Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
4.
Blood Cancer Discov ; 2(3): 216-225, 2021 05.
Article in English | MEDLINE | ID: mdl-34164626

ABSTRACT

Clonal hematopoiesis predisposes to hematological malignancies. However, clonal hematopoiesis is understudied in classical Hodgkin lymphoma (cHL), a mature B-cell neoplasm exhibiting the most abundant microenvironment. We analyzed clonal hematopoiesis in 40 cHL cases by sequencing microdissected tumor cells and matched normal cells from blood and/or lymph nodes. Five patients had blood and/or tissue clonal hematopoiesis. In three of five patients (all failing first-line therapy), clonal hematopoiesis spread through the tissue microenvironment extensively, and featured mutant DNMT3AR882H , KRASG60D and DNMT3AR882H +TET2Q1274 * in 33%, 92% and 60% of non-neoplastic cells, respectively. In the latter case, DNMT3A/TET2-mutant clonal hematopoiesis seeded the neoplastic clone, which was infected by the Epstein-Barr virus and showed almost no other somatic mutations exome-wide. In the former case, DNMT3A-mutant clonal hematopoiesis did not originate the neoplastic clone despite dominating the blood and B-cell lineage (~94% leukocytes; ~96% mature blood B cells), yet led to NPM1-mutated acute myeloid leukemia 6 years after therapy for cHL. Our results expand to cHL the spectrum of hematologic malignancies associated with clonal hematopoiesis.


Subject(s)
Epstein-Barr Virus Infections , Hodgkin Disease , Clonal Hematopoiesis/genetics , Herpesvirus 4, Human , Hodgkin Disease/genetics , Humans , Mutation , Tumor Microenvironment
5.
Br J Haematol ; 194(1): 78-82, 2021 07.
Article in English | MEDLINE | ID: mdl-33932027

ABSTRACT

We investigated the current role of interferon-alpha (IFNα) in hairy cell leukaemia (HCL) in a retrospective analysis of patients with HCL. A cohort of 74 patients with HCL was divided in to three groups: (A) patients aged >65 years with first-line treatment; (B) patients with comorbidities with first-line treatment; (C) patients who were purine analogues resistant. In total, 94% achieved a response, with a complete response rate of 24%. After a median (range) follow-up of 60 (7-236) months, 55 patients (78%) are still responding. The 5-year progression-free survival was 95%, 68%, and 96% in groups A, B and C respectively. A proportion of patients were monitored through their B-Raf proto-oncogene, serine/threonine kinase (BRAF)-V600E status. IFNα remains a possible option in select patients with HCL, where minimal residual disease negativity is achievable.


Subject(s)
Antineoplastic Agents/therapeutic use , Interferon-alpha/therapeutic use , Leukemia, Hairy Cell/drug therapy , Adult , Aged , Aged, 80 and over , Alopecia/chemically induced , Antineoplastic Agents/adverse effects , Biomarkers, Tumor/blood , Comorbidity , Disease-Free Survival , Drug Resistance, Neoplasm , Drug Substitution , Female , Follow-Up Studies , Humans , Interferon-alpha/adverse effects , Kaplan-Meier Estimate , Leukemia, Hairy Cell/blood , Leukemia, Hairy Cell/mortality , Male , Middle Aged , Mutation, Missense , Point Mutation , Progression-Free Survival , Proto-Oncogene Proteins B-raf/blood , Proto-Oncogene Proteins B-raf/genetics , Retrospective Studies , Salvage Therapy
7.
Blood ; 131(22): 2454-2465, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29650799

ABSTRACT

Dissecting the pathogenesis of classical Hodgkin lymphoma (cHL), a common cancer in young adults, remains challenging because of the rarity of tumor cells in involved tissues (usually <5%). Here, we analyzed the coding genome of cHL by microdissecting tumor and normal cells from 34 patient biopsies for a total of ∼50 000 singly isolated lymphoma cells. We uncovered several recurrently mutated genes, namely, STAT6 (32% of cases), GNA13 (24%), XPO1 (18%), and ITPKB (16%), and document the functional role of mutant STAT6 in sustaining tumor cell viability. Mutations of STAT6 genetically and functionally cooperated with disruption of SOCS1, a JAK-STAT pathway inhibitor, to promote cHL growth. Overall, 87% of cases showed dysregulation of the JAK-STAT pathway by genetic alterations in multiple genes (also including STAT3, STAT5B, JAK1, JAK2, and PTPN1), attesting to the pivotal role of this pathway in cHL pathogenesis and highlighting its potential as a new therapeutic target in this disease.


Subject(s)
Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , Janus Kinases/genetics , Mutation , STAT Transcription Factors/genetics , Cell Line, Tumor , DNA Mutational Analysis , Hodgkin Disease/metabolism , Hodgkin Disease/pathology , Humans , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction
8.
Oncotarget ; 8(37): 60826-60840, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-28977828

ABSTRACT

Fever plays a role in activating innate immunity while its relevance in activating adaptive immunity is less clear. Even brief exposure to elevated temperatures significantly impacts on the immunostimulatory capacity of dendritic cells (DCs), but the consequences on immune response remain unclear. To address this issue, we analyzed the gene expression profiles of normal human monocyte-derived DCs from nine healthy adults subjected either to fever-like thermal conditions (39°C) or to normal temperature (37°C) for 180 minutes. Exposure of DCs to 39°C caused upregulation of 43 genes and downregulation of 24 genes. Functionally, the up/downregulated genes are involved in post-translational modification, protein folding, cell death and survival, and cellular movement. Notably, when compared to monocytes, DCs differentially upregulated transcription of the secreted protein IGFBP-6, not previously known to be specifically linked to hyperthermia. Exposure of DCs to 39°C induced apoptosis/necrosis and resulted in accumulation of IGFBP-6 in the conditioned medium at 48 h. IGFBP-6 may have a functional role in the hyperthermic response as it induced chemotaxis of monocytes and T lymphocytes, but not of B lymphocytes. Thus, temperature regulates complex biological DC functions that most likely contribute to their ability to induce an efficient adaptive immune response.

9.
Blood ; 125(8): 1207-16, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25480661

ABSTRACT

Hairy cell leukemia (HCL) shows unique clinicopathological and biological features. HCL responds well to purine analogs but relapses are frequent and novel therapies are required. BRAF-V600E is the key driver mutation in HCL and distinguishes it from other B-cell lymphomas, including HCL-like leukemias/lymphomas (HCL-variant and splenic marginal zone lymphoma). The kinase-activating BRAF-V600E mutation also represents an ideal therapeutic target in HCL. Here, we investigated the biological and therapeutic importance of the activated BRAF-mitogen-activated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway in HCL by exposing in vitro primary leukemic cells purified from 26 patients to clinically available BRAF (vemurafenib; dabrafenib) or MEK (trametinib) inhibitors. Results were validated in vivo in samples from vemurafenib-treated HCL patients within a phase 2 clinical trial. BRAF and MEK inhibitors caused, specifically in HCL (but not HCL-like) cells, marked MEK/ERK dephosphorylation, silencing of the BRAF-MEK-ERK pathway transcriptional output, loss of the HCL-specific gene expression signature, downregulation of the HCL markers CD25, tartrate-resistant acid phosphatase, and cyclin D1, smoothening of leukemic cells' hairy surface, and, eventually, apoptosis. Apoptosis was partially blunted by coculture with bone marrow stromal cells antagonizing MEK-ERK dephosphorylation. This protective effect could be counteracted by combined BRAF and MEK inhibition. Our results strongly support and inform the clinical use of BRAF and MEK inhibitors in HCL.


Subject(s)
Antineoplastic Agents , Imidazoles , Indoles , Leukemia, Hairy Cell/drug therapy , Leukemia, Hairy Cell/genetics , Oximes , Pyridones , Pyrimidinones , Sulfonamides , Transcriptome/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Gene Expression Regulation, Leukemic/drug effects , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Oximes/pharmacology , Oximes/therapeutic use , Phosphorylation/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Tumor Cells, Cultured , Vemurafenib
11.
Histopathology ; 61(5): 930-41, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22716304

ABSTRACT

AIMS: The aim of this study was to search for a molecule selectively expressed by marginal zone (MZ) lymphomas (MZLs), whose diagnosis is currently based on morphological criteria and negativity for markers detectable in other B-cell lymphomas. METHODS AND RESULTS: Two thousand one hundred and four peripheral lymphomas of various types were immunostained with a monoclonal antibody against immunoglobulin superfamily receptor translocation-associated 1 (IRTA1), which recognizes the equivalents of MZ in human lymphoid tissues other than spleen. IRTA1 expression was restricted to extranodal (93%) and nodal MZLs (73%) and to lymphomas with MZ differentiation. Extranodal MZL cells with the strongest IRTA1 expression were usually located adjacent to epithelia, mimicking the IRTA1 expression pattern of normal and acquired mucosa-associated lymphoid tissue (MALT). The cytological features, growth pattern and IRTA1 positivity in nodal MZLs suggest they may derive from IRTA1(+) perifollicular B cells or monocytoid B cells detectable in reactive lymph nodes. Double immunostaining for IRTA1/bcl-6 tracked the colonization of B-cell follicles by MZL cells, and showed modulation of their phenotype (e.g. acquisition of bcl-6) during recirculation through germinal centres. MZL cells differentiating into plasma cells usually lost IRTA1. CONCLUSIONS: These results further expand our knowledge of the biology of MZLs, and highlight IRTA1 as the first positive marker for MZLs, enabling more accurate diagnosis of these neoplasms.


Subject(s)
Lymphoma, B-Cell, Marginal Zone/immunology , Lymphoma, B-Cell, Marginal Zone/pathology , Receptors, Fc/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell, Marginal Zone/genetics , Spleen/immunology , Spleen/pathology , Translocation, Genetic
12.
N Engl J Med ; 364(24): 2305-15, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21663470

ABSTRACT

BACKGROUND: Hairy-cell leukemia (HCL) is a well-defined clinicopathological entity whose underlying genetic lesion is still obscure. METHODS: We searched for HCL-associated mutations by performing massively parallel sequencing of the whole exome of leukemic and matched normal cells purified from the peripheral blood of an index patient with HCL. Findings were validated by Sanger sequencing in 47 additional patients with HCL. RESULTS: Whole-exome sequencing identified five missense somatic clonal mutations that were confirmed on Sanger sequencing, including a heterozygous mutation in BRAF that results in the BRAF V600E variant protein. Since BRAF V600E is oncogenic in other tumors, further analyses were focused on this genetic lesion. The same BRAF mutation was noted in all the other 47 patients with HCL who were evaluated by means of Sanger sequencing. None of the 195 patients with other peripheral B-cell lymphomas or leukemias who were evaluated carried the BRAF V600E variant, including 38 patients with splenic marginal-zone lymphomas or unclassifiable splenic lymphomas or leukemias. In immunohistologic and Western blot studies, HCL cells expressed phosphorylated MEK and ERK (the downstream targets of the BRAF kinase), indicating a constitutive activation of the RAF-MEK-ERK mitogen-activated protein kinase pathway in HCL. In vitro incubation of BRAF-mutated primary leukemic hairy cells from 5 patients with PLX-4720, a specific inhibitor of active BRAF, led to a marked decrease in phosphorylated ERK and MEK. CONCLUSIONS; The BRAF V600E mutation was present in all patients with HCL who were evaluated. This finding may have implications for the pathogenesis, diagnosis, and targeted therapy of HCL. (Funded by Associazione Italiana per la Ricerca sul Cancro and others.).


Subject(s)
Leukemia, Hairy Cell/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adult , Aged , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Leukemia, Hairy Cell/metabolism , Leukemia, Hairy Cell/pathology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , MAP Kinase Kinase Kinases/metabolism , Male , Middle Aged , Sequence Analysis, DNA
13.
Haematologica ; 93(5): 775-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18367491

ABSTRACT

We investigated the NPM1 mutation status or subcellular expression of NPM protein (nuclear vs. aberrant cytoplasmic) at diagnosis and relapse in 125 patients with acute myeloid leukemia from Italy and Germany. All 52 patients with acute myeloidleukemia carrying at diagnosis mutated or cytoplasmic NPM (NPMc(+) acute myeloid leukemia) retained this feature at relapse. Notably, cytoplasmic mutated NPM has now been retained for eight years in a xenotransplant model of NPMc(+) acute myeloid leukemia in immunodeficient mice. None of 73 acute myeloid leukemia patients carrying at diagnosis wild-type NPM1 gene or showing at immunohistochemistry nucleus-restricted expression of nucleophosmin (NPMc(-) acute myeloid leukemia), which is predictive of NPM1 gene in germline configuration, acquired cytoplasmic mutated NPM at relapse. This finding further confirms that NPMc(+) acute myeloid leukemia represents a primary event rather than a transformation stage of NPMc(-) acute myeloid leukemia. The stability of cytoplasmic mutated NPM in patients with acute myeloid leukemia, even at relapse in extramedullary sites, and in a xenotransplant model, suggest this event is crucial for leukemogenesis and represents the rationale for monitoring minimal residual disease and molecular targeted therapy in NPMc(+) acute myeloid leukemia.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation , Nuclear Proteins/genetics , Adult , Animals , Chromosome Aberrations , Cytoplasm/metabolism , Female , Humans , Immunohistochemistry/methods , Karyotyping , Mice , Mice, SCID , Neoplasm Transplantation , Nuclear Proteins/metabolism , Nucleophosmin , Recurrence
14.
Cancer Res ; 67(13): 6230-7, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616680

ABSTRACT

Creation of a nuclear export signal (NES) motif and loss of tryptophans (W) 288 and 290 (or 290 only) at the COOH terminus of nucleophosmin (NPM) are both crucial for NPM aberrant cytoplasmic accumulation in acute myelogenous leukemia (AML) carrying NPM1 mutations. Hereby, we clarify how these COOH-terminal alterations functionally cooperate to delocalize NPM to the cytoplasm. Using a Rev(1.4)-based shuttling assay, we measured the nuclear export efficiency of six different COOH-terminal NES motifs identified in NPM mutants and found significant strength variability, the strongest NES motifs being associated with NPM mutants retaining W288. When artificially coupled with a weak NES, W288-retaining NPM mutants are not exported efficiently into cytoplasm because the force (W288) driving the mutants toward the nucleolus overwhelms the force (NES) exporting the mutants into cytoplasm. We then used this functional assay to study the physiologic NH(2)-terminal NES motifs of wild-type NPM and found that they are weak, which explains the prominent nucleolar localization of wild-type NPM. Thus, the opposing balance of forces (tryptophans and NES) seems to determine the subcellular localization of NPM. The fact that W288-retaining mutants always combine with the strongest NES reveals mutational selective pressure toward efficient export into cytoplasm, pointing to this event as critical for leukemogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Amino Acid Motifs , Animals , Cell Transformation, Neoplastic , Cytoplasm/metabolism , DNA Mutational Analysis , Fibroblasts/metabolism , Leukemia, Myeloid, Acute/metabolism , Mice , Microscopy, Fluorescence , Mutation , NIH 3T3 Cells , Nucleophosmin , Plasmids/metabolism , Protein Structure, Tertiary
15.
Blood ; 108(13): 4146-55, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16926285

ABSTRACT

Because of a lack of specific clonality markers, information on lineage involvement and cell of origin of acute myeloid leukemia with normal karyotype (AML-NK), is missing. Because Nucleophosmin (NPM) gene is frequently mutated in AML-NK and causes aberrant NPM cytoplasmic localization (NPMc+), it was used as an AML lineage clonality marker. Clonal NPM exon 12 mutations were detected in myeloid, monocytic, erythroid, and megakaryocytic cells but not in fibroblasts or endothelia that were laser-microdissected from 3 patients with NPMc+ AML. Aberrant cytoplasmic expression of mutated NPM proteins was identified with anti-NPM antibodies in 2 or more myeloid hemopoietic cell lineages in 99 (61.5%) of 161 of NPMc+ AML paraffin-embedded bone marrow biopsies; lymphoid involvement was excluded in 3 investigated cases. These findings suggest that NPMc+ AML derives from either a common myeloid or earlier progenitor. Immunohistochemical studies show that varying combinations and ratios of NPMc+ leukemic cells from distinct lineages are responsible for heterogeneity within each French-American-British (FAB) classification type and for NPMc+ AML falling into different FAB categories. These findings question the value of FAB criteria in subdividing the WHO category of "AML not otherwise characterized" and suggest that, for clinical use, NPMc+ AML be provisionally regarded as a separate AML with prognostic significance.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Mutation , Nuclear Proteins/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , Humans , Karyotyping/methods , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Nucleophosmin , Protein Transport/genetics , World Health Organization
16.
Blood ; 107(11): 4514-23, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16455950

ABSTRACT

We recently identified aberrant cytoplasmic expression of nucleophosmin (NPM) as the immunohistochemical marker of a large subgroup of acute myeloid leukemia (AML) (about one-third of adult AML) that is characterized by normal karyotype and mutations occurring at the exon-12 of the NPM gene. In this paper, we have elucidated the molecular mechanism underlying the abnormal cytoplasmic localization of NPM. All 29 AML-associated mutated NPM alleles so far identified encode abnormal proteins which have acquired at the C-terminus a nuclear export signal (NES) motif and lost both tryptophan residues 288 and 290 (or only the residue 290) which determine nucleolar localization. We show for the first time that both alterations are crucial for NPM mutant export from nucleus to cytoplasm. In fact, the cytoplasmic accumulation of NPM is blocked by leptomycin-B and ratjadones, specific exportin-1/Crm1-inhibitors, and by reinsertion of tryptophan residues 288 and 290, which respectively relocate NPM mutants in the nucleoplasm and nucleoli. NPM leukemic mutants in turn recruit the wild-type NPM from nucleoli to nucleoplasm and cytoplasm. These findings indicate that potential therapeutic strategies aimed to retarget NPM to its physiological sites will have to overcome 2 obstacles, the new NES motif and the mutated tryptophan(s) at the NPM mutant C-terminus.


Subject(s)
Active Transport, Cell Nucleus/genetics , Leukemia/genetics , Mutation , Nuclear Export Signals/genetics , Nuclear Proteins/genetics , Tryptophan/genetics , Amino Acid Motifs , Animals , Base Sequence , Cell Line , Cell Nucleolus/metabolism , Cytoplasm/metabolism , Humans , Leukemia/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Transfection
17.
N Engl J Med ; 352(3): 254-66, 2005 Jan 20.
Article in English | MEDLINE | ID: mdl-15659725

ABSTRACT

BACKGROUND: Nucleophosmin (NPM), a nucleocytoplasmic shuttling protein with prominent nucleolar localization, regulates the ARF-p53 tumor-suppressor pathway. Translocations involving the NPM gene cause cytoplasmic dislocation of the NPM protein. METHODS: We used immunohistochemical methods to study the subcellular localization of NPM in bone marrow-biopsy specimens from 591 patients with primary acute myelogenous leukemia (AML). We then correlated the presence of cytoplasmic NPM with clinical and biologic features of the disease. RESULTS: Cytoplasmic NPM was detected in 208 (35.2 percent) of the 591 specimens from patients with primary AML but not in 135 secondary AML specimens or in 980 hematopoietic or extrahematopoietic neoplasms other than AML. It was associated with a wide spectrum of morphologic subtypes of the disease, a normal karyotype, and responsiveness to induction chemotherapy, but not with recurrent genetic abnormalities. There was a high frequency of FLT3 internal tandem duplications and absence of CD34 and CD133 in AML specimens with a normal karyotype and cytoplasmic dislocation of NPM, but not in those in which the protein was restricted to the nucleus. AML specimens with cytoplasmic NPM carried mutations of the NPM gene that were predicted to alter the protein at its C-terminal; this mutant gene caused cytoplasmic localization of NPM in transfected cells. CONCLUSIONS: Cytoplasmic NPM is a characteristic feature of a large subgroup of patients with AML who have a normal karyotype, NPM gene mutations, and responsiveness to induction chemotherapy.


Subject(s)
Bone Marrow/pathology , Cytoplasm/chemistry , Leukemia, Myeloid, Acute/genetics , Mutation , Nuclear Proteins/genetics , Adolescent , Adult , Antibodies, Monoclonal , Antineoplastic Agents/therapeutic use , Base Sequence , Cell Nucleolus , DNA Mutational Analysis , Humans , Karyotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Middle Aged , Nuclear Proteins/analysis , Nuclear Proteins/immunology , Nucleophosmin , Remission Induction , Transfection , Translocation, Genetic
18.
Blood ; 105(7): 2812-20, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15576473

ABSTRACT

We generated a murine monoclonal antibody (B28p) detecting an antigenic determinant shared by the immunoglobulin superfamily receptor translocation-associated 1 (IRTA1) receptor (the immunogen used to raise B28p) and an unrelated 28-kDa protein that was subsequently subjected to extensive characterization. The expression of the 28-kDa protein in normal lymphohematopoietic tissues was restricted to B cells and plasma cells and clearly differed from that expected for IRTA1 (selectively expressed by mucosa-associated lymphoid tissue [MALT] marginal zone B cells). Two-dimensional polyacrylamide gel electrophoresis (2D-PAGE)/mass-spectrometry analysis identified the 28-kDa protein as human tumor protein D52 (TPD52), whose expression had been previously described only in normal and neoplastic epithelia. Specific B28p reactivity with TPD52 was confirmed by immunostaining/immunoblotting of TPD52-transfected cells. TPD52 expression pattern in normal and neoplastic B cells was unique. In fact, unlike other B-cell molecules (paired box 5 [PAX5], CD19, CD79a, CD20, CD22), which are down-regulated during differentiation from B cells to plasma cells, TPD52 expression reached its maximum levels at the plasma cell stage. In the Thiel myeloma cell line, TPD52 bound to annexin VI in a Ca(2+)-dependent manner, suggesting that these molecules may act in concert to regulate secretory processes in plasma cells, similarly to what was observed in pancreatic acinar cells. Finally, the anti-TPD52 monoclonal antibody served as a valuable tool for the diagnosis of B-cell malignancies.


Subject(s)
Annexin A6/metabolism , Calcium/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Plasma Cells/physiology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , B-Lymphocytes/physiology , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Epitopes, B-Lymphocyte/immunology , Gene Expression Regulation, Leukemic/immunology , Leukemia, B-Cell/diagnosis , Leukemia, B-Cell/physiopathology , Mass Spectrometry , Mice , Mice, Inbred BALB C , Molecular Weight , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Receptors, Cell Surface/immunology , Receptors, Fc
19.
Blood ; 102(10): 3684-92, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12881317

ABSTRACT

IRTA1 (immunoglobulin superfamily receptor translocation-associated 1) is a novel surface B-cell receptor related to Fc receptors, inhibitory receptor superfamily (IRS), and cell adhesion molecule (CAM) family members and we mapped for the first time its distribution in human lymphoid tissues, using newly generated specific antibodies. IRTA1 was selectively and consistently expressed by a B-cell population located underneath and within the tonsil epithelium and dome epithelium of Peyer patches (regarded as the anatomic equivalents of marginal zone). Similarly, in mucosa-associated lymphoid tissue (MALT) lymphomas IRTA1 was mainly expressed by tumor cells involved in lympho-epithelial lesions. In contrast, no or a low number of IRTA1+ cells was usually observed in the marginal zone of mesenteric lymph nodes and spleen. Interestingly, monocytoid B cells in reactive lymph nodes were strongly IRTA1+. Tonsil IRTA1+ cells expressed the memory B-cell marker CD27 but not mantle cell-, germinal center-, and plasma cell-associated molecules. Polymerase chain reaction (PCR) analysis of single tonsil IRTA1+ cells showed they represent a mixed B-cell population carrying mostly mutated, but also unmutated, IgV genes. The immunohistochemical finding in the tonsil epithelial areas of aggregates of IRTA1+ B cells closely adjacent to plasma cells surrounding small vessels suggests antigen-triggered in situ proliferation/differentiation of memory IRTA1+ cells into plasma cells. Collectively, these results suggest a role of IRTA1 in the immune function of B cells within epithelia.


Subject(s)
B-Lymphocytes/chemistry , Epithelial Cells/chemistry , Lymphoma, B-Cell, Marginal Zone/pathology , Receptors, Immunologic/metabolism , Antibodies, Monoclonal/biosynthesis , Genes, Immunoglobulin/genetics , Humans , Immunophenotyping , Lymph Nodes/cytology , Palatine Tonsil/cytology , Peyer's Patches/cytology , Plasma Cells/cytology , Receptors, Immunologic/analysis , Receptors, Immunologic/immunology , Spleen/cytology , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...