided a framework for examining HDAC inhibitors as monotherapy, as well as combination therapy with bortezomib in WM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Waldenstrom Macroglobulinemia/drug therapy , Apoptosis/drug effects , Bone Marrow/enzymology , Boronic Acids/administration & dosage , Bortezomib , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival , Female , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylases/biosynthesis , Histone Deacetylases/genetics , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/therapeutic use , Immunoblotting , Male , Polymerase Chain Reaction , Pyrazines/administration & dosage , Waldenstrom Macroglobulinemia/enzymology , Waldenstrom Macroglobulinemia/genetics , Waldenstrom Macroglobulinemia/pathology
12.
Clin Lymphoma Myeloma Leuk ; 11(1): 160-3, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21454222

ABSTRACT

Waldenström's macroglobulinemia (WM) patients often present with anemia as their primary disease manifestation that may be related to hepcidin, an important regulator of iron homeostasis. We therefore determined hepcidin levels in 53 WM patients, and 20 age-matched healthy patient donors by hepcidin-25 ELISA. Serum hepcidin levels were elevated in WM patients versus healthy patients (P=.04), and correlated with BM disease involvement (P=.004), beta-2-microglobulin levels (P=.029), and inversely with hemoglobin (P=.05). No correlation with serum iron indices was observed, though in patients with high hepcidin levels, increased iron deposition in bone marrow macrophages was observed. Importantly, hepcidin transcripts and protein were produced by primary WM cells. Hepcidin levels correlated with serum IL-6 (P<.001) and C-Reactive Protein (P=.033) levels. The results of this study implicate hepcidin as a contributor to anemia in WM, and suggest that an iron re-utilization defect accompanies hepcidin overproduction leading to its sequestration in WM patients.


Subject(s)
Anemia/blood , Antimicrobial Cationic Peptides/blood , Waldenstrom Macroglobulinemia/blood , Aged , Aged, 80 and over , Female , Hepcidins , Humans , Lymphocytes/metabolism , Male , Middle Aged , Plasma Cells/metabolism
13.
Haematologica ; 95(3): 470-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19903677

ABSTRACT

BACKGROUND: Hypogammaglobulinemia is common in Waldenström's macroglobulinemia. The etiology of this finding remains unclear, but it has been speculated to be based on tumor-induced suppression of the 'uninvolved' immunoglobulin production DESIGN AND METHODS: We evaluated the incidence of IgA and IgG hypogammaglobulinemia in 207 untreated patients with Waldenström's macroglobulinemia and investigated the associated clinicopathological findings and impact of therapy. We also sequenced eight genes (AICDA, BTK, CD40, CD154, NEMO, TACI, SH2D1A, UNG) implicated in immunoglobulin deficiency in 19 Waldenström's macroglobulinemia patients with IgA and/or IgG hypogammaglobulinemia. RESULTS: At baseline 63.3%, 58.0% and 49.3% of the 207 patients had abnormally low serum levels of IgA, IgG, or both. No association between IgA and IgG hypogammaglobulinemia and disease burden, serum IgM levels, beta(2)-microglobulin, International Prognostic Scoring System score, or incidence of recurrent infections was observed, although the presence of adenopathy and/or splenomegaly was associated with a lower incidence of hypogammaglobulinemia. Lower IgA and IgG levels were associated with disease progression in patients managed with a 'watch and wait' strategy. IgA and/or IgG levels remained abnormally low despite response to treatment, including complete remissions. A missense mutation in the highly conserved catalytic site of UNG was observed in a patient with hypogammaglobulinemia, warranting further study of this pathway in Waldenström's macroglobulinemia. CONCLUSIONS: IgA and IgG hypogammaglobulinemia is common in Waldenström's macroglobulinemia and persists despite therapeutic intervention and response. IgA and IgG hypogammaglobulinemia does not predict the risk of recurrent infections in patients with Waldenström's macroglobulinemia, although lower levels of serum IgA and IgG are associated with disease progression in Waldenström's macroglobulinemia patients being managed with a 'watch and wait' strategy.


Subject(s)
Agammaglobulinemia/etiology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Waldenstrom Macroglobulinemia/complications , Adult , Agammaglobulinemia/immunology , Aged , Aged, 80 and over , Biomarkers/metabolism , Female , Humans , Infections/immunology , Male , Middle Aged , Risk Factors , Waldenstrom Macroglobulinemia/therapy
14.
Clin Lymphoma Myeloma ; 9(1): 56-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19362974

ABSTRACT

BACKGROUND: The assessment of disease burden is often difficult in patients with Waldenström's macroglobulinemia (WM) who receive rituximab due to the induction of an IgM flare, and following the removal of serum IgM by plasmapheresis. Soluble CD27 (sCD27) is a tumor necrosis factor family member secreted by WM cells which is strongly correlated with serum IgM levels and clinical responses in patients with WM. As such, we attempted to delineate its potential role in WM patients experiencing a rituximab-induced IgM flare and following plasmapheresis. PATIENTS AND METHODS: sCD27 levels were serially measured by serum-based ELISA in 8 patients who ultimately demonstrated a response to therapy, and in whom a rituximab-mediated IgM flare was observed, as well as in 3 WM patients undergoing plasmapheresis. RESULTS: Among the 8 patients who experienced a rituximab-mediated IgM flare, IgM levels rose from 3515 to a peak of 5270 mg/dL (P = .008), while sCD27 levels decreased from 174.1 to 155.9 U/mL (P = .012), with a decline observed in all patients. Among 3 patients undergoing plasmapheresis, IgM levels declined from a median of 6940 to 4770 mg/dL (P = .031), while median sCD27 levels remained without significant change (P = .317). CONCLUSION: sCD27 is a faithful marker of disease burden and is unaffected by the rituximab-induced IgM flare, as well as plasmapheresis in WM. The use of this marker may aid in correctly predicting clinical outcome in patients undergoing treatment with rituximab and/or plasmapheresis in WM.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Tumor Necrosis Factor Receptor Superfamily, Member 7/blood , Waldenstrom Macroglobulinemia/blood , Waldenstrom Macroglobulinemia/drug therapy , Aged , Antibodies, Monoclonal, Murine-Derived , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin M/blood , Middle Aged , Plasmapheresis , Rituximab
15.
J Virol ; 82(14): 7047-58, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18448528

ABSTRACT

Flavivirus methyltransferase catalyzes both guanine N7 and ribose 2'-OH methylations of the viral RNA cap (GpppA-RNA-->m(7)GpppAm-RNA). The methyltransferase is physically linked to an RNA-dependent RNA polymerase (RdRp) in the flaviviral NS5 protein. Here, we report genetic interactions of West Nile virus (WNV) methyltransferase with the RdRp and the 5'-terminal stem-loop of viral genomic RNA. Genome-length RNAs, containing amino acid substitutions of D146 (a residue essential for both cap methylations) in the methyltransferase, were transfected into BHK-21 cells. Among the four mutant RNAs (D146L, D146P, D146R, and D146S), only D146S RNA generated viruses in transfected cells. Sequencing of the recovered viruses revealed that, besides the D146S change in the methyltransferase, two classes of compensatory mutations had reproducibly emerged. Class 1 mutations were located in the 5'-terminal stem-loop of the genomic RNA (a G35U substitution or U38 insertion). Class 2 mutations resided in NS5 (K61Q in methyltransferase and W751R in RdRp). Mutagenesis analysis, using a genome-length RNA and a replicon of WNV, demonstrated that the D146S substitution alone was lethal for viral replication; however, the compensatory mutations rescued replication, with the highest rescuing efficiency occurring when both classes of mutations were present. Biochemical analysis showed that a low level of N7 methylation of the D146S methyltransferase is essential for the recovery of adaptive viruses. The methyltransferase K61Q mutation facilitates viral replication through improved N7 methylation activity. The RdRp W751R mutation improves viral replication through an enhanced polymerase activity. Our results have clearly established genetic interactions among flaviviral methyltransferase, RdRp, and the 5' stem-loop of the genomic RNA.


Subject(s)
Methyltransferases/metabolism , Protein Interaction Mapping , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , West Nile virus/metabolism , Amino Acid Substitution/genetics , Animals , Base Sequence , Cell Line , Cricetinae , DNA Mutational Analysis , Methyltransferases/genetics , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Nucleic Acid Conformation , Protein Binding , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Suppression, Genetic , Viral Plaque Assay , Virus Replication/genetics , West Nile virus/enzymology , West Nile virus/genetics
16.
J Virol ; 82(9): 4295-307, 2008 May.
Article in English | MEDLINE | ID: mdl-18305027

ABSTRACT

Flaviviruses encode a single methyltransferase domain that sequentially catalyzes two methylations of the viral RNA cap, GpppA-RNA-->m(7)GpppA-RNA-->m(7)GpppAm-RNA, by using S-adenosyl-l-methionine (SAM) as a methyl donor. Crystal structures of flavivirus methyltransferases exhibit distinct binding sites for SAM, GTP, and RNA molecules. Biochemical analysis of West Nile virus methyltransferase shows that the single SAM-binding site donates methyl groups to both N7 and 2'-O positions of the viral RNA cap, the GTP-binding pocket functions only during the 2'-O methylation, and two distinct sets of amino acids in the RNA-binding site are required for the N7 and 2'-O methylations. These results demonstrate that flavivirus methyltransferase catalyzes two cap methylations through a substrate-repositioning mechanism. In this mechanism, guanine N7 of substrate GpppA-RNA is first positioned to SAM to generate m(7)GpppA-RNA, after which the m(7)G moiety is repositioned to the GTP-binding pocket to register the 2'-OH of the adenosine with SAM, generating m(7)GpppAm-RNA. Because N7 cap methylation is essential for viral replication, inhibitors designed to block the pocket identified for the N7 cap methylation could be developed for flavivirus therapy.


Subject(s)
Methyltransferases/chemistry , Methyltransferases/metabolism , RNA Caps/metabolism , West Nile virus/enzymology , Binding Sites , Crystallography, X-Ray , Guanosine Triphosphate/metabolism , Methylation , S-Adenosylmethionine/metabolism
17.
J Virol ; 81(8): 3891-903, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17267492

ABSTRACT

The plus-strand RNA genome of flavivirus contains a 5' terminal cap 1 structure (m7GpppAmG). The flaviviruses encode one methyltransferase, located at the N-terminal portion of the NS5 protein, to catalyze both guanine N-7 and ribose 2'-OH methylations during viral cap formation. Representative flavivirus methyltransferases from dengue, yellow fever, and West Nile virus (WNV) sequentially generate GpppA-->m7GpppA-->m7GpppAm. The 2'-O methylation can be uncoupled from the N-7 methylation, since m7GpppA-RNA can be readily methylated to m7GpppAm-RNA. Despite exhibiting two distinct methylation activities, the crystal structure of WNV methyltransferase at 2.8 A resolution showed a single binding site for S-adenosyl-L-methionine (SAM), the methyl donor. Therefore, substrate GpppA-RNA should be repositioned to accept the N-7 and 2'-O methyl groups from SAM during the sequential reactions. Electrostatic analysis of the WNV methyltransferase structure showed that, adjacent to the SAM-binding pocket, is a highly positively charged surface that could serve as an RNA binding site during cap methylations. Biochemical and mutagenesis analyses show that the N-7 and 2'-O cap methylations require distinct buffer conditions and different side chains within the K61-D146-K182-E218 motif, suggesting that the two reactions use different mechanisms. In the context of complete virus, defects in both methylations are lethal to WNV; however, viruses defective solely in 2'-O methylation are attenuated and can protect mice from later wild-type WNV challenge. The results demonstrate that the N-7 methylation activity is essential for the WNV life cycle and, thus, methyltransferase represents a novel target for flavivirus therapy.


Subject(s)
Methyltransferases/chemistry , Methyltransferases/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , West Nile virus/chemistry , West Nile virus/physiology , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Dengue Virus/chemistry , Flavivirus , Mice , Mice, Inbred C3H , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , RNA Caps/metabolism , RNA, Viral/metabolism , S-Adenosylmethionine/metabolism , Virulence Factors/chemistry , Virulence Factors/metabolism , West Nile Fever/virology , West Nile virus/pathogenicity , Yellow fever virus/chemistry
18.
J Gen Virol ; 88(Pt 3): 865-874, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17325359

ABSTRACT

West Nile virus (WNV) has successfully spread throughout the USA, Canada, Mexico, the Caribbean and parts of Central and South America since its 1999 introduction into North America. Despite infecting a broad range of both mosquito and avian species, the virus remains highly genetically conserved. This lack of evolutionary change over space and time is common with many arboviruses and is frequently attributed to the adaptive constraints resulting from the virus cycling between vertebrate hosts and invertebrate vectors. WNV, like most RNA viruses studied thus far, has been shown in nature to exist as a highly genetically diverse population of genotypes. Few studies have directly evaluated the role of these mutant spectra in viral fitness and adaptation. Using clonal analysis and reverse genetics experiments, this study evaluated genotype diversity and the importance of consensus change in producing the adaptive phenotype of WNV following sequential mosquito cell passage. The results indicated that increases in the replicative ability of WNV in mosquito cells correlate with increases in the size of the mutant spectrum, and that consensus change is not solely responsible for alterations in viral fitness and adaptation of WNV. These data provide evidence of the importance of quasispecies dynamics in the adaptation of a flavivirus to new and changing environments and hosts, with little evidence of significant genetic change.


Subject(s)
Adaptation, Biological/genetics , Genetic Variation , West Nile virus/physiology , Animals , Chlorocebus aethiops , RNA, Viral/genetics , Sequence Analysis, DNA , Serial Passage , Vero Cells , Viral Plaque Assay , Virus Replication/genetics , West Nile virus/genetics
19.
Virology ; 361(1): 229-41, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17178141

ABSTRACT

An adaptive mutation (E249G) within West Nile virus (WNV) NS4B gene was consistently recovered from replicon RNAs in C3H/He mouse cells. The E249G is located at the C-terminal tail of NS4B predicted to be on the cytoplasmic side of the endoplasmic reticulum membrane. The E249G substitution reduced replicon RNA synthesis. Compared with the wild-type NS4B, the E249G mutant protein exhibited a similar efficiency in evasion of interferon-beta response. Recombinant E249G virus exhibited smaller plaques, slower growth kinetics, and lower RNA synthesis than the wild-type virus in a host-dependent manner, with the greatest difference in rodent cells (C3H/He and BHK-21) and the least difference in mosquito cells (C3/36). Selection of revertants of E249G virus identified a second site mutation at residue 246, which could compensate for the low replication phenotype in cell culture. These results demonstrate that distinct residues within the C-terminal tail of flavivirus NS4B are critical for viral replication.


Subject(s)
RNA, Viral/biosynthesis , Viral Nonstructural Proteins/genetics , West Nile Fever/virology , West Nile virus/physiology , Adaptation, Physiological , Animals , Cell Line , Female , Interferon-beta/pharmacology , Mice , Mice, Inbred C3H , Mutation , Virus Replication/drug effects , West Nile Fever/prevention & control
20.
J Virol ; 80(17): 8362-70, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16912287

ABSTRACT

Many flaviviruses are globally important human pathogens. Their plus-strand RNA genome contains a 5'-cap structure that is methylated at the guanine N-7 and the ribose 2'-OH positions of the first transcribed nucleotide, adenine (m(7)GpppAm). Using West Nile virus (WNV), we demonstrate, for the first time, that the nonstructural protein 5 (NS5) mediates both guanine N-7 and ribose 2'-O methylations and therefore is essential for flavivirus 5'-cap formation. We show that a recombinant full-length and a truncated NS5 protein containing the methyltransferase (MTase) domain methylates GpppA-capped and m(7)GpppA-capped RNAs to m(7)GpppAm-RNA, using S-adenosylmethionine as a methyl donor. Furthermore, methylation of GpppA-capped RNA sequentially yielded m(7)GpppA- and m(7)GpppAm-RNA products, indicating that guanine N-7 precedes ribose 2'-O methylation. Mutagenesis of a K(61)-D(146)-K(182)-E(218) tetrad conserved in other cellular and viral MTases suggests that NS5 requires distinct amino acids for its N-7 and 2'-O MTase activities. The entire K(61)-D(146)-K(182)-E(218) motif is essential for 2'-O MTase activity, whereas N-7 MTase activity requires only D(146). The other three amino acids facilitate, but are not essential for, guanine N-7 methylation. Amino acid substitutions within the K(61)-D(146)-K(182)-E(218) motif in a WNV luciferase-reporting replicon significantly reduced or abolished viral replication in cells. Additionally, the mutant MTase-mediated replication defect could not be trans complemented by a wild-type replicase complex. These findings demonstrate a critical role for the flavivirus MTase in viral reproduction and underscore this domain as a potential target for antiviral therapy.


Subject(s)
5' Untranslated Regions/chemistry , Methyltransferases/metabolism , Ribose/analogs & derivatives , Ribose/metabolism , Viral Nonstructural Proteins/metabolism , West Nile virus/metabolism , 5' Untranslated Regions/metabolism , Animals , Cell Line , Cricetinae , Genome, Viral , Methylation , RNA Caps , Virus Replication , West Nile virus/chemistry , West Nile virus/genetics , West Nile virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...