Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 241
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Br J Haematol ; 204(6): 2162-2164, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38719212

RESUMO

Metabolic reprogramming has been investigated in haematological malignancies. To date, a few studies have analysed the metabolic profile of paroxysmal nocturnal haemoglobinuria (PNH). The study by Chen and colleagues sheds light on the involvement of metabolic changes in the proliferation of PNH clones. Commentary on: Chen et al. The histone demethylase JMJD1C regulates CPS1 expression and promotes the proliferation of PNH clones through cell metabolic reprogramming. Br J Haematol 2024;204:2468-2479.


Assuntos
Hemoglobinúria Paroxística , Humanos , Hemoglobinúria Paroxística/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética
2.
Br J Haematol ; 204(6): 2468-2479, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38650379

RESUMO

Paroxysmal nocturnal haemoglobinuria (PNH) is a disorder resulting from erythrocyte membrane deficiencies caused by PIG-A gene mutations. While current treatments alleviate symptoms, they fail to address the underlying cause of the disease-the pathogenic PNH clones. In this study, we found that the expression of carbamoyl phosphate synthetase 1 (CPS1) was downregulated in PNH clones, and the level of CPS1 was negatively correlated with the proportion of PNH clones. Using PIG-A knockout K562 (K562 KO) cells, we demonstrated that CPS1 knockdown increased cell proliferation and altered cell metabolism, suggesting that CPS1 participates in PNH clonal proliferation through metabolic reprogramming. Furthermore, we observed an increase in the expression levels of the histone demethylase JMJD1C in PNH clones, and JMJD1C expression was negatively correlated with CPS1 expression. Knocking down JMJD1C in K562 KO cells upregulated CPS1 and H3K36me3 expression, decreased cell proliferation and increased cell apoptosis. Chromatin immunoprecipitation analysis further demonstrated that H3K36me3 regulated CPS1 expression. Finally, we demonstrated that histone demethylase inhibitor JIB-04 can suppressed K562 KO cell proliferation and reduced the proportion of PNH clones in PNH mice. In conclusion, aberrant regulation of the JMJD1C-H3K36me3-CPS1 axis contributes to PNH clonal proliferation. Targeting JMJD1C with a specific inhibitor unveils a potential strategy for treating PNH patients.


Assuntos
Proliferação de Células , Hemoglobinúria Paroxística , Histona Desmetilases com o Domínio Jumonji , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Animais , Camundongos , Células K562 , Hemoglobinúria Paroxística/patologia , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/metabolismo , Masculino , Feminino , Apoptose , Reprogramação Metabólica , Oxirredutases N-Desmetilantes
3.
J Immunol ; 199(2): 477-488, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28630090

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired disorder originating from hematopoietic stem cells and is a life-threating disease characterized by intravascular hemolysis, bone marrow (BM) failure, and venous thrombosis. The etiology of PNH is a somatic mutation in the phosphatidylinositol glycan class A gene (PIG-A) on the X chromosome, which blocks synthesis of the glycolipid moiety and causes deficiency in GPI-anchored proteins. PNH is closely related to aplastic anemia, in which T cells mediate destruction of BM. To identify aberrant molecular mechanisms involved in immune targeting of hematopoietic stem cells in BM, we applied RNA-seq to examine the transcriptome of T cell subsets (CD4+ naive, CD4+ memory, CD8+ naive, and CD8+ memory) from PNH patients and healthy control subjects. Differentially expressed gene analysis in four different T cell subsets from PNH and healthy control subjects showed distinct transcriptional profiles, depending on the T cell subsets. By pathway analysis, we identified novel signaling pathways in T cell subsets from PNH, including increased gene expression involved in TNFR, IGF1, NOTCH, AP-1, and ATF2 pathways. Dysregulation of several candidate genes (JUN, TNFAIP3, TOB1, GIMAP4, GIMAP6, TRMT112, NR4A2, CD69, and TNFSF8) was validated by quantitative real-time RT-PCR and flow cytometry. We have demonstrated molecular signatures associated with positive and negative regulators in T cells, suggesting novel pathophysiologic mechanisms in PNH. These pathways may be targets for new strategies to modulate T cell immune responses in BM failure.


Assuntos
Hemoglobinúria Paroxística/imunologia , Redes e Vias Metabólicas/genética , Subpopulações de Linfócitos T/imunologia , Transcriptoma , Fator 2 Ativador da Transcrição/genética , Fator 2 Ativador da Transcrição/metabolismo , Adulto , Ligante CD30/genética , Ligante CD30/metabolismo , Linfócitos T CD4-Positivos/imunologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/fisiopatologia , Humanos , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Metiltransferases/genética , Metiltransferases/metabolismo , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Transdução de Sinais/genética , Subpopulações de Linfócitos T/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/genética , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Adulto Jovem
4.
J Biol Chem ; 292(11): 4593-4601, 2017 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-28154186

RESUMO

Naturally occurring mutations in the ribonucleoprotein reverse transcriptase, telomerase, are associated with the bone marrow failure syndromes dyskeratosis congenita, aplastic anemia, and idiopathic pulmonary fibrosis. However, the mechanism by which these mutations impact telomerase function remains unknown. Here we present the structure of the human telomerase C-terminal extension (or thumb domain) determined by the method of single-wavelength anomalous diffraction to 2.31 Å resolution. We also used direct telomerase activity and nucleic acid binding assays to explain how naturally occurring mutations within this portion of telomerase contribute to human disease. The single mutations localize within three highly conserved regions of the telomerase thumb domain referred to as motifs E-I (thumb loop and helix), E-II, and E-III (the FVYL pocket, comprising the hydrophobic residues Phe-1012, Val-1025, Tyr-1089, and Leu-1092). Biochemical data show that the mutations associated with dyskeratosis congenita, aplastic anemia, and idiopathic pulmonary fibrosis disrupt the binding between the protein subunit reverse transcriptase of the telomerase and its nucleic acid substrates leading to loss of telomerase activity and processivity. Collectively our data show that although these mutations do not alter the overall stability or expression of telomerase reverse transcriptase, these rare genetic disorders are associated with an impaired telomerase holoenzyme that is unable to correctly assemble with its nucleic acid substrates, leading to incomplete telomere extension and telomere attrition, which are hallmarks of these diseases.


Assuntos
Anemia Aplástica/genética , Doenças da Medula Óssea/genética , Hemoglobinúria Paroxística/genética , Mutação Puntual , Telomerase/genética , Sequência de Aminoácidos , Anemia Aplástica/metabolismo , Medula Óssea/metabolismo , Doenças da Medula Óssea/metabolismo , Transtornos da Insuficiência da Medula Óssea , Cristalografia por Raios X , Disceratose Congênita/genética , Disceratose Congênita/metabolismo , Células HEK293 , Hemoglobinúria Paroxística/metabolismo , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/metabolismo , Modelos Moleculares , Ácidos Nucleicos/metabolismo , Conformação Proteica , Domínios Proteicos , Telomerase/química , Telomerase/metabolismo , Telômero/metabolismo
5.
Blood ; 128(3): 337-47, 2016 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-27121470

RESUMO

Clonal hematopoiesis (CH) in aplastic anemia (AA) has been closely linked to the evolution of late clonal disorders, including paroxysmal nocturnal hemoglobinuria and myelodysplastic syndromes (MDS)/acute myeloid leukemia (AML), which are common complications after successful immunosuppressive therapy (IST). With the advent of high-throughput sequencing of recent years, the molecular aspect of CH in AA has been clarified by comprehensive detection of somatic mutations that drive clonal evolution. Genetic abnormalities are found in ∼50% of patients with AA and, except for PIGA mutations and copy-neutral loss-of-heterozygosity, or uniparental disomy (UPD) in 6p (6pUPD), are most frequently represented by mutations involving genes commonly mutated in myeloid malignancies, including DNMT3A, ASXL1, and BCOR/BCORL1 Mutations exhibit distinct chronological profiles and clinical impacts. BCOR/BCORL1 and PIGA mutations tend to disappear or show stable clone size and predict a better response to IST and a significantly better clinical outcome compared with mutations in DNMT3A, ASXL1, and other genes, which are likely to increase their clone size, are associated with a faster progression to MDS/AML, and predict an unfavorable survival. High frequency of 6pUPD and overrepresentation of PIGA and BCOR/BCORL1 mutations are unique to AA, suggesting the role of autoimmunity in clonal selection. By contrast, DNMT3A and ASXL1 mutations, also commonly seen in CH in the general population, indicate a close link to CH in the aged bone marrow, in terms of the mechanism for selection. Detection and close monitoring of somatic mutations/evolution may help with prediction and diagnosis of clonal evolution of MDS/AML and better management of patients with AA.


