Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
2.
Medicina (Kaunas) ; 56(8)2020 Jul 27.
Article in English | MEDLINE | ID: mdl-32727068

ABSTRACT

Myelodysplastic syndrome (MDS) is a clonal disease characterized by multilineage dysplasia, peripheral blood cytopenias, and a high risk of transformation to acute myeloid leukemia. In theory, from clonal hematopoiesis of indeterminate potential to hematologic malignancies, there is a complex interplay between genetic and epigenetic factors, including miRNA. In practice, karyotype analysis assigns patients to different prognostic groups, and mutations are often associated with a particular disease phenotype. Among myeloproliferative disorders, secondary MDS is a group of special entities with a typical spectrum of genetic mutations and cytogenetic rearrangements resembling those in de novo MDS. This overview analyzes the present prognostic systems of MDS and the most recent efforts in the search for genetic and epigenetic markers for the diagnosis and prognosis of MDS.


Subject(s)
Biomarkers/analysis , Myelodysplastic Syndromes/diagnosis , Prognosis , Core Binding Factor Alpha 2 Subunit/analysis , DNA (Cytosine-5-)-Methyltransferases/analysis , DNA Methyltransferase 3A , DNA-Binding Proteins/analysis , Dioxygenases , Humans , Mutation/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/physiopathology , Phosphoproteins/analysis , Proto-Oncogene Proteins/analysis , RNA Splicing Factors/analysis , Repressor Proteins/analysis , Serine-Arginine Splicing Factors/analysis
3.
Front Immunol ; 11: 368, 2020.
Article in English | MEDLINE | ID: mdl-32265900

ABSTRACT

Introduction: Leprosy is an infectious disease caused by Mycobacterium leprae, a debilitating disease that affects the skin and peripheral nerves. It is possible that tissue changes during infection with leprosy are related to alterations in the activity of the Notch signaling pathway, an innate signaling pathway in the physiology of the skin and peripheral nerves. Methods: This is a descriptive observational study. Thirty skin biopsies from leprosy patients and 15 from individuals with no history of this disease were evaluated. In these samples, gene expressions of cellular components associated with the Notch signaling pathway, Hes-1, Hey-1, Runx-1 Jagged-1, Notch-1, and Numb, were evaluated using q-PCR, and protein expression was evaluated using immunohistochemistry of Runx-1 and Hes-1. Results: Changes were observed in the transcription of Notch signaling pathway components; Hes-1 was downregulated and Runx-1 upregulated in the skin of infected patients. These results were confirmed by immunohistochemistry, where reduction of Hes-1 expression was found in the epidermis, eccrine glands, and hair follicles. Increased expression of Runx-1 was found in inflammatory cells in the dermis of infected patients; however, it is not related to tissue changes. With these results, a multivariate analysis was performed to determine the causes of transcription factor Hes-1 reduction. It was concluded that tissue inflammation was the main cause. Conclusions: The tissue changes found in the skin of infected patients could be associated with a reduction in the expression of Hes-1, a situation that would promote the survival and proliferation of M. leprae in this tissue.


Subject(s)
Leprosy/metabolism , Nerve Fibers/pathology , Receptors, Notch/physiology , Skin/metabolism , Adult , Aged , Core Binding Factor Alpha 2 Subunit/analysis , Cyclin D1/analysis , Female , Humans , Immunohistochemistry , Leprosy/pathology , Male , Middle Aged , Nerve Fibers/chemistry , Signal Transduction/physiology , Skin/pathology , Transcription Factor HES-1/analysis
5.
Blood ; 134(19): 1608-1618, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31554635

ABSTRACT

We performed serial measurable residual disease (MRD) monitoring in bone marrow (BM) and peripheral blood (PB) samples of 155 intensively treated patients with RUNX1-RUNX1T1+ AML, using a qRT-PC-based assay with a sensitivity of up to 10-6. We assessed both reduction of RUNX1-RUNX1T1 transcript levels (TLs) and achievement of MRD negativity (MRD-) for impact on prognosis. Achievement of MR2.5 (>2.5 log reduction) after treatment cycle 1 and achievement of MR3.0 after treatment cycle 2 were significantly associated with a reduced risk of relapse (P = .034 and P = .028, respectively). After completion of therapy, achievement of MRD- in both BM and PB was an independent, favorable prognostic factor in cumulative incidence of relapse (4-year cumulative incidence relapse: BM, 17% vs 36%, P = .021; PB, 23% vs 55%, P = .001) and overall survival (4-year overall survival rate BM, 93% vs 70%, P = .007; PB, 87% vs 47%, P < .0001). Finally, during follow-up, serial qRT-PCR analyses allowed prediction of relapse in 77% of patients exceeding a cutoff value of 150 RUNX1-RUNX1T1 TLs in BM, and in 84% of patients exceeding a value of 50 RUNX1-RUNX1T1 TLs in PB. The KIT mutation was a significant factor predicting a lower CR rate and inferior outcome, but its prognostic impact was outweighed by RUNX1-RUNX1T1 TLs during treatment. Virtually all relapses occurred within 1 year after the end of treatment, with a very short latency from molecular to morphologic relapse, necessitating MRD assessment at short intervals during this time period. Based on our data, we propose a refined practical guideline for MRD assessment in RUNX1-RUNX1T1+ AML.


Subject(s)
Leukemia, Myeloid, Acute/diagnosis , Neoplasm, Residual/diagnosis , Oncogene Proteins, Fusion/analysis , Adolescent , Adult , Aged , Core Binding Factor Alpha 2 Subunit/analysis , Core Binding Factor Alpha 2 Subunit/genetics , Female , Humans , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Neoplasm, Residual/genetics , Oncogene Proteins, Fusion/genetics , Prognosis , RUNX1 Translocation Partner 1 Protein/analysis , RUNX1 Translocation Partner 1 Protein/genetics , Real-Time Polymerase Chain Reaction/methods , Translocation, Genetic , Young Adult
6.
BMJ Case Rep ; 20172017 Oct 09.
Article in English | MEDLINE | ID: mdl-28993357

ABSTRACT

We describe a unique presentation of acute myeloid leukaemia (AML) with myeloid sarcoma (MS), manifested as proptosis with multiple cranial nerve palsies in a 9-year-old boy. MRI of the brain revealed multiple enhancing lesions and bilateral mastoiditis, in addition to sagittal sinus thrombosis. Peripheral blood smear demonstrated blasts showing Auer rods. Bone marrow examination confirmed the diagnosis of AML. PCR was positive for RUNX1-RUNX1T1. Neurological deficits improved with induction chemotherapy for AML. Extramedullary MS can present simultaneously with or antedate AML. Common genetic aberrations include t(8;21) and inv(16). Therapy is akin to AML. An effect of MS on survival outcomes is variable.


Subject(s)
Core Binding Factor Alpha 2 Subunit/analysis , Cranial Nerve Diseases/genetics , Exophthalmos/genetics , Leukemia, Myeloid, Acute/complications , RUNX1 Translocation Partner 1 Protein/analysis , Child , Humans , Leukemia, Myeloid, Acute/genetics , Male
7.
Proc Natl Acad Sci U S A ; 113(32): 9075-80, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27457952

ABSTRACT

Chromosomal translocation 8;21 is found in 40% of the FAB M2 subtype of acute myeloid leukemia (AML). The resultant in-frame fusion protein AML1-ETO (AE) acts as an initiating oncogene for leukemia development. AE immortalizes human CD34(+) cord blood cells in long-term culture. We assessed the transforming properties of the alternatively spliced AE isoform AE9a (or alternative splicing at exon 9), which is fully transforming in a murine retroviral model, in human cord blood cells. Full activity was realized only upon increased fusion protein expression. This effect was recapitulated in the AE9a murine AML model. Cotransduction of AE and AE9a resulted in a strong selective pressure for AE-expressing cells. In the context of AE, AE9a did not show selection for increased expression, affirming observations of human t(8;21) patient samples where full-length AE is the dominant protein detected. Mechanistically, AE9a showed defective transcriptional regulation of AE target genes that was partially corrected at high expression. Together, these results bring an additional perspective to our understanding of AE function and highlight the contribution of oncogene expression level in t(8;21) experimental models.


Subject(s)
Cell Transformation, Neoplastic , Core Binding Factor Alpha 2 Subunit/physiology , Leukemia, Myeloid, Acute/etiology , Oncogene Proteins, Fusion/physiology , RUNX1 Translocation Partner 1 Protein/physiology , Animals , Cells, Cultured , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/analysis , Disease Models, Animal , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Oncogene Proteins, Fusion/analysis , Protein Isoforms , RUNX1 Translocation Partner 1 Protein/analysis , Translocation, Genetic
8.
Dev Biol ; 415(1): 111-121, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27105579

ABSTRACT

The de novo generation of hematopoietic cells occurs during midgestation when a population of endothelial cells called hemogenic endothelium transitions into hematopoietic progenitors and stem cells. In mammalian embryos, the newly formed hematopoietic cells form clusters in the lumens of the major arteries in the embryo proper and in the vascular plexus of the yolk sac. Small clusters of hematopoietic cells that are independent of the vasculature (referred to here as extravascular islands) were shown to form in the mesentery during vascular remodeling of the vitelline artery. Using three-dimensional imaging of whole mouse embryos we demonstrate that extravascular budding of hematopoietic clusters is a more widespread phenomenon that occurs from the vitelline and the umbilical arteries both proximal to the embryo proper and distal in the extraembryonic yolk sac and placenta. Furthermore, we show that there are several mechanisms by which hematopoietic clusters leave the arteries, including vascular remodeling and extrusion. Lastly, we provide static images suggesting that extravascular islands contribute to the formation of new blood vessels. Thus, extravascular islands may represent a novel mechanism of vasculogenesis whereby established vessels contribute endothelial and hematopoietic cells to developing vascular beds.


Subject(s)
Hematopoietic Stem Cells/cytology , Mesentery/embryology , Neovascularization, Physiologic/physiology , Animals , Antigens, Ly/analysis , Core Binding Factor Alpha 2 Subunit/analysis , Lymphatic System/embryology , Membrane Proteins/analysis , Mesentery/cytology , Mice , Microscopy, Confocal , Organ Specificity , Umbilical Arteries/embryology , Vascular Remodeling , Yolk Sac/blood supply
9.
Zhongguo Dang Dai Er Ke Za Zhi ; 18(1): 34-8, 2016 Jan.
Article in Chinese | MEDLINE | ID: mdl-26781410

ABSTRACT

OBJECTIVE: To investigate the application of multiplex ligation-dependent probe amplification (MLPA) in the detection of copy number variations (CNVs) in pediatric ETV6/RUNX1-positive acute lymphoblastic leukemia (ALL), to compare this method with conventional karyotype analysis and fluorescence in situ hybridization (FISH), and to evaluate the value of MLPA. METHODS: The clinical data of 95 children with ETV6/RUNX1-positive ALL who were treated from January 2006 to November 2012 were analyzed retrospectively, including clinical features, results of karyotype analysis, and results of FISH. CNVs were detected with MLPA. RESULTS: CNVs were detected in 73 (77%), and the median number of CNVs was 1 (range 0-6). The CNVs of EBF1, CDKN2A/2B, PAX5, ETV6, RB1, and BTG1 were detected in more than 10% of all the patients. The changes in the chromosome segments carrying the genes with CNVs detected by MLPA were not detected by conventional karyotype analysis. The coincidence rate between the CNVs in ETV6 gene detected by FISH and those detected by MLPA was 66%. CONCLUSIONS: MLPA is an efficient and convenient method to detect CNVs in children with ETV6/RUNX1-positive ALL.


Subject(s)
Core Binding Factor Alpha 2 Subunit/analysis , DNA Copy Number Variations , Multiplex Polymerase Chain Reaction/methods , Oncogene Proteins, Fusion/analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Child , Child, Preschool , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Male
10.
Asia Pac J Clin Oncol ; 12(3): e375-82, 2016 Sep.
Article in English | MEDLINE | ID: mdl-25227983

ABSTRACT

AIM: CD44v6 is an isoform of CD44 that can be present in soluble form (sCD44v6). The aim of this study is to evaluate the presence of soluble CD44 (sCD44) and sCD44v6 in serum of children with B-cell precursor acute lymphoblastic leukemia (B-ALL) and their relationship with prognosis. METHODS: sCD44v6 and sCD44 levels were measured in the sera of patients and healthy children by enzyme-linked immunosorbent assay. The level of the molecules was analyzed in relation to laboratory and clinical characteristics of the patients at presentation and response to therapy. RESULT: sCD44v6 was significantly lower in patients (103.4 ± 44 ng/mL) than in controls (173.5 ± 73.6 ng/mL) whereas the serum level of sCD44 showed no significant difference between the groups. In patients, sCD44v6 quantity was inversely correlated with sCD44 level (r = -0.57, P < 0.01). The mean serum level of sCD44 in patients with >20% positivity for CD44 surface expression was greater than that in patients with ≤20% positivity (1345 ± 409 ng/mL vs 1111 ± 390 ng/mL, P = 0.05). sCD44v6 showed no significant association with response to therapy and prognostic factors except the TEL/AML1 positivity, as it was higher in TEL/AML1 positive patients (157.3 ± 55.6 ng/mL) than negative ones (92 ± 43.6 ng/mL, P = 0.036). Conversely, sCD44 was lower in TEL/AML1 positive patients and showed a significant association with white blood cell number, blast percentage and extramedullary involvement. CONCLUSION: The lower level of sCD44v6 in patients than in controls suggests the possible diagnostic value of this molecule for B-ALL. The presence of an association with established prognostic factors despite of no relationship with disease outcome suggested these molecules for more studies in larger patient cohorts.


Subject(s)
Hyaluronan Receptors/blood , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Adolescent , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit/analysis , Female , Humans , Infant , Male , Oncogene Proteins, Fusion/analysis , Prognosis
11.
Leukemia ; 30(1): 32-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26202931

ABSTRACT

Deletions in IKZF1 are found in ~15% of children with B-cell precursor acute lymphoblastic leukemia (BCP-ALL). There is strong evidence for the poor prognosis of IKZF1 deletions affecting exons 4-7 and exons 1-8, but evidence for the remaining 33% of cases harboring other variants of IKZF1 deletions is lacking. In an international multicenter study we analyzed the prognostic value of these rare variants in a case-control design. Each IKZF1-deleted case was matched to three IKZF1 wild-type controls based on cytogenetic subtype, treatment protocol, risk stratification arm, white blood cell count and age. Hazard ratios for the prognostic impact of rare IKZF1 deletions on event-free survival were calculated by matched pair Cox regression. Matched pair analysis for all 134 cases with rare IKZF1 deletions together revealed a poor prognosis (P<0.001) that was evident in each risk stratification arm. Rare variant types with the most unfavorable event-free survival were DEL 2-7 (P=0.03), DEL 2-8 (P=0.002) and DEL-Other (P<0.001). The prognosis of each type of rare variant was equal or worse compared with the well-known major DEL 4-7 and DEL 1-8 IKZF1 deletion variants. We therefore conclude that all variants of rare IKZF1 deletions are associated with an unfavorable prognosis in pediatric BCP-ALL.


Subject(s)
Gene Deletion , Ikaros Transcription Factor/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adolescent , Adult , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit/analysis , Humans , Infant , International Cooperation , Oncogene Proteins, Fusion/analysis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Prognosis , Proportional Hazards Models
12.
Jpn J Clin Oncol ; 46(2): 185-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26685324

ABSTRACT

Stage IA non-small-cell lung cancer cases have been recognized as having a low risk of relapse; however, occasionally, relapse may occur. To predict clinical outcome in Stage IA non-small-cell lung cancer patients, we searched for chimeric transcripts that can be used as biomarkers and identified a novel chimeric transcript, RUNX1-GLRX5, comprising RUNX1, a transcription factor, and GLRX5. This chimera was detected in approximately half of the investigated Stage IA non-small-cell lung cancer patients (44/104 cases, 42.3%). Although there was no significant difference in the overall survival rate between RUNX1-GLRX5-positive and -negative cases (P = 0.088), a significantly lower relapse rate was observed in the RUNX1-GLRX5-positive cases (P = 0.039), indicating that this chimera can be used as a biomarker for good prognosis in Stage IA patients. Detection of the RUNX1-GLRX5 chimeric transcript may therefore be useful for the determination of a postoperative treatment plan for Stage IA non-small-cell lung cancer patients.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/pathology , Core Binding Factor Alpha 2 Subunit/analysis , Glutaredoxins/analysis , Lung Neoplasms/chemistry , Lung Neoplasms/pathology , Aged , Chimera , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Neoplasm Staging , Postoperative Period , Predictive Value of Tests , Prognosis , Survival Analysis
13.
Int J Hematol ; 101(4): 319-29, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25749719

ABSTRACT

The differentiation from multipotent hematopoietic stem cells (HSC) to mature and functional blood cells requires the finely tuned regulation of gene expression at each stage of development. Specific transcription factors play a key role in this process as they modulate the expression of their target genes in an exquisitely lineage-specific manner. A large number of important transcriptional regulators have been identified which establish and maintain specific gene expression patterns during hematopoietic development. Hematopoiesis is therefore a paradigm for investigating how transcription factors function in mammalian cells, thanks also to the evolution of genome-wide and the next-generation sequencing technologies. In this review, we focus on the current knowledge of the biological and functional properties of the hematopoietic master regulator RUNX1 (also known as AML1, CBFA2, PEBP2aB) transcription factor and its main downstream target PU.1. We will outline their relationship in determining the fate of the myeloid lineage during normal stem cell development and under conditions when hematopoietic development is subverted by leukemic transformation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoiesis , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcriptional Activation , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Core Binding Factor Alpha 2 Subunit/analysis , Core Binding Factor Alpha 2 Subunit/genetics , Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Trans-Activators/analysis , Trans-Activators/genetics
14.
Pediatr Blood Cancer ; 61(10): 1761-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24920269

ABSTRACT

BACKGROUND: To determine the prognostic significance of the detection of the minimal residual disease (MRD) in children with AML1/ETO AML, we compared the results of reverse-transcription polymerase chain reaction (RT-PCR) and quantitative reverse-transcription polymerase chain reaction (RQ-PCR). PROCEDURE: Between January 2006 and February 2013, 70 patients (≤16 years of age) with AML1/ETO AML were included in our study. Bone marrow samples were evaluated using by both RT-PCR and RQ-PCR assays. AML1/ETO transcripts were normalized to 10(5) ABL copies. RESULTS: When treated with fewer than four courses of therapy, no association was found between positive RT-PCR results and relapse. After four courses of therapy, a positive RT-PCR result was correlated with a probability of relapse. After induction chemotherapy, a >1.8 log reduction in AML1/ETO transcripts in BM determined by RQ-PCR may represent a subgroup of patients at low risk for relapse. MRD levels after consolidation (Courses 2 and 3) were also informative. CONCLUSION: Both RT-PCR and RQ-PCR can be used to detect MRD in childhood AML1/ETO AML. RQ-PCR can identify patients who are at high risk of relapse earlier than can RT-PCR.


Subject(s)
Biomarkers, Tumor/analysis , Core Binding Factor Alpha 2 Subunit/analysis , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Neoplasm, Residual , Oncogene Proteins, Fusion/genetics , Prognosis , Proportional Hazards Models , ROC Curve , RUNX1 Translocation Partner 1 Protein , Transcription, Genetic , Treatment Outcome
17.
Int J Cancer ; 132(6): 1260-71, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23180629

ABSTRACT

The Runt-related transcription factors (RUNX) belong to an ancient family of metazoan genes involved in developmental processes. Through multiple protein-interacting partners, RUNX proteins have been implicated in diverse signaling pathways and cellular processes. The frequent inactivation of RUNX genes in cancer indicates crucial roles for RUNX in tumor suppression. This review discusses the abilities of RUNX proteins, in particular RUNX3, to integrate oncogenic signals or environmental cues and to initiate appropriate tumor suppressive responses.


Subject(s)
Core Binding Factor alpha Subunits/physiology , Neoplasms/etiology , Animals , Cell Cycle , Core Binding Factor Alpha 2 Subunit/analysis , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor Alpha 3 Subunit/analysis , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/physiology , Humans , Mutation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/physiology , Serine-Threonine Kinase 3 , Signal Transduction/physiology , Transcription, Genetic , Transforming Growth Factor beta/physiology , Tumor Suppressor Protein p53/physiology , Wnt Signaling Pathway/physiology
18.
Chin Med J (Engl) ; 125(6): 1182-5, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22613552

ABSTRACT

Childhood leukemia bottleneck phenomenon is the most mysterious corollary of the prenatal origin discovery of leukemogenic chromosome translocations. The bottleneck is evidence that leukemia initiation, by in utero acquired chromosome translocations that generate functional fusion genes, is far more common than the incidence rate of corresponding leukemia. For childhood TEL-AML1(+) acute lymphoblastic leukemia (ALL) this equates to approximately 100 times. Practically this means that among a hundred children born with TEL-AML1 fusion gene, only one child will later in its life develop ALL. The key data necessary for unraveling of this mystery were discovered in 2002. It was the level of TEL-AML1(+) cells' frequency. The bottleneck is caused by the very low body TEL-AML1(+) cell count. Only one out of a thousand B cells carries TEL-AML1 fusion gene. TEL-AML1(+) body cell count is low because TEL-AML1 fusion is generated at cell level of 10(-3) to 10(-4) just during the late fetal lymphopoiesis i.e. after the 36th gestational week.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , Core Binding Factor Alpha 2 Subunit/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic , Child , Core Binding Factor Alpha 2 Subunit/analysis , Humans , Infant, Newborn , Models, Genetic , Oncogene Proteins, Fusion/analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology
19.
Dev Biol ; 364(2): 114-27, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22326227

ABSTRACT

The sensory neurons of the dorsal root ganglia (DRG) must project accurately to their central targets to convey proprioceptive, nociceptive and mechanoreceptive information to the spinal cord. How these different sensory modalities and central connectivities are specified and coordinated still remains unclear. Given the expression of the POU homeodomain transcription factors Brn3a/Pou4f1 and Brn3b/Pou4f2 in DRG and spinal cord sensory neurons, we determined the subtype specification of DRG and spinal cord sensory neurons as well as DRG central projections in Brn3a and Brn3b single and double mutant mice. Inactivation of either or both genes causes no gross abnormalities in early spinal cord neurogenesis; however, in Brn3a single and Brn3a;Brn3b double mutant mice, sensory afferent axons from the DRG fail to form normal trajectories in the spinal cord. The TrkA(+) afferents remain outside the dorsal horn and fail to extend into the spinal cord, while the projections of TrkC(+) proprioceptive afferents into the ventral horn are also impaired. Moreover, Brn3a mutant DRGs are defective in sensory neuron specification, as marked by the excessive generation of TrkB(+) and TrkC(+) neurons as well as TrkA(+)/TrkB(+) and TrkA(+)/TrkC(+) double positive cells at early embryonic stages. At later stages in the mutant, TrkB(+), TrkC(+) and parvalbumin(+) neurons diminish while there is a significant increase of CGRP(+) and c-ret(+) neurons. In addition, Brn3a mutant DRGs display a dramatic down-regulation of Runx1 expression, suggesting that the regulation of DRG sensory neuron specification by Brn3a is mediated in part by Runx1. Our results together demonstrate a critical role for Brn3a in generating DRG sensory neuron diversity and regulating sensory afferent projections to the central targets.


Subject(s)
Axons/physiology , Ganglia, Spinal/embryology , Spinal Cord/embryology , Transcription Factor Brn-3A/physiology , Animals , Calcitonin Gene-Related Peptide/analysis , Core Binding Factor Alpha 2 Subunit/analysis , Down-Regulation , Female , Ganglia, Spinal/growth & development , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Male , Mice , Neurogenesis/physiology , Proto-Oncogene Proteins c-ret/analysis , Receptor, trkA/analysis , Receptor, trkB/analysis , Receptor, trkC/analysis , Spinal Cord/growth & development , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3B/genetics , Transcription Factor Brn-3B/physiology
20.
Pediatr Blood Cancer ; 59(3): 461-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22238115

ABSTRACT

BACKGROUND: Genetic examination of childhood leukemia has not been available in Iraq. We here report the frequency of TEL-AML1, E2A-PBX1, MLL-AF4, and BCR-ABL chimeric transcripts in 264 Iraqi children newly diagnosed with acute lymphoblastic leukemia (ALL), using FTA cards impregnated with bone marrow aspirate or whole blood. PATIENTS AND METHODS: The diagnosis of ALL was made according to standard French-American-British morphologic criteria. Based on the results of storage temperature and duration, most of the FTA samples were preserved at 4°C for up to 6 weeks in five Iraqi hospitals and then transferred to Japan for molecular analysis. Nested reverse transcription-polymerase chain reaction was adopted for the analysis. RESULTS: TEL-AML1 chimeric transcript product was found in 32 (12.1%) of 264 ALL patients. Eleven (4.2%) patients, 4 (1.5%) patients, and 11 (4.2%) patients had E2A-PBX1 mRNA, MLL-AF4 mRNA, and BCR-ABL mRNA, respectively. One patient had both TEL-AML1 and E2A-PBX1 fusion genes. The incidence of TEL-AML1 in Iraqi ALL children appears to be similar to or slightly higher than those of Jordan (12%) and Kuwait (7%). The prevalence and clinical findings of ALL patients with either E2A-PBX1 or BCR-ABL were comparable to the data reported elsewhere. CONCLUSION: International collaboration via FTA cards may be helpful to improve diagnosis and management of patients with hematological malignancies in low-income and underdeveloped countries.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit/analysis , Female , Fusion Proteins, bcr-abl/analysis , Homeodomain Proteins/analysis , Humans , Infant , Iraq , Male , Myeloid-Lymphoid Leukemia Protein/analysis , Oncogene Proteins, Fusion/analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , RNA, Messenger , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL