Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 227
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
EMBO Rep ; 25(5): 2418-2440, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38605277

RESUMO

Microcephaly is a common feature in inherited bone marrow failure syndromes, prompting investigations into shared pathways between neurogenesis and hematopoiesis. To understand this association, we studied the role of the microcephaly gene Mcph1 in hematological development. Our research revealed that Mcph1-knockout mice exhibited congenital macrocytic anemia due to impaired terminal erythroid differentiation during fetal development. Anemia's cause is a failure to complete cell division, evident from tetraploid erythroid progenitors with DNA content exceeding 4n. Gene expression profiling demonstrated activation of the p53 pathway in Mcph1-deficient erythroid precursors, leading to overexpression of Cdkn1a/p21, a major mediator of p53-dependent cell cycle arrest. Surprisingly, fetal brain analysis revealed hypertrophied binucleated neuroprogenitors overexpressing p21 in Mcph1-knockout mice, indicating a shared pathophysiological mechanism underlying both erythroid and neurological defects. However, inactivating p53 in Mcph1-/- mice failed to reverse anemia and microcephaly, suggesting that p53 activation in Mcph1-deficient cells resulted from their proliferation defect rather than causing it. These findings shed new light on Mcph1's function in fetal hematopoietic development, emphasizing the impact of disrupted cell division on neurogenesis and erythropoiesis - a common limiting pathway.


Assuntos
Proteínas de Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21 , Eritropoese , Camundongos Knockout , Microcefalia , Proteína Supressora de Tumor p53 , Animais , Camundongos , Anemia Macrocítica/genética , Anemia Macrocítica/patologia , Anemia Macrocítica/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoese/genética , Microcefalia/genética , Microcefalia/patologia , Mutação , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
2.
Mol Cell ; 67(1): 55-70.e4, 2017 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-28673543

RESUMO

Ribosomal protein (RP) expression in higher eukaryotes is regulated translationally through the 5'TOP sequence. This mechanism evolved to more rapidly produce RPs on demand in different tissues. Here we show that 40S ribosomes, in a complex with the mRNA binding protein LARP1, selectively stabilize 5'TOP mRNAs, with disruption of this complex leading to induction of the impaired ribosome biogenesis checkpoint (IRBC) and p53 stabilization. The importance of this mechanism is underscored in 5q− syndrome, a macrocytic anemia caused by a large monoallelic deletion, which we found to also encompass the LARP1 gene. Critically, depletion of LARP1 alone in human adult CD34+ bone marrow precursor cells leads to a reduction in 5'TOP mRNAs and the induction of p53. These studies identify a 40S ribosome function independent of those in translation that, with LARP1, mediates the autogenous control of 5'TOP mRNA stability, whose disruption is implicated in the pathophysiology of 5q− syndrome.


Assuntos
Autoantígenos/metabolismo , Biossíntese de Proteínas , Sequência de Oligopirimidina na Região 5' Terminal do RNA , Estabilidade de RNA , RNA Mensageiro/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Anemia Macrocítica/genética , Anemia Macrocítica/metabolismo , Autoantígenos/genética , Células da Medula Óssea/metabolismo , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Cromossomos Humanos Par 5/metabolismo , Células HCT116 , Humanos , Complexos Multiproteicos , Ligação Proteica , Interferência de RNA , RNA Mensageiro/genética , Ribonucleoproteínas/genética , Proteínas Ribossômicas/genética , Ribossomos/genética , Fatores de Tempo , Transfecção , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Antígeno SS-B
3.
Am J Hematol ; 97(11): 1495-1496, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35441393

RESUMO

We diagnosed a 13-month-old girl with severe neurological deficits and hyporegenerative macrocytic anemiawith Brown-Vialetto-Van Laere syndrome type 2 (BVVL 2), a rare disorder of the riboflavin transporter, caused by variants in the SLC52A2 gene. Bone marrow aspiration revealed hypoplastic erythropoiesis and vacuolization of myelocytes, proerythroblasts, and micromegakaryocytes. We suggest BVVL 2 as an important differential diagnosis in hyporegenerative macrocytic anemia as rapid diagnosis and initiation of therapy are crucial for the remedy of hematological and neurological impairment.


Assuntos
Anemia Macrocítica , Paralisia Bulbar Progressiva , Perda Auditiva Neurossensorial , Anemia Macrocítica/diagnóstico , Anemia Macrocítica/genética , Paralisia Bulbar Progressiva/etiologia , Paralisia Bulbar Progressiva/genética , Feminino , Perda Auditiva Neurossensorial/genética , Humanos , Lactente , Riboflavina
4.
Blood Cells Mol Dis ; 87: 102522, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33260083

RESUMO

Stathmin 1 (STMN1) is a cytosolic phosphoprotein that was discovered as a result of its high level of expression in leukemic cells. It plays an important role in the regulation of mitosis by promoting depolymerization of the microtubules that make up the mitotic spindle and, aging has been shown to impair STMN1 levels and change microtubule stability. We have previously demonstrated that a high level of STMN1 expression during early megakaryopoiesis is necessary for proliferation of megakaryocyte progenitors and that down-regulation of STMN1 expression during late megakaryopoiesis is important for megakaryocyte maturation and platelet production. In this report, we examined the effects of STMN1 deficiency on erythroid and megakaryocytic lineages in the mouse. Our studies show that STMN1 deficiency results in mild thrombocytopenia in young animals which converts into profound thrombocythemia as the mice age. STMN1 deficiency also lead to macrocytic changes in both erythrocytes and megakaryocytes that persisted throughout the life of STMN1 knock-out mice. Furthermore, STMN1 knock-out mice displayed a lower number of erythroid and megakaryocytic progenitor cells and had delayed recovery of their blood counts after chemotherapy. These studies show an important role for STMN1 in normal erythro-megakaryopoietic development and suggests potential implications for disorders affecting these hematopoietic lineages.


Assuntos
Anemia Macrocítica/genética , Células Precursoras Eritroides/patologia , Megacariócitos/patologia , Estatmina/genética , Trombocitose/genética , Anemia Macrocítica/patologia , Animais , Plaquetas/patologia , Eritropoese , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Trombocitose/patologia
5.
Am J Med Genet A ; 185(12): 3844-3850, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34322994

RESUMO

Microdeletions at 5q11.2 are rare. Subjects show a phenotypic spectrum that overlaps CHARGE syndrome and 22q11.2 deletion syndrome. A growing number of subjects present with learning difficulty and/or intellectual disability, immune deficiency, congenital heart malformation, and dysmorphism. DHX29 and IL6ST have been proposed as candidate genes for the development of the major clinical manifestations. We present a new case and narrow down the shortest region of overlap to evaluate possible candidate genes. Our case does not present developmental delay or immune deficiency indicating a reduced penetrance for some of the main clinical manifestations. The shortest region of overlap between subjects with deletions at 5q11.2 is approximately 450 kb (position 54.3-54.7 Mb). The narrowed region comprises 10 protein coding genes, including DHX29. DHX29 is a strong candidate gene for the main features of 5q11.2-microdeletion syndrome; however, our findings suggest a joined impact of several genes as the cause of the syndrome.


Assuntos
Anormalidades Múltiplas/genética , Anemia Macrocítica/genética , Cardiopatias Congênitas/genética , Deficiência Intelectual/genética , RNA Helicases/genética , Anormalidades Múltiplas/fisiopatologia , Anemia Macrocítica/fisiopatologia , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Hibridização Genômica Comparativa , Receptor gp130 de Citocina/genética , Deficiências do Desenvolvimento/genética , Deficiências do Desenvolvimento/fisiopatologia , Fácies , Cardiopatias Congênitas/fisiopatologia , Humanos , Lactente , Recém-Nascido , Deficiência Intelectual/fisiopatologia , Deficiências da Aprendizagem/genética , Deficiências da Aprendizagem/fisiopatologia , Masculino , Fenótipo
6.
Am J Med Genet A ; 182(11): 2781-2787, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32909658

RESUMO

Riboflavin transporter deficiency (RTD) (MIM #614707) is a neurogenetic disorder with its most common manifestations including sensorineural hearing loss, peripheral neuropathy, respiratory insufficiency, and bulbar palsy. Here, we present a 2-year-old boy whose initial presentation was severe macrocytic anemia necessitating multiple blood transfusions and intermittent neutropenia; he subsequently developed ataxia and dysarthria. Trio-exome sequencing detected compound heterozygous variants in SLC52A2 that were classified as pathogenic and a variant of uncertain significance. Bone marrow evaluation demonstrated megaloblastic changes. Notably, his anemia and neutropenia resolved after treatment with oral riboflavin, thus expanding the clinical phenotype of this disorder. We reiterate the importance of starting riboflavin supplementation in a young child who presents with macrocytic anemia and neurological features while awaiting biochemical and genetic work up. We detected multiple biochemical abnormalities with the help of untargeted metabolomics analysis associated with abnormal flavin adenine nucleotide function which normalized after treatment, emphasizing the reversible pathomechanisms involved in this disorder. The utility of untargeted metabolomics analysis to monitor the effects of riboflavin supplementation in RTD has not been previously reported.


Assuntos
Anemia Macrocítica/patologia , Paralisia Bulbar Progressiva/patologia , Perda Auditiva Neurossensorial/patologia , Metaboloma , Deficiência de Riboflavina/patologia , Riboflavina/metabolismo , Adulto , Anemia Macrocítica/genética , Anemia Macrocítica/metabolismo , Paralisia Bulbar Progressiva/genética , Paralisia Bulbar Progressiva/metabolismo , Feminino , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/metabolismo , Humanos , Lactente , Masculino , Mutação , Receptores Acoplados a Proteínas G/genética , Deficiência de Riboflavina/genética , Deficiência de Riboflavina/metabolismo
7.
Rev Med Chil ; 148(9): 1357-1361, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33399713

RESUMO

Myelodysplastic syndrome with deletion of chromosome 5q (5q-syndrome) has a favorable prognosis and a low risk of transformation to acute myeloid leukemia, when treated with lenalidomide. Azacitidine leads to complete remission even as second-line therapy and in patients with clonal evolution. We report a 70 years old female without previous exposure to myelotoxic drugs, presenting with three weeks with fatigue and dyspnea. She had anemia with normal white blood cell and platelet count. Bone marrow biopsy showed 50% cellularity and the karyotype analysis revealed a (5) (q33q34) deletion in 22% of the metaphases. A diagnosis of 5q-syndrome with low risk calculated using the Revised International Prognostic Scoring System (IPSS-R), was made. Since lenalidomide was not affordable, thalidomide 100 mg/day was initiated, achieving transfusion independence for three years. Afterwards, she developed pancytopenia and a bone marrow biopsy showed erythroid and megakaryocyte dysplasia with a complex karyotype, which worsened prognosis (IPSS-R of five points). Therefore, azacitidine (by donation) was administered. She achieved complete remission with a normal karyotype and completed 12 cycles of treatment. Thereafter, she relapsed and received only supportive care for a year. She suffered an ischemic stroke and died two weeks later.


Assuntos
Anemia Macrocítica , Inibidores da Angiogênese , Deleção Cromossômica , Síndromes Mielodisplásicas , Talidomida , Idoso , Anemia Macrocítica/tratamento farmacológico , Anemia Macrocítica/genética , Inibidores da Angiogênese/uso terapêutico , Cromossomos Humanos Par 5/genética , Feminino , Humanos , Lenalidomida , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/genética , Talidomida/uso terapêutico , Resultado do Tratamento
8.
Pediatr Hematol Oncol ; 36(4): 236-243, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31361176

RESUMO

Here we report a case of refractory macrocytic anemia with a spliceosomal point mutation involving the ZRSR2 gene in a child with Down syndrome (DS). Such mutations have been shown to cause refractory macrocytic anemia and myelodysplastic syndrome (MDS) in elderly individuals. We report the hematological indices of a child with DS and a ZRSR2 spliceosomal mutation. DS is known to produce macrocytic anemia but does not lead to transfusion dependence. In this case, the ZRSR2 mutation was the likely implicating factor for severe transfusion-dependent anemia in a child with DS. The clinical implication of a ZRSR2 mutation in a child with DS has not been previously described and warrants close surveillance to detect potential insidious transformation to MDS.


Assuntos
Anemia Macrocítica/genética , Síndrome de Down/genética , Mutação Puntual , Ribonucleoproteínas/genética , Anemia Macrocítica/sangue , Anemia Macrocítica/terapia , Criança , Síndrome de Down/sangue , Síndrome de Down/terapia , Humanos , Masculino , Ribonucleoproteínas/metabolismo
9.
Blood ; 127(6): 749-60, 2016 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-26626993

RESUMO

Non-del(5q) transfusion-dependent low/intermediate-1 myelodysplastic syndrome (MDS) patients achieve an erythroid response with lenalidomide in 25% of cases. Addition of an erythropoiesis-stimulating agent could improve response rate. The impact of recurrent somatic mutations identified in the diseased clone in response to lenalidomide and the drug's effects on clonal evolution remain unknown. We investigated recurrent mutations by next-generation sequencing in 94 non-del(5q) MDS patients randomized in the GFM-Len-Epo-08 clinical trial to lenalidomide or lenalidomide plus epoetin ß. Clonal evolution was analyzed after 4 cycles of treatment in 42 cases and reanalyzed at later time points in 18 cases. The fate of clonal architecture of single CD34(+)CD38(-) hematopoietic stem cells was also determined in 5 cases. Mutation frequency was >10%: SF3B1 (74.5%), TET2 (45.7%), DNMT3A (20.2%), and ASXL1 (19.1%). Analysis of variant allele frequencies indicated a decrease of major mutations in 15 of 20 responders compared with 10 of 22 nonresponders after 4 cycles. The decrease in the variant allele frequency of major mutations was more significant in responders than in nonresponders (P < .001). Genotyping of single CD34(+)CD38(-) cell-derived colonies showed that the decrease in the size of dominant subclones could be associated with the rise of founding clones or of hematopoietic stem cells devoid of recurrent mutations. These effects remained transient, and disease escape was associated with the re-emergence of the dominant subclones. In conclusion, we show that, although the drug initially modulates the distribution of subclones, loss of treatment efficacy coincides with the re-expansion of the dominant subclone. This trial was registered at www.clinicaltrials.gov as #NCT01718379.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Evolução Clonal/efeitos dos fármacos , Síndromes Mielodisplásicas/tratamento farmacológico , Talidomida/análogos & derivados , Idoso , Anemia Macrocítica/tratamento farmacológico , Anemia Macrocítica/genética , Anemia Macrocítica/patologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Evolução Clonal/genética , Células Clonais/efeitos dos fármacos , Células Clonais/metabolismo , Células Clonais/patologia , Análise Mutacional de DNA , Eritropoetina/administração & dosagem , Feminino , Humanos , Lenalidomida , Masculino , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Proteínas Recombinantes/administração & dosagem , Talidomida/administração & dosagem , Talidomida/farmacologia , Resultado do Tratamento
10.
Int J Mol Sci ; 19(10)2018 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-30347879

RESUMO

Myelodysplastic syndrome (MDS) can easily transform into acute myeloid leukemia (AML), a process which is often associated with clonal evolution and development of complex karyotypes. Deletion of 5q (del(5q)) is the most frequent aberration in complex karyotypes. This prompted us to analyze clonal evolution in MDS patients with del(5q). There were 1684 patients with low and intermediate-risk MDS and del(5q) with or without one additional cytogenetic abnormality, who were investigated cytogenetically in our department, involving standard karyotyping, fluorescence in situ hybridization (FISH) and multicolor FISH. We identified 134 patients (8%) with aspects of clonal evolution. There are two main routes of cytogenetic clonal evolution: a stepwise accumulation of cytogenetic events over time and a catastrophic event, which we defined as the occurrence of two or more aberrations present at the same time, leading to a sudden development of highly complex clones. Of the 134 patients, 61% underwent a stepwise accumulation of events whereas 39% displayed a catastrophic event. Patients with isolated del(5q) showed significantly more often a stepwise accumulation of events rather than a catastrophic event. The most frequent aberrations in the group of stepwise accumulation were trisomy 8 and trisomy 21 which were significantly more frequent in this group compared to the catastrophic event group. In the group with catastrophic events, del(7q)/-7 and del(17p)/-17 were the most common aberrations. A loss of 17p, containing the tumor suppressor gene TP53, was found significantly more frequent in this group compared to the group of stepwise accumulation. This leads to the assumption that the loss of TP53 is the driving force in patients with del(5q) who undergo a sudden catastrophic event and evolve into complex karyotypes.


Assuntos
Anemia Macrocítica/genética , Evolução Clonal , Cariótipo , Células Cultivadas , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Humanos
11.
Curr Opin Hematol ; 24(3): 191-197, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28072603

RESUMO

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.


Assuntos
Células da Medula Óssea/metabolismo , Medula Óssea/metabolismo , Eritropoese , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/metabolismo , Anemia Macrocítica/genética , Anemia Macrocítica/metabolismo , Anemia Macrocítica/patologia , Anemia Sideroblástica/etiologia , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patologia , Animais , Medula Óssea/patologia , Células da Medula Óssea/patologia , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Cromossomos Humanos Par 5/metabolismo , Células Eritroides/citologia , Células Eritroides/metabolismo , Células Eritroides/patologia , Eritropoese/genética , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Mitocôndrias/genética , Mitocôndrias/imunologia , Mitocôndrias/metabolismo , Síndromes Mielodisplásicas/diagnóstico , Splicing de RNA , Transdução de Sinais
13.
Am J Hum Genet ; 93(5): 906-14, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-24119684

RESUMO

We used exome sequencing to identify mutations in sideroflexin 4 (SFXN4) in two children with mitochondrial disease (the more severe case also presented with macrocytic anemia). SFXN4 is an uncharacterized mitochondrial protein that localizes to the mitochondrial inner membrane. sfxn4 knockdown in zebrafish recapitulated the mitochondrial respiratory defect observed in both individuals and the macrocytic anemia with megaloblastic features of the more severe case. In vitro and in vivo complementation studies with fibroblasts from the affected individuals and zebrafish demonstrated the requirement of SFXN4 for mitochondrial respiratory homeostasis and erythropoiesis. Our findings establish mutations in SFXN4 as a cause of mitochondriopathy and macrocytic anemia.


Assuntos
Anemia Macrocítica/genética , Proteínas de Membrana/genética , Doenças Mitocondriais/genética , Adolescente , Animais , Criança , Eritropoese/genética , Exoma , Feminino , Técnicas de Silenciamento de Genes , Humanos , Proteínas Mitocondriais/genética , Mutação , Peixe-Zebra/genética
14.
Am J Physiol Heart Circ Physiol ; 311(6): H1392-H1408, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27694217

RESUMO

The HDL receptor SR-BI mediates the transfer of cholesteryl esters from HDL to cells and controls HDL abundance and structure. Depending on the genetic background, loss of SR-BI causes hypercholesterolemia, anemia, reticulocytosis, splenomegaly, thrombocytopenia, female infertility, and fatal coronary heart disease (CHD). The carboxy terminus of SR-BI (505QEAKL509) must bind to the cytoplasmic adaptor PDZK1 for normal hepatic-but not steroidogenic cell-expression of SR-BI protein. To determine whether SR-BI's carboxy terminus is also required for normal protein levels in steroidogenic cells, we introduced into SR-BI's gene a 507Ala/STOP mutation that produces a truncated receptor (SR-BIΔCT). As expected, the dramatic reduction of hepatic receptor protein in SR-BIΔCT mice was similar to that in PDZK1 knockout (KO) mice. Unlike SR-BI KO females, SR-BIΔCT females were fertile. The severity of SR-BIΔCT mice's hypercholesterolemia was intermediate between those of SR-BI KO and PDZK1 KO mice. Substantially reduced levels of the receptor in adrenal cortical cells, ovarian cells, and testicular Leydig cells in SR-BIΔCT mice suggested that steroidogenic cells have an adaptor(s) functionally analogous to hepatic PDZK1. When SR-BIΔCT mice were crossed with apolipoprotein E KO mice (SR-BIΔCT/apoE KO), pathologies including hypercholesterolemia, macrocytic anemia, hepatic and splenic extramedullary hematopoiesis, massive splenomegaly, reticulocytosis, thrombocytopenia, and rapid-onset and fatal occlusive coronary arterial atherosclerosis and CHD (median age of death: 9 wk) were observed. These results provide new insights into the control of SR-BI in steroidogenic cells and establish SR-BIΔCT/apoE KO mice as a new animal model for the study of CHD.


Assuntos
Córtex Suprarrenal/metabolismo , Hipercolesterolemia/genética , Células Intersticiais do Testículo/metabolismo , Fígado/metabolismo , Ovário/metabolismo , Receptores Depuradores Classe B/genética , Anemia Macrocítica/genética , Animais , Apolipoproteínas E/genética , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/mortalidade , Doença das Coronárias/genética , Doença das Coronárias/mortalidade , Oclusão Coronária/genética , Oclusão Coronária/mortalidade , Feminino , Técnicas de Introdução de Genes , Hematopoese Extramedular/genética , Immunoblotting , Lipoproteínas HDL/genética , Masculino , Camundongos , Mutação , Reação em Cadeia da Polimerase , Receptores de Lipoproteínas/genética , Reticulocitose/genética , Esplenomegalia/genética , Trombocitopenia/genética , Transcriptoma
15.
Cytogenet Genome Res ; 150(1): 35-39, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27838684

RESUMO

Constitutional trisomy 8 mosaicism (CT8M) is a rare chromosomal abnormality. The phenotype varies from normal features to severe malformations. CT8M increases the risk of developing leukemia and myelodysplastic syndrome. As CT8M is very rare, its diagnosis can easily be overlooked, especially in cases with mild phenotypes. Here, we report the diagnostic process of a 40-year-old female patient with CT8M and discuss the importance of follow-up in monitoring for hematological malignancies.


Assuntos
Anemia Macrocítica/genética , Trissomia/diagnóstico , Trissomia/genética , Dissomia Uniparental/diagnóstico , Dissomia Uniparental/genética , Adulto , Anemia Macrocítica/complicações , Cromossomos Humanos Par 8/genética , Feminino , Predisposição Genética para Doença/genética , Neoplasias Hematológicas/etiologia , Neoplasias Hematológicas/genética , Humanos , Mosaicismo , Doenças Raras/diagnóstico , Doenças Raras/genética
16.
Blood ; 123(2): 228-38, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24264229

RESUMO

Therapy-related myeloid neoplasms (t-MN) are a late complication of the successful use of cytotoxic therapy for patients with cancer. A heterozygous deletions of the long arm of chromosome 5 [del(5q)], observed in 40% of patients, is associated with prior exposure to alkylating agents, and a high frequency of TP53 loss or mutation. In previous studies, we demonstrated that haploinsufficiency of 2 del(5q) genes, Egr1, and Apc, individually play a role in the pathogenesis of hematologic disease in mice. We now show that loss of one copy of Egr1 or Tp53 in an Apc haploinsufficient background (Apc (del/+)) accelerated the development of a macrocytic anemia with monocytosis, early features of t-MN. The development of anemia was significantly accelerated by treatment of mice with the alkylating agent, N-ethyl-N-nitrosourea (ENU), regardless of the levels of expression of Egr1 and Tp53. Transplantation of either wild type; Egr1(+/-); Tp53(+/-); Apc(del/+); or Egr1(+/-), Apc(del/+) bone marrow cells into lethally irradiated Apc(del/+) recipients resulted in rapid development of anemia that was further accelerated by administration of ENU to recipients, demonstrating that the Apc(del/+)-induced anemia was cell extrinsic and potentiated by ENU mutagenesis. These data emphasize the synergistic role of cell intrinsic and cell extrinsic (microenvironment) factors in the pathogenesis of t-MN, and raise awareness of the deleterious effects of cytotoxic therapy on the stromal microenvironment.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Deleção Cromossômica , Cromossomos Humanos Par 5 , Proteína 1 de Resposta de Crescimento Precoce/genética , Haploinsuficiência , Proteína Supressora de Tumor p53/genética , Alelos , Anemia Macrocítica/induzido quimicamente , Anemia Macrocítica/genética , Anemia Macrocítica/mortalidade , Animais , Apoptose/genética , Medula Óssea/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Eritroblastos/citologia , Eritroblastos/metabolismo , Eritropoese/genética , Etilnitrosoureia/efeitos adversos , Genes Letais , Genótipo , Heterozigoto , Humanos , Camundongos , Camundongos Transgênicos , Baço/metabolismo , Baço/patologia
17.
Ann Hematol ; 95(11): 1805-10, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27510179

RESUMO

Lenalidomide (LEN) leads to erythroid improvement in the majority of patients with myelodysplastic syndrome and isolated deletion of the long arm of chromosome 5 (MDS-del(5q)). This effect is believed to be exerted via its immunomodulatory properties, although the precise nature is still incompletely understood. We prospectively performed immune profiling in the bone marrow and blood of MDS-del(5q) patients undergoing LEN therapy for a median of 6 cycles. Therapy with LEN led to a significant increase in the median absolute lymphocyte count (1.3-fold, p = 0.013) without changes in the distribution of the T helper cells within the entire compartment. In parallel, the frequency of Treg increased significantly during treatment both in the peripheral blood (5.0 vs. 9.6 %, p = 0.001) and bone marrow (3.4 vs. 8.1 %, p = 0.001). Surprisingly, LEN treatment led to a decrease in TGFbeta levels, both in the peripheral blood (4.9 vs. 2.3 ng/ml, p = 0.039) and bone marrow (4.5 vs. 0.8 ng/ml, p = 0.023). These changes were not associated with an increase in pro-inflammatory Th17 cells. Taken together, our results demonstrate that LEN induces a shift in lymphocytic populations towards immunosuppression in MDS-del(5q) patients.


Assuntos
Anemia Macrocítica/tratamento farmacológico , Fatores Imunológicos/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Talidomida/análogos & derivados , Idoso , Idoso de 80 Anos ou mais , Anemia Macrocítica/genética , Anemia Macrocítica/imunologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Cromossomos Humanos Par 5/imunologia , Feminino , Humanos , Fatores Imunológicos/uso terapêutico , Lenalidomida , Masculino , Pessoa de Meia-Idade , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Talidomida/farmacologia , Talidomida/uso terapêutico , Fator de Crescimento Transformador beta/sangue
19.
Int J Gynecol Pathol ; 35(6): 561-565, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27015438

RESUMO

Microcystic stromal tumor (MST) is a rare tumor of presumed sex-cord stromal differentiation. We present a case of MST arising within a patient with constitutional 5q deletion syndrome, whose deletion encompassed the APC gene. Genomic analysis of the MST revealed a point mutation in the remaining APC allele, predicted to result in abnormal splicing of Exon 7. Subsequent clinical investigation revealed multiple gastrointestinal polyps qualifying for a diagnosis of familial adenomatous polyposis. This case emphasizes the importance of an aberrant Wnt/ß-catenin pathway in the development of MST and adds credence to the inclusion of MST as a rare phenotype of familial adenomatous polyposis. In a search for additional genetic aberrations which may contribute to the development of this rare tumor, genomic analysis revealed a frameshift mutation in FANCD2, a protein which plays a key role in DNA repair. This protein is expressed in human ovarian stromal cells and FANCD2-knockout mice are known to develop sex cord-stromal tumors, factors which further support a possible role of aberrant FANCD2 in the development of MST.


Assuntos
Polipose Adenomatosa do Colo/complicações , Anemia Macrocítica/complicações , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Neoplasias Ovarianas/genética , Tumores do Estroma Gonadal e dos Cordões Sexuais/genética , Polipose Adenomatosa do Colo/genética , Anemia Macrocítica/genética , Deleção Cromossômica , Cromossomos Humanos Par 5/genética , Feminino , Mutação da Fase de Leitura , Genes APC , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias Ovarianas/patologia , Tumores do Estroma Gonadal e dos Cordões Sexuais/patologia , Adulto Jovem
20.
Adv Exp Med Biol ; 907: 189-213, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27256387

RESUMO

RNA editing is a cellular process used to expand and diversify the RNA transcripts produced from a generally immutable genome. In animals, the most prevalent type of RNA editing is adenosine (A) to inosine (I) deamination catalyzed by the ADAR family. Throughout development, A-to-I editing levels increase while ADAR expression is constant, suggesting cellular mechanisms to regulate A-to-I editing exist. Furthermore, in several disease states, ADAR expression levels are similar to the normal state, but A-to-I editing levels are altered. Therefore, understanding how these enzymes are regulated in normal tissues and misregulated in disease states is of profound importance. This chapter will both discuss how to identify A-to-I editing sites across the transcriptome and explore the mechanisms that regulate ADAR editing activity, with particular focus on the diverse types of RNA-binding proteins implicated in regulating A-to-I editing in vivo.


Assuntos
Adenosina Desaminase/fisiologia , Adenosina/metabolismo , Inosina/metabolismo , Edição de RNA , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/fisiologia , Anemia Macrocítica/enzimologia , Anemia Macrocítica/genética , Animais , Pareamento de Bases , Proteínas de Caenorhabditis elegans/fisiologia , Deleção Cromossômica , Cromossomos Humanos Par 5/enzimologia , Cromossomos Humanos Par 5/genética , Proteínas de Drosophila/fisiologia , Humanos , Camundongos , Doenças do Sistema Nervoso/enzimologia , Doenças do Sistema Nervoso/genética , Conformação de Ácido Nucleico , Splicing de RNA , Spliceossomos/fisiologia , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA