Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 195
Filter
2.
Leuk Lymphoma ; 63(1): 199-204, 2022 01.
Article in English | MEDLINE | ID: mdl-34448437

ABSTRACT

Myelodysplastic syndrome/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T) is characterized by anemia, ring sideroblast erythroid precursors, and persistent thrombocytosis. Case reports suggest lenalidomide may be effective in treating MDS/MPN-RS-T. We evaluated a large series of patients with MDS/MPN-RS-T to compare hematological improvement (HI) response rates among different drug therapies including lenalidomide. We identified 167 patients with MDS/MPN-RS-T. Among the patients tested, 84% had SF3B1 mutations and 43% had JAK2 V617F mutations. The median OS for the cohort was 81 months. Overall, 76 patients (46%) received erythropoiesis-stimulating agents (ESAs), 47 patients (28%) received lenalidomide, and 45 patients (27%) received hypomethylating agents (HMAs). The HI rates were 58%, 53%, and 24%, respectively. The median duration of treatment was 11 months for lenalidomide compared to 6 months for HMAs. Rates of HI improvement were higher in patients with MDS/MPN-RS-T treated with ESAs or lenalidomide, in comparison to those treated with HMAs.


Subject(s)
Anemia, Sideroblastic , Myelodysplastic-Myeloproliferative Diseases , Neoplasms , Thrombocytosis , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/drug therapy , Anemia, Sideroblastic/etiology , Humans , Mutation , Myelodysplastic-Myeloproliferative Diseases/complications , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Myelodysplastic-Myeloproliferative Diseases/drug therapy , Thrombocytosis/drug therapy , Thrombocytosis/genetics , Treatment Outcome
3.
Front Immunol ; 12: 586320, 2021.
Article in English | MEDLINE | ID: mdl-33936027

ABSTRACT

Since the first description of the syndrome of sideroblastic anemia with immunodeficiency, fevers and development delay (SIFD), clinical pictures lacking both neurological and hematological manifestations have been reported. Moreover, prominent skin involvement, such as with relapsing erythema nodosum, is not a common finding. Up to this moment, no genotype and phenotype correlation could be done, but mild phenotypes seem to be located in the N or C part. B-cell deficiency is a hallmark of SIFD syndrome, and multiple others immunological defects have been reported, but not high levels of double negative T cells. Here we report a Brazilian patient with a novel phenotype of SFID syndrome, carrying multiple immune defects and harboring a novel mutation on TRNT1 gene.


Subject(s)
Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/etiology , Developmental Disabilities/diagnosis , Disease Susceptibility , Fever , Immunologic Deficiency Syndromes/diagnosis , Phenotype , Alleles , Biopsy , DNA Mutational Analysis , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Mutation
4.
Am J Hematol ; 96(3): 379-394, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33428785

ABSTRACT

DISEASE OVERVIEW: Ring sideroblasts (RS) are erythroid precursors with abnormal perinuclear mitochondrial iron accumulation. Two myeloid neoplasms defined by the presence of RS, include myelodysplastic syndromes with RS (MDS-RS) and MDS/myeloproliferative neoplasm with RS and thrombocytosis (MDS/MPN-RS-T). DIAGNOSIS: MDS-RS is a lower risk MDS, with single or multilineage dysplasia (MDS-RS-SLD/MLD), <5% bone marrow (BM) blasts, <1% peripheral blood blasts and ≥15% BM RS (≥5% in the presence of SF3B1 mutations). MDS/MPN-RS-T, now a formal entity in the MDS/MPN overlap syndromes, has diagnostic features of MDS-RS-SLD, along with a platelet count ≥450 × 109 /L and large atypical megakaryocytes. MUTATIONS AND KARYOTYPE: Mutations in SF3B1 are seen in ≥80% of patients with MDS-RS-SLD and MDS/MPN-RS-T, and strongly correlate with the presence of BM RS; MDS/MPN-RS-T patients also demonstrate JAK2V617F (50%), DNMT3A, TET2 and ASXL1 mutations. Cytogenetic abnormalities are uncommon in both. RISK STRATIFICATION: Most patients with MDS-RS-SLD are stratified into lower risk groups by the revised-IPSS. Disease outcome in MDS/MPN-RS-T is better than that of MDS-RS-SLD, but worse than that of essential thrombocythemia (MPN). Both diseases are associated with a low risk of leukemic transformation. TREATMENT: Anemia and iron overload are complications seen in both and are managed similar to lower risk MDS and MPN. Luspatercept, a first-in-class erythroid maturation agent is now approved for the management of anemia in patients with MDS-RS and MDS/MPN-RS-T. Aspirin therapy is reasonable in MDS/MPN-RS-T, especially in the presence of JAK2V617F, but the value of platelet-lowering drugs remains to be defined.


Subject(s)
Anemia, Sideroblastic , Myelodysplastic-Myeloproliferative Diseases , Allografts , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/etiology , Anemia, Sideroblastic/pathology , Anemia, Sideroblastic/therapy , Bone Marrow/pathology , Cell Lineage , Clone Cells/pathology , Combined Modality Therapy , DNA Methylation/drug effects , Disease Management , Erythroblasts/ultrastructure , Ferritins/analysis , Hematinics/therapeutic use , Hematopoietic Stem Cell Transplantation , Humans , Iron Chelating Agents/therapeutic use , Mitochondria/chemistry , Mutation , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Myelodysplastic-Myeloproliferative Diseases/genetics , Myelodysplastic-Myeloproliferative Diseases/therapy , Phosphoproteins/genetics , Prognosis , RNA Splicing Factors/genetics , Risk Assessment , Thrombocytosis/diagnosis , Thrombocytosis/therapy
5.
Probl Radiac Med Radiobiol ; 25: 390-401, 2020 Dec.
Article in English, Ukrainian | MEDLINE | ID: mdl-33361849

ABSTRACT

OBJECTIVE: To determine the influence of iron metabolism on the prognosis of acute lymphoblastic (ALL) and (AML)myeloblastic leukemia at the different phases of chemotherapy in children after Chоrnobyl accident. MATERIALS AND METHODS: 333 children (295 - ALL, 38 - AML) were examined at the stages of chemotherapy. Thecomparison group included 93 children without leukemia. Acute leukemia variants, patients survival, relapses, thenature of disease (live child or died), iron methabolism (morphometric parameters of erythrocytes, SI, SF, STf, TS),manifestations of dyserythropoiesis, bone marrow sideroblast and patients radiation dose were taken into account. RESULTS: In 295 patients with ALL the following variants of leukemia were established: pro-B-ALL in 23, «common¼type of ALL in 224, pre-B-ALL in 29, T-ALL in 19. Thirty eight patients were diagnosed with AML (11 - M1, 19 - M2,8 - M4). Doses of radiation in patients with AL were (2.78 ± 0.10) mSv and they did not correlate with clinical andhematological parameters, disease variant. Relapse rates and shorter survival were in patients with T-ALL, pro-B-ALLand AML with SF levels > 500 ng/ml (p < 0.05). The amount of children with normochromic-normocytic anemias andmanifestations of dysplasia of erythroid lineage elements was greater in the AML than in ALL. SF content in patientswas elevated during chemotherapy and was lower than the initial one only in the remission period. Transferrin wasreliably overloaded with iron: TS (70.2 ± 2.3) % compared with the control group (32.7 ± 2.1) %. Correlationbetween TS and survival of patients was detected (rs = -0.45). Direct correlation between the number of iron granules in erythrocariocytes and SF level (rs = 0.43) was established, indicating the phenomena of ineffective erythropoiesis. CONCLUSIONS: The negative influence of iron excess in the patients body on the hemopoiesis function, manifestations of ineffective erythropoiesis and the course of acute leukemia in children have been established. Changes inferrokinetic processes in children can be the basis of leukemоgenesis development.


Subject(s)
Anemia, Sideroblastic/blood , Chernobyl Nuclear Accident , Erythropoiesis/radiation effects , Iron/blood , Leukemia, Myeloid, Acute/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Adolescent , Anemia, Sideroblastic/drug therapy , Anemia, Sideroblastic/etiology , Anemia, Sideroblastic/mortality , Antineoplastic Agents/therapeutic use , Bone Marrow/pathology , Bone Marrow/radiation effects , Child , Child, Preschool , Erythroid Cells/pathology , Erythroid Cells/radiation effects , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/mortality , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Radiation Exposure/adverse effects , Radiation, Ionizing , Recurrence , Remission Induction , Survival Analysis , Transferrin/metabolism , Ukraine/epidemiology
8.
Hematol Oncol Clin North Am ; 34(2): 401-420, 2020 04.
Article in English | MEDLINE | ID: mdl-32089219

ABSTRACT

Sideroblastic anemias are a heterogeneous group of disorders unified by the presence of abnormal erythroid precursors with perinuclear mitochondrial iron deposition in the bone marrow. Based on etiology, they are classified into clonal and nonclonal. Clonal sideroblastic anemias refer to myeloid neoplasms with ring sideroblasts (RS) and frequently have somatic perturbations in the SF3B1 gene. Anemia is a major cause of morbidity in patients, and restoration of effective erythropoiesis is a major treatment goal. Morbidity includes transfusion and disease-related complications. This article focuses on treatment of acquired sideroblastic anemias and highlights areas of future investigation.


Subject(s)
Anemia, Sideroblastic/therapy , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/etiology , Biomarkers , Clonal Evolution , Combined Modality Therapy , Disease Management , Disease Progression , Disease Susceptibility , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Erythroid Cells/pathology , Erythropoiesis/drug effects , Hematinics/pharmacology , Hematinics/therapeutic use , Humans , Hyperplasia , Immunologic Factors/immunology , Immunologic Factors/therapeutic use , Mutation , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/therapy , Treatment Outcome
10.
Int J Lab Hematol ; 41(3): 345-352, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30811101

ABSTRACT

INTRODUCTION: Myelodysplastic/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T) is a rare disease in the 2016 revised World Health Organization (WHO) classification. Diagnostic criteria include the following: persistent thrombocytosis (>450 × 109 /L) with clustering of atypical megakaryocytes, refractory anemia, dyserythropoiesis with ring sideroblasts, and the presence of the spliceosome factor 3b subunit (SF3B1) mutation. It is unclear if anemia should be a required criterion for this diagnosis as cases which show all other features of MDS/MPN-RS-T but without anemia exist. METHODS: We searched for borderline cases of MDS/MPN-RS-T in which refractory anemia was absent at diagnosis in two major academic institutes. RESULTS: Three cases without anemia were identified. These cases all showed other classic morphologic and clinical features of MDS/MPN-RS-T, including thrombocytosis, atypical megakaryocytes with clustering, and characteristic SF3B1 and JAK2 V617F mutations. CONCLUSION: Given these findings, the requirement of refractory anemia as a diagnostic criterion for MDS/MPN-RS-T should be re-evaluated. Removal of refractory anemia as a diagnostic criterion would incorporate current borderline cases and extend the spectrum of this disorder.


Subject(s)
Anemia, Sideroblastic/blood , Myelodysplastic-Myeloproliferative Diseases/blood , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Thrombocytosis/blood , Aged, 80 and over , Anemia, Sideroblastic/etiology , Biomarkers , Biopsy , Bone Marrow/pathology , Bone Marrow Cells/pathology , Cytogenetic Analysis , Female , Flow Cytometry , High-Throughput Nucleotide Sequencing , Humans , Janus Kinase 2/genetics , Male , Middle Aged , Mutation , Myelodysplastic-Myeloproliferative Diseases/complications , Myelodysplastic-Myeloproliferative Diseases/etiology , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Sequence Analysis, DNA , Thrombocytosis/etiology
12.
JAMA Neurol ; 74(6): 686-694, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28395030

ABSTRACT

Importance: YARS2 mutations have been associated with a clinical triad of myopathy, lactic acidosis, and sideroblastic anemia in predominantly Middle Eastern populations. However, the identification of new patients expands the clinical and molecular spectrum of mitochondrial disorders. Objectives: To review the clinical, molecular, and genetic features of YARS2-related mitochondrial disease and to demonstrate a new Scottish founder variant. Design, Setting, and Participants: An observational case series study was conducted at a national diagnostic center for mitochondrial disease in Newcastle upon Tyne, England, and review of cases published in the literature. Six adults in a well-defined mitochondrial disease cohort and 11 additional cases described in the literature were identified with YARS2 variants between January 1, 2000, and January 31, 2015. Main Outcome and Measures: The spectrum of clinical features and disease progression in unreported and reported patients with pathogenic YARS2 variants. Results: Seventeen patients (median [interquartile range] age at onset, 1.5 [9.8] years) with YARS2-related mitochondrial myopathy were identified. Fifteen individuals (88%) exhibited an elevated blood lactate level accompanied by generalized myopathy; only 12 patients (71%) manifested with sideroblastic anemia. Hypertrophic cardiomyopathy (9 [53%]) and respiratory insufficiency (8 [47%]) were also prominent clinical features. Central nervous system involvement was rare. Muscle studies showed global cytochrome-c oxidase deficiency in all patients tested and severe, combined respiratory chain complex activity deficiencies. Microsatellite genotyping demonstrated a common founder effect shared between 3 Scottish patients with a p.Leu392Ser variant. Immunoblotting from fibroblasts and myoblasts of an affected Scottish patient showed normal YARS2 protein levels and mild respiratory chain complex defects. Yeast modeling of novel missense YARS2 variants closely correlated with the severity of clinical phenotypes. Conclusions and Relevance: The p.Leu392Ser variant is likely a newly identified founder YARS2 mutation. Testing for pathogenic YARS2 variants should be considered in patients presenting with mitochondrial myopathy, characterized by exercise intolerance and muscle weakness even in the absence of sideroblastic anemia irrespective of ethnicity. Regular surveillance and early treatment for cardiomyopathy and respiratory muscle weakness is advocated because early treatment may mitigate the significant morbidity and mortality associated with this genetic disorder.


Subject(s)
Acidosis, Lactic/genetics , Anemia, Sideroblastic/genetics , Cardiomyopathies/genetics , Mitochondrial Myopathies/genetics , Muscle Weakness/genetics , Respiratory Insufficiency/genetics , Tyrosine-tRNA Ligase/genetics , Acidosis, Lactic/ethnology , Acidosis, Lactic/etiology , Adult , Aged , Anemia, Sideroblastic/ethnology , Anemia, Sideroblastic/etiology , Cardiomyopathies/ethnology , Cardiomyopathies/etiology , England/ethnology , Female , Humans , Male , Middle Aged , Mitochondrial Myopathies/complications , Mitochondrial Myopathies/ethnology , Muscle Weakness/ethnology , Muscle Weakness/etiology , Mutation , Prognosis , Respiratory Insufficiency/ethnology , Respiratory Insufficiency/etiology , Scotland/ethnology
14.
Curr Opin Hematol ; 24(3): 191-197, 2017 May.
Article in English | MEDLINE | ID: mdl-28072603

ABSTRACT

PURPOSE OF REVIEW: Myelodysplastic syndromes (MDS) are heterogeneous diseases of the hematopoietic stem cell in the elderly. Anemia is the main symptom that mostly correlates with dysplastic erythropoiesis in the bone marrow. We will review the recent advances in understanding the diverse mechanisms of dyserythropoiesis. RECENT FINDINGS: Dyserythropoiesis defined as 10% dysplastic erythroid cells in the bone marrow is found in more than 80% of early MDS. Immature erythroblasts accumulate at the expense of mature erythroblasts due to differentiation arrest and apoptosis. In early MDS with dyserythropoiesis, caspase-dependent cleavage of the erythroid transcription factor GATA-1 occurring in basophilic erythroblasts accounts for impairment of final maturation. Depending on initiating genetic alteration, specific mechanisms contribute to erythroid defect. In MDS with 5q deletion, the haploinsufficiency of ribosomal protein gene, RPS14, opposes the transition of immature to mature erythroblasts by inducing a p53-dependent ribosome stress, cell cycle arrest and apoptosis. Recent work identifies the activation of a p53-S100A8/9 innate immune pathway that both intrinsically and extrinsically contributes to defective erythropoiesis. In MDS with ring sideroblasts, a paradigm of dyserythropoiesis, a unique mutation in SF3B1 splicing factor gene induces a multiplicity of alterations at RNA level that deeply modifies the patterns of gene expression. SUMMARY: Insights in the pathophysiology of MDS with dyserythropoiesis may guide the choice of the appropriate therapy, for instance lenalidomide in MDS with del(5q). A better understanding of the mechanisms of dyserthropoiesis is required to treat anemia in non-del(5q) MDS, especially in case of resistance to first-line therapy by erythropoiesis-stimulating agents.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow/metabolism , Erythropoiesis , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/metabolism , Anemia, Macrocytic/genetics , Anemia, Macrocytic/metabolism , Anemia, Macrocytic/pathology , Anemia, Sideroblastic/etiology , Anemia, Sideroblastic/metabolism , Anemia, Sideroblastic/pathology , Animals , Bone Marrow/pathology , Bone Marrow Cells/pathology , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 5/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythroid Cells/pathology , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Humans , Immunity, Innate , Mitochondria/genetics , Mitochondria/immunology , Mitochondria/metabolism , Myelodysplastic Syndromes/diagnosis , RNA Splicing , Signal Transduction
15.
Leukemia ; 31(3): 720-727, 2017 03.
Article in English | MEDLINE | ID: mdl-27604819

ABSTRACT

Heterozygous somatic mutations affecting the spliceosome gene SF3B1 drive age-related clonal hematopoiesis, myelodysplastic syndromes (MDS) and other neoplasms. To study their role in such disorders, we generated knock-in mice with hematopoietic-specific expression of Sf3b1-K700E, the commonest type of SF3B1 mutation in MDS. Sf3b1K700E/+ animals had impaired erythropoiesis and progressive anemia without ringed sideroblasts, as well as reduced hematopoietic stem cell numbers and host-repopulating fitness. To understand the molecular basis of these observations, we analyzed global RNA splicing in Sf3b1K700E/+ hematopoietic cells. Aberrant splicing was associated with the usage of cryptic 3' splice and branchpoint sites, as described for human SF3B1 mutants. However, we found a little overlap between aberrantly spliced mRNAs in mouse versus human, suggesting that anemia may be a consequence of globally disrupted splicing. Furthermore, the murine orthologues of genes associated with ring sideroblasts in human MDS, including Abcb7 and Tmem14c, were not aberrantly spliced in Sf3b1K700E/+ mice. Our findings demonstrate that, despite significant differences in affected transcripts, there is overlap in the phenotypes associated with SF3B1-K700E between human and mouse. Future studies should focus on understanding the basis of these similarities and differences as a means of deciphering the consequences of spliceosome gene mutations in MDS.


Subject(s)
Anemia, Sideroblastic/etiology , Anemia, Sideroblastic/pathology , Hematopoiesis/genetics , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , RNA Splicing Factors/genetics , RNA Splicing , Anemia, Sideroblastic/mortality , Animals , Disease Models, Animal , Gene Targeting , Humans , Mice , Mice, Transgenic , Mutation , Phenotype , RNA Splicing Factors/metabolism
17.
Br J Haematol ; 174(6): 847-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27391606

ABSTRACT

Myeloid neoplasms with ring sideroblasts are currently categorized within the myelodysplastic syndromes (MDS) or myelodysplastic/myeloproliferative neoplasms (MDS/MPN) in the World Health Organization classification. Recent findings have identified that the presence of ring sideroblasts in these disorders has a unique molecular basis, i.e., the somatic mutation of SF3B1, a gene encoding a splicing factor. Mutations of SF3B1 occur in up to 90% of patients with refractory anaemia with unilineage dysplasia (RARS) and 70% of those with refractory cytopenia with multilineage dysplasia and ring sideroblasts or RARS associated with marked thrombocytosis. Experimental evidence has shown that mutant SF3B1 results in the abnormal splicing of several genes, primarily due to misrecognition of 3' splice sites. The resulting aberrant mRNAs undergo nonsense-mediated mRNA decay (NMD), resulting in haploinsufficiency of canonical transcripts and protein expression. In addition, it is also possible that NMD-insensitive aberrant transcripts are translated into proteins with altered function. Patients with MDS carrying the SF3B1 mutation show a homogeneous disease phenotype characterized by isolated erythroid dysplasia and mild dysplasia in granulocytic or megakaryocytic lineages, supporting the notion that the SF3B1 mutation identifies a distinct entity within MDS. The available evidence suggests that these findings may have relevant impact on the diagnosis, classification and management of patients with these neoplasms.


Subject(s)
Erythroid Precursor Cells/pathology , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/etiology , Anemia, Sideroblastic/etiology , Anemia, Sideroblastic/metabolism , Anemia, Sideroblastic/pathology , Bone Marrow/metabolism , Bone Marrow/pathology , Erythroid Precursor Cells/metabolism , Humans , Iron/metabolism , Mitochondria/metabolism , Mutation , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/etiology , Phenotype , Phosphoproteins/genetics , Prognosis , RNA Splicing , RNA Splicing Factors/genetics
18.
Rinsho Ketsueki ; 55(3): 345-9, 2014 03.
Article in Japanese | MEDLINE | ID: mdl-24681939

ABSTRACT

A 64-year-old man underwent kidney transplantation for progressive chronic renal failure which had developed 8 years after allogeneic bone marrow transplantation for acute myeloid leukemia. Because of post-operative complications, he had been placed on intravenous hyperalimentation. Three months after the transplantation, anemia rapidly progressed (hemoglobin, 7.9 g/dl). The proportion of reticulocytes was 0.2%, but white blood cell and platelet counts remained within normal ranges. Serum iron, vitamin B12, and folate levels were normal. Bone marrow examination showed the presence of ringed sideroblasts and cytoplasmic vacuoles in a fraction of erythroid cells. Megakaryocytes were adequate in number with normal morphology. Although the findings were consistent with refractory anemia with ringed sideroblasts according to the WHO classification, cytoplasmic vacuolations were also observed in myeloid cells, suggesting copper deficiency. Indeed, serum copper and ceruloplasmin levels were found to be low (33 µg/dl and 11 mg/dl, respectively), and oral copper supplementation at a daily dose of 1 mg was initiated. There was a prompt increase in reticulocytes, and the hemoglobin level was normalized within one month, in response to this regimen. In progressive anemia cases with ringed sideroblasts in the bone marrow, copper deficiency should be considered in the differential diagnosis.


Subject(s)
Anemia, Sideroblastic/etiology , Anemia, Sideroblastic/pathology , Bone Marrow/pathology , Copper/deficiency , Administration, Oral , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/drug therapy , Biomarkers/blood , Bone Marrow Transplantation , Ceruloplasmin , Copper/administration & dosage , Copper/blood , Diagnosis, Differential , Humans , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/therapy , Kidney Transplantation , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Myelodysplastic Syndromes
20.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(4): 1088-90, 2013 Aug.
Article in Chinese | MEDLINE | ID: mdl-23998618

ABSTRACT

Myelodysplastic syndromes (MDS) are heterogeneous clonal hematopoietic stem cell disorders with different mechanisms and diverse prognosis. The excess of ring sideroblasts (RS) is an important presentation MDS, but the mechanisms of RS appearance are obscure and the treatment of MDS-RS is intractable. Splicing factors play a very important role in the maturation process of eucaryon mRNA, recent studies indicate that there is a significant causal relationship between splicing factor 3B subunit 1 (SF3B1) mutation and the presence of ring sideroblasts. Lucubrating the downstream molecular of the mutated SF3B1 can facilitate exploring the mechanisms and new therapeutic strategies of MDS-RS.


Subject(s)
Anemia, Sideroblastic/genetics , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Anemia, Sideroblastic/etiology , Animals , Humans , Mutation , Myelodysplastic Syndromes/complications , RNA Splicing Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...