Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Arthritis Rheumatol ; 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38272827

ABSTRACT

OBJECTIVE: In this study, we examine how advancements in novel antirheumatic drugs affect the clinicopathologic features of lymphoproliferative disorder (LPD) in patients with rheumatoid arthritis (RA). METHODS: In this multicenter study across 53 hospitals in Japan, we characterized patients with RA who developed LPDs and visited the hospitals between January 1999 and March 2021. The statistical tools used included Fisher's exact test, the Mann-Whitney U-test, the log-rank test, logistic regression analysis, and Cox proportional hazards models. RESULTS: Overall, 752 patients with RA-associated LPD (RA-LPD) and 770 with sporadic LPD were included in the study. We observed significant differences in the clinicopathologic features between patients with RA-LPD and those with sporadic LPD. Histopathological analysis revealed a high frequency of LPD-associated immunosuppressive conditions. Furthermore, patients with RA-LPD were evaluated based on the antirheumatic drugs administered. The methotrexate (MTX) plus tacrolimus and MTX plus tumor necrosis factor inhibitor (TNFi) groups had different affected site frequencies and histologic subtypes than the MTX-only group. Moreover, MTX and TNFi may synergistically affect susceptibility to Epstein-Barr virus infection. In case of antirheumatic drugs administered after LPD onset, tocilizumab (TCZ)-only therapy was associated with lower frequency of regrowth after spontaneous regression than other regimens. CONCLUSION: Antirheumatic drugs administered before LPD onset may influence the clinicopathologic features of RA-LPD, with patterns changing over time. Furthermore, TCZ-only regimens are recommended after LPD onset.

2.
Acta Haematol ; 143(1): 33-39, 2020.
Article in English | MEDLINE | ID: mdl-31216534

ABSTRACT

Immune-mediated processes are considered important in the pathogenesis of bone marrow failure syndromes (BFS). We previously reported that natural killer group 2D (NKG2D) ligands were expressed on pathological blood cells of patients with BFS and that NKG2D immunity may be involved in bone marrow failure. In addition to membranous NKG2D ligands on the cell surface, soluble NKG2D ligands can exist in plasma. We therefore examined the relationship between soluble NKG2D ligands and blood cell counts in 86 patients with BFS, including aplastic anemia, myelodysplastic syndrome with single lineage dysplasia, and paroxysmal nocturnal hemoglobinuria. Approximately half of the BFS patients were positive for soluble NKG2D ligands in the plasma by enzyme-linked immunosorbent assay, and soluble NKG2D ligand-positive BFS patients exhibited severe cytopenia regardless of membranous NKG2D ligand expression. In vitroanalyses demonstrated that soluble ULBP1, an NKG2D ligand, down-regulated NKG2D receptors on CD2-positive cells in peripheral blood. Moreover, soluble ULBP1 attenuated the cytotoxic effects of peripheral blood mononuclear cells on K562, which express membranous ULBP1. Our results suggest that soluble NKG2D ligands can be easy-to-measure biomarkers for the prediction of activity of immune-meditated bone marrow injury in BFS and that soluble NKG2D ligands suppress redundant immune-mediated bone marrow injury.


Subject(s)
Biomarkers/blood , Bone Marrow Failure Disorders/diagnosis , Intracellular Signaling Peptides and Proteins/blood , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/diagnosis , Blood Cell Count , Bone Marrow Failure Disorders/complications , CD2 Antigens/metabolism , Down-Regulation , GPI-Linked Proteins/blood , Hematologic Diseases/complications , Hematologic Diseases/diagnosis , Hemoglobinuria, Paroxysmal/diagnosis , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Young Adult
5.
Br J Haematol ; 146(5): 538-45, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19594748

ABSTRACT

It is considered that a similar immune mechanism acts in the pathogenesis of bone marrow (BM) failure in paroxysmal nocturnal haemoglobinuria (PNH) and its related disorders, such as aplastic anaemia (AA) and myelodysplastic syndromes (MDS). However, the molecular events in immune-mediated marrow injury have not been elucidated. We recently reported an abnormal expression of stress-inducible NKG2D (natural-killer group 2, member D) ligands, such as ULBP (UL16-binding protein) and MICA/B (major histocompatibility complex class I chain-related molecules A/B), on granulocytes in some PNH patients and the granulocyte killing by autologous lymphocytes in vitro. The present study found that the expression of NKG2D ligands was common to both granulocytes and BM cells of patients with PNH, AA, and MDS, indicating their exposure to some incitement to induce the ligands. The haematopoietic colony formation in vitro by the patients' marrow cells significantly improved when their BM cells were pretreated with antibodies against NKG2D receptor, suggesting that the antibodies rescued haematopoietic cells expressing NKG2D ligands from damage by autologous lymphocytes with NKG2D. Clinical courses of patients with PNH and AA showed a close association of the expression of NKG2D ligands with BM failure and a favourable response to immunosuppressive therapy. We therefore propose that NKG2D-mediated immunity may underlie the BM failure in PNH and its-related marrow diseases.


Subject(s)
Anemia, Aplastic/immunology , Antibodies, Monoclonal/pharmacology , Hemoglobinuria, Paroxysmal/immunology , Myelodysplastic Syndromes/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Adult , Aged , Aged, 80 and over , Bone Marrow Cells/immunology , Case-Control Studies , Cells, Cultured , Colony-Forming Units Assay , Female , Flow Cytometry , GPI-Linked Proteins , Granulocytes/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Ligands , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Middle Aged , Minor Histocompatibility Antigens , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Young Adult
6.
Rinsho Ketsueki ; 50(4): 304-8, 2009 Apr.
Article in Japanese | MEDLINE | ID: mdl-19404025

ABSTRACT

We report 2 cases of serum sickness after rituximab infusion. Case 1 is a patient with Waldenström's macroglobulinemia, and case 2 is a patient with marginal-zone B-cell lymphoma of mucosa-associated lymphoid tissue (MALT) type and Sjögren's syndrome. Both patients had polyclonal hypergammaglobulinemia, were treated with rituximab monotherapy, developed serum sickness between 9 and 17 days after the first rituximab infusion, developed fever and arthralgia, and improved soon after corticosteroid treatment. Serum sickness after rituximab treatment for hematological malignancies is very rare as far as we know. We identified three risk factors of serum sickness after rituximab infusion from previous reports and our cases; administration of rituximab alone, the existence of Sjögren's syndrome, and polyclonal hypergammaglobulinemia.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/adverse effects , Lymphoma, B-Cell, Marginal Zone/drug therapy , Serum Sickness/etiology , Waldenstrom Macroglobulinemia/drug therapy , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/administration & dosage , Female , Humans , Hypergammaglobulinemia/complications , Infusions, Intravenous , Lymphoma, B-Cell, Marginal Zone/complications , Middle Aged , Risk Factors , Rituximab , Sjogren's Syndrome/complications
8.
Blood ; 107(3): 1184-91, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16195329

ABSTRACT

The mechanism by which paroxysmal nocturnal hemoglobinuria (PNH) clones expand is unknown. PNH clones harbor PIGA mutations and do not synthesize glycosylphosphatidylinositol (GPI), resulting in deficiency of GPI-linked membrane proteins. GPI-deficient blood cells often expand in patients with aplastic anemia who sustain immune-mediated marrow injury putatively induced by cytotoxic cells, hence suggesting that the injury allows PNH clones to expand selectively. We previously reported that leukemic K562 cells preferentially survived natural killer (NK) cell-mediated cytotoxicity in vitro when they acquired PIGA mutations. We herein show that the survival is ascribable to the deficiency of stress-inducible GPI-linked membrane proteins ULBP1 and ULBP2, which activate NK and T cells. The ULBPs were detected on GPI-expressing but not on GPI-deficient K562 cells. In the presence of antibodies to either the ULBPs or their receptor NKG2D on NK cells, GPI-expressing cells were as less NK sensitive as GPI-deficient cells. NK cells therefore spared ULBP-deficient cells in vitro. The ULBPs were identified only on GPI-expressing blood cells of a proportion of patients with PNH but none of healthy individuals. Granulocytes of the patients partly underwent killing by autologous cytotoxic cells, implying ULBP-associated blood cell injury. In this setting, the lack of ULBPs may allow immunoselection of PNH clones.


Subject(s)
Carrier Proteins/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Membrane Proteins/immunology , Mutation , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/complications , Anemia, Aplastic/genetics , Anemia, Aplastic/immunology , Carrier Proteins/genetics , Erythrocytes/immunology , Female , GPI-Linked Proteins , Glycosylphosphatidylinositols/biosynthesis , Glycosylphosphatidylinositols/deficiency , Glycosylphosphatidylinositols/immunology , Granulocytes/immunology , Hemoglobinuria, Paroxysmal/complications , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/immunology , Histocompatibility Antigens Class I/genetics , Humans , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , K562 Cells , Male , Membrane Proteins/genetics , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Immunologic/immunology , Receptors, Natural Killer Cell , Stress, Physiological/genetics , Stress, Physiological/immunology , T-Lymphocytes/immunology
9.
Blood ; 100(3): 1031-7, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12130519

ABSTRACT

The cloning of the PIG-A gene has facilitated the unraveling of the complex pathophysiology of paroxysmal nocturnal hemoglobinuria (PNH). Of current major concern is the mechanism by which a PNH clone expands. Many reports have suggested that an immune mechanism operates to cause bone marrow failure in some patients with PNH, aplastic anemia, and myelodysplastic syndromes. Because blood cells of PNH phenotype are often found in patients with these marrow diseases, one hypothesis is that the PNH clone escapes immune attack, producing a survival advantage by immunoselection. To test this hypothesis, we examined the sensitivity of blood cells, with or without PIG-A mutations, to killing by natural killer (NK) cells, using 51Cr-release assay in vitro. To both peripheral blood and cultured NK cells, PIG-A mutant cells prepared from myeloid and lymphoid leukemic cell lines were less susceptible than their control counterparts (reverted from the mutant cells by transfection with a PIG-A cDNA). NK activity was completely abolished with concanamycin A and by calcium chelation, indicating that killing was perforin-dependent. There were no differences in major histocompatibility (MHC) class I expression or sensitivity to either purified perforin or to interleukin-2-activated NK cells between PIG-A mutant and control cells. From these results, we infer that PIG-A mutant cells lack molecules needed for NK activation or to trigger perforin-mediated killing. Our experiments suggest that PIG-A mutations confer a relative survival advantage to a PNH clone, contributing to selective expansion of these cells in the setting of marrow injury by cytotoxic lymphocytes.


Subject(s)
Killer Cells, Natural/immunology , Leukemia/pathology , Membrane Proteins/genetics , Mutation/immunology , Cell Survival/genetics , Clone Cells/immunology , Clone Cells/metabolism , Clone Cells/pathology , Cytotoxicity Tests, Immunologic , DNA, Complementary , Hemoglobinuria, Paroxysmal/immunology , Hemoglobinuria, Paroxysmal/pathology , Humans , Interleukin-2/pharmacology , K562 Cells , Leukemia/immunology , Membrane Glycoproteins , Membrane Proteins/immunology , Perforin , Pore Forming Cytotoxic Proteins , Transfection
10.
Blood ; 99(1): 24-9, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11756148

ABSTRACT

Acquired mutations of the PIG-A gene result in the hemolysis characteristic of paroxysmal nocturnal hemoglobinuria (PNH). Although the etiology of the mutation(s) is unclear, mutable conditions have been suggested by the coexistence of multiple clones with different mutations of PIG-A and by the appearance of leukemic clones in patients with PNH. This study sought to test this hypothesis by examining the frequency of hypoxanthine-guanine phosphoribosyl transferase (HPRT) gene mutations, identified by both resistance to 6-thioguanine (6-TG) and gene analysis. T-cell colonies resistant to 6-TG formed in methylcellulose culture were found in 8 (67%) of 12 PNH patients and 3 (18%) of 17 age-matched healthy volunteers (P <.02, Fisher exact probability test). The incidence of resistant colonies ranged from 40 to 367 (mean 149, x 10(-7)) in the 8 patients and from 1 to 16 (mean 7, x 10(-7)) in the 3 healthy donors. Thus, the HRPT gene mutated more frequently in patients with PNH than in healthy controls (P <.02, Mann-Whitney test). Analysis of bone marrow cells supported these findings. Like the PIG-A mutations in PNH, the HPRT mutations were widely distributed in the coding regions and consisted primarily of base deletions. Unlike PNH cells, 6-TG-resistant cells expressed CD59, indicating that the HPRT mutations did not occur in PNH clones. No correlation was noted between HPRT mutation frequency and content of therapy received by the patients. It is concluded that in PNH patients, conditions exist that favor the occurrence of diverse somatic mutations in blood cells.


Subject(s)
Hemoglobinuria, Paroxysmal/enzymology , Hypoxanthine Phosphoribosyltransferase/genetics , Mutation , T-Lymphocytes/enzymology , Adult , Aged , Colony-Forming Units Assay , DNA Mutational Analysis , Drug Resistance , Female , Flow Cytometry , Glycosylphosphatidylinositols/analysis , Hemoglobinuria, Paroxysmal/genetics , Humans , Male , Middle Aged , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , Thioguanine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...