Assuntos
Anemia Aplástica , Hematopoese/genética , Mutação , Proteínas de Neoplasias , Anemia Aplástica/genética , Anemia Aplástica/metabolismo , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
6.
Clin Immunol ; 175: 133-142, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28039017

RESUMO

Monocytes and macrophage combat infections and maintain homeostatic balance by engulfing microbes and apoptotic cells, and releasing inflammatory cytokines. Studies have described that these cells develop anti-inflammatory properties upon recycling the free-hemoglobin (Hb) in hemolytic conditions. While investigating the phenotype of monocytes in two hemolytic disorders-paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease (SCD), we observed a high number of pro-inflammatory (CD14+CD16hi) monocytes in these patients. We further investigated in vitro the phenotype of these monocytes and found an estimated 55% of CD14+ cells were transformed into the CD14+CD16hi subset after engulfing Hb-activated platelets. The CD14+CD16hi monocytes, which were positive for both intracellular Hb and CD42b (platelet marker), secreted significant amounts of TNF-α and IL-1ß, unlike monocytes treated with only free Hb, which secreted more IL-10. We have shown recently the presence of a high number of Hb-bound hyperactive platelets in patients with both diseases, and further investigated if the monocytes engulfed these activated platelets in vivo. As expected, we found 95% of CD14+CD16hi monocytes with both intracellular Hb and CD42b in both diseases, and they expressed high TNF-α. Furthermore our data showed that these monocytes whether from patients or developed in vitro after treatment with Hb-activated platelets, secreted significant amounts of tissue factor. Besides, these CD14+CD16hi monocytes displayed significantly decreased phagocytosis of E. coli. Our study therefore suggests that this alteration of monocyte phenotype may play a role in the increased propensity to pro-inflammatory/coagulant complications observed in these hemolytic disorders-PNH and SCD.


Assuntos
Anemia Falciforme/patologia , Plaquetas/patologia , Hemoglobinas/metabolismo , Hemoglobinúria Paroxística/patologia , Inflamação/patologia , Monócitos/patologia , Anemia Falciforme/metabolismo , Plaquetas/metabolismo , Hemoglobinúria Paroxística/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Monócitos/metabolismo , Fenótipo , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Receptores de IgG/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Haematologica ; 102(3): 466-475, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27810992

RESUMO

Paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome are diseases of excess activation of the alternative pathway of complement that are treated with eculizumab, a humanized monoclonal antibody against the terminal complement component C5. Eculizumab must be administered intravenously, and moreover some patients with paroxysmal nocturnal hemoglobinuria on eculizumab have symptomatic extravascular hemolysis, indicating an unmet need for additional therapeutic approaches. We report the activity of two novel small-molecule inhibitors of the alternative pathway component Factor D using in vitro correlates of both paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. Both compounds bind human Factor D with high affinity and effectively inhibit its proteolytic activity against purified Factor B in complex with C3b. When tested using the traditional Ham test with cells from paroxysmal nocturnal hemoglobinuria patients, the Factor D inhibitors significantly reduced complement-mediated hemolysis at concentrations as low as 0.01 µM. Additionally the compound ACH-4471 significantly decreased C3 fragment deposition on paroxysmal nocturnal hemoglobinuria erythrocytes, indicating a reduced potential relative to eculizumab for extravascular hemolysis. Using the recently described modified Ham test with serum from patients with atypical hemolytic uremic syndrome, the compounds reduced the alternative pathway-mediated killing of PIGA-null reagent cells, thus establishing their potential utility for this disease of alternative pathway of complement dysregulation and validating the modified Ham test as a system for pre-clinical drug development for atypical hemolytic uremic syndrome. Finally, ACH-4471 blocked alternative pathway activity when administered orally to cynomolgus monkeys. In conclusion, the small-molecule Factor D inhibitors show potential as oral therapeutics for human diseases driven by the alternative pathway of complement, including paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/etiologia , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Fator D do Complemento/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Via Alternativa do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/imunologia , Hemoglobinúria Paroxística/etiologia , Hemoglobinúria Paroxística/metabolismo , Adulto , Idoso , Animais , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Biomarcadores , Complemento C3/imunologia , Complemento C3/metabolismo , Fator D do Complemento/imunologia , Fator D do Complemento/metabolismo , Inativadores do Complemento/administração & dosagem , Citotoxicidade Imunológica , Modelos Animais de Doenças , Eritrócitos/imunologia , Eritrócitos/metabolismo , Feminino , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/tratamento farmacológico , Hemólise , Humanos , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Ligação Proteica , Proteólise , Resultado do Tratamento , Adulto Jovem
9.
Rinsho Ketsueki ; 58(4): 353-362, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-28484166

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) manifests by clonal expansion of mutant hematopoietic stem cells (HSCs) bearing a somatic mutation in the X-linked PIGA gene. PIGA mutations cause defective biosynthesis of GPI and cell surface deficiency of GPI-anchored proteins such as DAF and CD59, leading to intravascular hemolysis and thrombosis. These two major symptoms of PNH can be controlled by eculizumab, an anti-C5 monoclonal antibody. Bone marrow failure, the third major symptom of PNH, is autoimmune-mediated and contributes to the clonal expansion of GPI-defective HSCs by selectively attacking GPI-positive wild-type HSCs. GPI-defective erythrocytes, being protected from intravascular hemolysis by eculizumab, accumulate C3-derived fragments, C3b, iC3b, and C3dg, because of DAF deficiency and in turn become susceptible to CR3-mediated phagocytosis by spleen macrophages. Approximately 3% of Japanese patients with PNH are refractory to eculizumab therapy. Approximately 3% of Japanese people are heterozygous for a single nucleotide polymorphism that changes an amino acid near the eculizumab binding site. New therapeutic measures are needed to solve these issues.


Assuntos
Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/metabolismo , Anemia Hemolítica/complicações , Animais , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/tratamento farmacológico , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação , Ligação Proteica
11.
Blood ; 124(20): 3151-4, 2014 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25293775

RESUMO

A rare point mutation in the core promoter -270GC-rich box of PIGM, a housekeeping gene, disrupts binding of the generic transcription factor (TF) Sp1 and causes inherited glycosylphosphatidylinositol (GPI) deficiency (IGD). We show that whereas PIGM messenger RNA levels and surface GPI expression in IGD B cells are low, GPI expression is near normal in IGD erythroid cells. This divergent phenotype results from differential promoter chromatin accessibility and binding of Sp1. Specifically, whereas PIGM transcription in B cells is dependent on Sp1 binding to the -270GC-rich box and is associated with lower promoter accessibility, in erythroid cells, Sp1 activates PIGM transcription by binding upstream of (but not to) the -270GC-rich box. These findings explain intact PIGM transcription in IGD erythroid cells and the lack of clinically significant intravascular hemolysis in patients with IGD. Furthermore, they provide novel insights into tissue-specific transcriptional control of a housekeeping gene by a generic TF.


Assuntos
Glicosilfosfatidilinositóis/deficiência , Hemoglobinúria Paroxística/genética , Manosiltransferases/genética , Ativação Transcricional , Linfócitos B/metabolismo , Linfócitos B/patologia , Eritrócitos/metabolismo , Eritrócitos/patologia , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/patologia , Humanos , Mutação , Fenótipo , Regiões Promotoras Genéticas , Convulsões , Fator de Transcrição Sp1/metabolismo
12.
Blood ; 123(13): 2094-101, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24497537

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by complement-mediated intravascular hemolysis due to the lack of CD55 and CD59 on affected erythrocytes. The anti-C5 antibody eculizumab has proven clinically effective, but uncontrolled C3 activation due to CD55 absence may result in opsonization of erythrocytes, possibly leading to clinically meaningful extravascular hemolysis. We investigated the effect of the peptidic C3 inhibitor, compstatin Cp40, and its long-acting form (polyethylene glycol [PEG]-Cp40) on hemolysis and opsonization of PNH erythrocytes in an established in vitro system. Both compounds demonstrated dose-dependent inhibition of hemolysis with IC50 ∼4 µM and full inhibition at 6 µM. Protective levels of either Cp40 or PEG-Cp40 also efficiently prevented deposition of C3 fragments on PNH erythrocytes. We further explored the potential of both inhibitors for systemic administration and performed pharmacokinetic evaluation in nonhuman primates. A single intravenous injection of PEG-Cp40 resulted in a prolonged elimination half-life of >5 days but may potentially affect the plasma levels of C3. Despite faster elimination kinetics, saturating inhibitor concentration could be reached with unmodified Cp40 through repetitive subcutaneous administration. In conclusion, peptide inhibitors of C3 activation effectively prevent hemolysis and C3 opsonization of PNH erythrocytes, and are excellent, and potentially cost-effective, candidates for further clinical investigation.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Complemento C3/antagonistas & inibidores , Hemoglobinúria Paroxística/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Animais , Desenho de Fármacos , Eritrócitos/efeitos dos fármacos , Eritrócitos/fisiologia , Meia-Vida , Hemoglobinúria Paroxística/metabolismo , Hemólise/efeitos dos fármacos , Humanos , Macaca fascicularis , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Polietilenoglicóis/farmacologia , Polietilenoglicóis/uso terapêutico
13.
Eur J Haematol ; 97(3): 261-70, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26660446

RESUMO

Heterozygous mutations in the transcriptional regulator GATA-2 associate with multilineage immunodeficiency, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML). The majority of these mutations localize in the zinc finger (ZnF) domains, which mediate GATA-2 DNA binding. Deregulated hematopoiesis with GATA-2 mutation frequently develops in adulthood, yet GATA-2 function in the bone marrow remains unresolved. To investigate this, we conditionally deleted the GATA-2 C-terminal ZnF (C-ZnF) coding sequences in adult mice. Upon Gata2 C-ZnF deletion, we observed rapid peripheral cytopenia, bone marrow failure, and decreased c-Kit expression on hematopoietic progenitors. Transplant studies indicated GATA-2 has a cell-autonomous role in bone marrow hematopoiesis. Moreover, myeloid lineage populations were particularly sensitive to Gata2 hemizygosity, while molecular assays indicated GATA-2 regulates c-Kit expression in multilineage progenitor cells. Enforced c-Kit expression in Gata2 C-ZnF-deficient hematopoietic progenitors enhanced myeloid colony activity, suggesting GATA-2 sustains myelopoiesis via a cell intrinsic role involving maintenance of c-Kit expression. Our results provide insight into mechanisms regulating hematopoiesis in bone marrow and may contribute to a better understanding of immunodeficiency and bone marrow failure associated with GATA-2 mutation.


Assuntos
Anemia Aplástica/genética , Doenças da Medula Óssea/genética , Medula Óssea/patologia , Fator de Transcrição GATA2/genética , Hemoglobinúria Paroxística/genética , Domínios e Motivos de Interação entre Proteínas/genética , Proteínas Proto-Oncogênicas c-kit/deficiência , Deleção de Sequência , Dedos de Zinco/genética , Anemia Aplástica/diagnóstico , Anemia Aplástica/metabolismo , Anemia Aplástica/mortalidade , Animais , Biomarcadores , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Doenças da Medula Óssea/diagnóstico , Doenças da Medula Óssea/metabolismo , Doenças da Medula Óssea/mortalidade , Transtornos da Insuficiência da Medula Óssea , Osso e Ossos/patologia , Imunoprecipitação da Cromatina , Descalcificação Patológica/genética , Modelos Animais de Doenças , Fator de Transcrição GATA2/química , Fator de Transcrição GATA2/metabolismo , Expressão Gênica , Regulação da Expressão Gênica , Frequência do Gene , Genes Reporter , Genótipo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/mortalidade , Sequenciamento de Nucleotídeos em Larga Escala , Imunofenotipagem , Camundongos , Camundongos Knockout , Prognóstico , Células da Side Population
14.
Rinsho Ketsueki ; 57(10): 1900-1907, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-27725586

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired GPI deficiency caused by somatic mutation of the PIGA gene in one or several hematopoietic stem cells. Recently, PNH caused by somatic mutation of one allele of the PIGT gene in combination with a germline mutation of the other allele was reported, showing that PIGA is not the only gene responsible for PNH, though other causes are rare. These mutant cells become GPI deficient, expand clonally and differentiate into all of the hematopoietic lineages. When GPI deficient erythrocytes increase in proportion, massive hemolysis occurs due to activated complement attack during infection. As the complement regulatory proteins such as CD59 and DAF are GPI anchored proteins, they are defective on GPI deficient erythrocytes and these abnormal erythrocytes are thereby left unprotected from complement attack. Hemolytic anemia, venous thrombosis, and bone marrow failure are thus the resulting triad of symptoms. Clonal expansion does not occur with PIGA deficiency alone. We hypothesize that PIGA deficient cells acquire a proliferative phenotype via additional gene mutations within the associated environment of bone marrow failure. This hypothesis will be explained by introducing recent reports.


Assuntos
Hemoglobinúria Paroxística , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Exoma , Glicosilfosfatidilinositóis/deficiência , Glicosilfosfatidilinositóis/metabolismo , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/terapia , Hemólise , Humanos , Análise de Sequência de DNA
15.
J Cell Mol Med ; 19(9): 2193-201, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26033571

RESUMO

The glycolipid glycosylphosphatidylinositol anchor (GPI-A) plays an important role in lipid raft formation, which is required for proper expression on the cell surface of two inhibitors of the complement cascade, CD55 and CD59. The absence of these markers from the surface of blood cells, including erythrocytes, makes the cells susceptible to complement lysis, as seen in patients suffering from paroxysmal nocturnal haemoglobinuria (PNH). However, the explanation for why PNH-affected hematopoietic stem/progenitor cells (HSPCs) expand over time in BM is still unclear. Here, we propose an explanation for this phenomenon and provide evidence that a defect in lipid raft formation in HSPCs leads to defective CXCR4- and VLA-4-mediated retention of these cells in BM. In support of this possibility, BM-isolated CD34(+) cells from PNH patients show a defect in the incorporation of CXCR4 and VLA-4 into membrane lipid rafts, respond weakly to SDF-1 stimulation, and show defective adhesion to fibronectin. Similar data were obtained with the GPI-A(-) Jurkat cell line. Moreover, we also report that chimeric mice transplanted with CD55(-/-)  CD59(-/-) BM cells but with proper GPI-A expression do not expand over time in transplanted hosts. On the basis of these findings, we propose that a defect in lipid raft formation in PNH-mutated HSPCs makes these cells more mobile, so that they expand and out-compete normal HSPCs from their BM niches over time.


Assuntos
Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/patologia , Microdomínios da Membrana/metabolismo , Animais , Antígenos CD/metabolismo , Toxinas Bacterianas/metabolismo , Medula Óssea/patologia , Adesão Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Quimiotaxia/efeitos dos fármacos , Fibronectinas/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Humanos , Integrina alfa4beta1/metabolismo , Células Jurkat , Microdomínios da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Hum Mol Genet ; 22(16): 3187-94, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23591993

RESUMO

How the transcription repressing complex Polycomb interacts with transcriptional regulators at housekeeping genes in somatic cells is not well understood. By exploiting a CpG island (CGI) point mutation causing a Mendelian disease, we show that DNA binding of activating transcription factor (TF) determines histone acetylation and nucleosomal depletion commensurate with Polycomb exclusion from the target promoter. Lack of TF binding leads to reversible transcriptional repression imposed by nucleosomal compaction and consolidated by Polycomb recruitment and establishment of bivalent chromatin status. Thus, within a functional hierarchy of transcriptional regulators, TF binding is the main determinant of Polycomb recruitment to the CGI of a housekeeping gene in somatic cells.


Assuntos
Fatores Ativadores da Transcrição/química , Linfócitos B/química , Linfócitos B/metabolismo , Ilhas de CpG/genética , Glicosilfosfatidilinositóis/deficiência , Hemoglobinúria Paroxística/genética , Manosiltransferases/genética , Proteínas do Grupo Polycomb/metabolismo , Fatores Ativadores da Transcrição/genética , Fatores Ativadores da Transcrição/metabolismo , Sequência de Bases , Células Cultivadas , Metilação de DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Genes Essenciais , Glicosilfosfatidilinositóis/genética , Glicosilfosfatidilinositóis/metabolismo , Hemoglobinúria Paroxística/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Manosiltransferases/metabolismo , Dados de Sequência Molecular , Nucleossomos/metabolismo , Mutação Puntual , Proteínas do Grupo Polycomb/química , Proteínas do Grupo Polycomb/genética , Regiões Promotoras Genéticas , Ligação Proteica , Convulsões
17.
Blood ; 121(14): 2753-61, 2013 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-23372165

RESUMO

The mechanism of bone marrow failure (BMF) in paroxysmal nocturnal hemoglobinuria (PNH) is not yet known. Because in PNH the biosynthesis of the glycolipid molecule glycosylphosphatidylinositol (GPI) is disrupted in hematopoietic stem and progenitor cells by a somatic mutation in the PIG-A gene, BMF might result from an autoimmune attack, whereby T cells target GPI in normal cells, whereas PIG-A mutant GPI-negative cells are spared. In a deliberate test of this hypothesis, we have demonstrated in PNH patients the presence of CD8(+) T cells reactive against antigen-presenting cells (APCs) loaded with GPI. These T cells were significantly more abundant in PNH patients than in healthy controls; their reactivity depended on CD1d expression and they increased upon coculture with CD1d-expressing, GPI-positive APCs. In GPI-specific T cells captured by CD1d dimer technology, we identified, through global T-cell receptor α (TCRα) analysis, an invariant TCRVα21 sequence, which was then found at frequencies higher than background in the TCR repertoire of 6 of 11 PNH patients. Thus, a novel, autoreactive, CD1d-restricted, GPI-specific T-cell population, enriched in an invariant TCRα chain, is expanded in PNH patients and may be responsible for BMF in PNH.


Assuntos
Antígenos CD1d/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Hemoglobinúria Paroxística/imunologia , Adulto , Idoso , Anemia Aplástica , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD1d/química , Antígenos CD1d/imunologia , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Linfócitos T CD8-Positivos/citologia , Técnicas de Cocultura , Dimerização , Feminino , Citometria de Fluxo , Biblioteca Gênica , Glicosilfosfatidilinositóis/química , Glicosilfosfatidilinositóis/farmacologia , Hemoglobinúria Paroxística/metabolismo , Humanos , Células K562 , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Adulto Jovem
18.
Haematologica ; 100(12): 1546-52, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26315930

RESUMO

Acquired aplastic anemia is an immune-mediated disease characterized by severe defects in stem cell number resulting in hypocellular marrow and peripheral blood cytopenias. Minor paroxysmal nocturnal hemoglobinuria populations and a short telomere length were identified as predictive biomarkers of immunosuppressive therapy responsiveness in aplastic anemia. We enrolled 113 aplastic anemia patients (63 boys and 50 girls) in this study to evaluate their response to immunosuppressive therapy. The paroxysmal nocturnal hemoglobinuria populations and telomere length were detected by flow cytometry. Forty-seven patients (42%) carried a minor paroxysmal nocturnal hemoglobinuria population. The median telomere length of aplastic anemia patients was -0.99 standard deviation (SD) (range -4.01-+3.01 SD). Overall, 60 patients (53%) responded to immunosuppressive therapy after six months. Multivariate logistic regression analysis identified the absence of a paroxysmal nocturnal hemoglobinuria population and a shorter telomere length as independent unfavorable predictors of immunosuppressive therapy response at six months. The cohort was stratified into a group of poor prognosis (paroxysmal nocturnal hemoglobinuria negative and shorter telomere length; 37 patients) and good prognosis (paroxysmal nocturnal hemoglobinuria positive and/or longer telomere length; 76 patients), respectively. The response rates of the poor prognosis and good prognosis groups at six months were 19% and 70%, respectively (P<0.001). The combined absence of a minor paroxysmal nocturnal hemoglobinuria population and a short telomere length is an efficient predictor of poor immunosuppressive therapy response, which should be considered while deciding treatment options: immunosuppressive therapy or first-line hematopoietic stem cell transplantation. The trial was registered in www.umin.ac.jp with number UMIN000017972.


Assuntos
Anemia Aplástica , Hemoglobinúria Paroxística , Terapia de Imunossupressão , Homeostase do Telômero , Telômero/metabolismo , Adolescente , Anemia Aplástica/diagnóstico , Anemia Aplástica/metabolismo , Anemia Aplástica/terapia , Criança , Pré-Escolar , Feminino , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/terapia , Humanos , Lactente , Masculino , Prognóstico
19.
Eur J Haematol ; 95(3): 230-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25404431

RESUMO

Trisomy 8 (+8), one of the most common chromosomal abnormalities found in patients with myelodysplastic syndromes (MDS), is occasionally seen in patients with otherwise typical aplastic anemia (AA). Although some studies have indicated that the presence of +8 is associated with the immune pathophysiology of bone marrow (BM) failure, its pathophysiology may be heterogeneous. We studied 53 patients (22 with AA and 31 with low-risk MDS) with +8 for the presence of increased glycosylphosphatidylinositol-anchored protein-deficient (GPI-AP(-) ) cells, their response to immunosuppressive therapy (IST), and their prognosis. A significant increase in the percentage of GPI-AP(-) cells was found in 14 (26%) of the 53 patients. Of the 26 patients who received IST, including nine with increased GPI-AP(-) cells and 17 without increased GPI-AP(-) cells, 14 (88% with increased GPI-AP(-) cells and 41% without increased GPI-AP(-) cells) improved. The overall and event-free survival rates of the +8 patients with and without increased GPI-AP(-) cells at 5 yr were 100% and 100% and 59% and 57%, respectively. Examining the peripheral blood for the presence of increased GPI-AP(-) cells may thus be helpful for choosing the optimal treatment for +8 patients with AA or low-risk MDS.


Assuntos
Proteínas Ligadas por GPI/deficiência , Granulócitos/metabolismo , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/metabolismo , Trissomia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anemia Aplástica , Medula Óssea/metabolismo , Medula Óssea/patologia , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Cromossomos Humanos Par 8 , Feminino , Proteínas Ligadas por GPI/metabolismo , Hemoglobinúria Paroxística/diagnóstico , Hemoglobinúria Paroxística/tratamento farmacológico , Hemoglobinúria Paroxística/epidemiologia , Hemoglobinúria Paroxística/mortalidade , Humanos , Imunossupressores/uso terapêutico , Incidência , Masculino , Pessoa de Meia-Idade , Prevalência , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento , Adulto Jovem
20.
Curr Top Membr ; 75: 269-303, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26015286

RESUMO

Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) are a class of lipid-anchored proteins attached to the membranes by a glycolipid anchor that is added, as posttranslation modification, in the endoplasmic reticulum. GPI-APs are expressed at the cell surface of eukaryotes where they play diverse vital functions. Like all plasma membrane proteins, GPI-APs must be correctly sorted along the different steps of the secretory pathway to their final destination. The presence of both a glycolipid anchor and a protein portion confers special trafficking features to GPI-APs. Here, we discuss the recent advances in the field of GPI-AP trafficking, focusing on the mechanisms regulating their biosynthetic pathway and plasma membrane organization. We also discuss how alterations of these mechanisms can result in different diseases. Finally, we will examine the strict relationship between the trafficking and function of GPI-APs in epithelial cells.


Assuntos
Anormalidades Múltiplas/metabolismo , Membrana Celular/metabolismo , Proteínas Ligadas por GPI/metabolismo , Hemoglobinúria Paroxística/metabolismo , Deficiência Intelectual/metabolismo , Neoplasias/metabolismo , Distúrbios do Metabolismo do Fósforo/metabolismo , Anormalidades Múltiplas/genética , Membrana Celular/ultraestrutura , Desenvolvimento Embrionário/genética , Retículo Endoplasmático/metabolismo , Células Epiteliais/metabolismo , Proteínas Ligadas por GPI/genética , Glicosilfosfatidilinositóis/metabolismo , Complexo de Golgi/metabolismo , Hemoglobinúria Paroxística/genética , Humanos , Deficiência Intelectual/genética , Microdomínios da Membrana/metabolismo , Neoplasias/genética , Distúrbios do Metabolismo do Fósforo/genética , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA