Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where João Agostinho Machado-Neto is active.

Publication


Featured researches published by João Agostinho Machado-Neto.


Journal of Clinical Pathology | 2017

Single-nucleotide polymorphism array (SNP-A) improves the identification of chromosomal abnormalities by metaphase cytogenetics in myelodysplastic syndrome.

da Silva Fb; João Agostinho Machado-Neto; Bertini Vh; Velloso Ed; Ratis Ca; Rodrigo T. Calado; Belinda Pinto Simões; Eduardo M. Rego; Fabiola Traina

Aims The myelodysplastic syndromes (MDS) are a heterogeneous group of clonal haematopoietic stem cell disorders characterised by inefficient haematopoiesis and risk of progression to acute myeloid leukaemia. Metaphase cytogenetics is an extremely valuable clinical tool in the management of haematological malignancies. However, metaphase cytogenetics requires cellular proliferation, its sensitivity and resolution depends on the proportion of clonal cells in the sample and size of the lesion, respectively. Single-nucleotide polymorphism array (SNP-A) does not depend on the presence of dividing cells, is able to detect copy number variations with a high resolution and to detect copy number neutral loss of heterozygosity or uniparental disomy (UPD). The aim of this study was to illustrate that the use of SNP-A can cover cryptic chromosomal lesions not identified by metaphase cytogenetics in patients with MDS. Methods Metaphase cytogenetics was performed on bone marrow aspirate using standard methods. Genomic DNA from total bone marrow cells were submitted to SNP-A using Affymetrix Genome-Wide Human SNP CytoScan HD. Results In our cohort of 15 patients with a diagnosis of MDS and related diseases, chromosomal abnormalities were found in 47% of the cases by SNP-A and in 33% by metaphase cytogenetics. SNP-A detected all lesions identified by metaphase cytogenetics, except a balanced translocation and a marker chromosome. Notably, SNP-A detected a total of 30 new lesions: 1 (3%) gain, 17 (57%) losses and 12 (40%) UPDs in 5 patients with MDS. Conclusions SNP-A may complement metaphase cytogenetics to improve the detection of chromosomal abnormalities in myeloid neoplasms.


Leukemia Research | 2016

Reversine triggers mitotic catastrophe and apoptosis in K562 cells.

Ana Paula Nunes Rodrigues Alves; João Agostinho Machado-Neto; Priscila Santos Scheucher; Helder Henrique Paiva; Belinda Pinto Simões; Eduardo M. Rego; Fabiola Traina

Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasm of the hematopoietic stem cell characterized by presence of the oncoprotein BCR-ABL1, which have constitutive tyrosine kinase activity. BCR-ABL1 activation induces aurora kinase A (AURKA) and aurora kinase B (AURKB) expression, which are serine-threonine kinases that play an important function in chromosome alignment, segregation and cytokinesis during mitosis. Acquisition of resistance to tyrosine kinase inhibitors has emerged as a problem for CML patients and the identification of novel targets with an important contribution for CML phenotype is of interest. In the present study, we explored the cellular effects of reversine, an AURKA and AURKB inhibitor, in the BCR-ABL1+ K562 cells. Our results indicate that reversine reduces AURKA and AURKB expression, leads to reduction of cell viability and increased apoptosis in a dose- and time-dependent manner, as well as, induces mitotic catastrophe in K562 cells. Our preclinical study establishes that reversine presents an effective antileukemia activity against K562 cells and provide new insights on anticancer opportunities for CML.


Journal of Cellular Biochemistry | 2017

IRS1/β-Catenin Axis Is Activated and Induces MYC Expression in Acute Lymphoblastic Leukemia Cells

Jaqueline Cristina Fernandes; Ana Paula Nunes Rodrigues Alves; João Agostinho Machado-Neto; Renata Scopim-Ribeiro; Bruna Alves Fenerich; Belinda Pinto Simões; Eduardo M. Rego; Fabiola Traina

Insulin‐like growth factor 1 (IGF1) and its receptor IGF1R regulate normal cell growth and contribute to cell transformation through activation of downstream signaling pathways. In fibroblast cells, insulin receptor substrate 1 (IRS1), through IGF1 signaling, was found to be the key protein for nuclear translocation of β‐catenin and MYC transcription activation. We herein investigated the IRS1/β‐catenin axis in acute lymphoblastic leukemia (ALL) cells. Samples were obtained from 45 patients with ALL and 13 healthy donors. ALL cell lines were used. Gene expression was measured by quantitative PCR. Protein expression, associations, and cellular localization were evaluated by immunoprecipitation, subcellular fractionation, and confocal microscopy. Cells were submitted to IGF1 stimulation and/or IGF1R pharmacological inhibition (OSI‐906). IRS1, β‐catenin, and MYC mRNA expression were significantly elevated in ALL patients, compared to normal controls. MYC mRNA expression positively correlated with β‐catenin and IRS1. Increased age and MYC expression negatively affected overall survival by univariate analysis. Total and phospho‐IGF1R and IRS1, MYC and β‐catenin protein expression were higher in ALL cells, compared to normal peripheral blood mononuclear cells (PBMC). IRS1 and β‐catenin were found to be colocalized in the nuclei and the cytoplasm of ALL cell lines, whereas both proteins were only slightly detected in the cytoplasm of normal PBMC. In Jurkat cells, a constitutive IRS1 and β‐catenin protein interaction were observed; OSI‐906 treatment decreased IGF1R tyrosine phosphorylation, IRS1 expression and phosphorylation, nuclear translocation of β‐catenin, IRS1 and β‐catenin association, and MYC protein expression. In conclusion, the IRS1/β‐catenin axis is activated in ALL cells. J. Cell. Biochem. 118: 1774–1781, 2017.


Cell Death and Disease | 2018

Metformin exerts multitarget antileukemia activity in JAK2V617F-positive myeloproliferative neoplasms

João Agostinho Machado-Neto; Bruna Alves Fenerich; Renata Scopim-Ribeiro; Christopher A. Eide; Juan Luiz Coelho-Silva; Carlos R.P. Dechandt; Jaqueline Cristina Fernandes; Ana Paula Nunes Rodrigues Alves; Priscila Santos Scheucher; Belinda Pinto Simões; Luciane C. Alberici; Lorena Lôbo de Figueiredo Pontes; Cristina E. Tognon; Brian J. Druker; Eduardo M. Rego; Fabiola Traina

The recurrent gain-of-function JAK2V617F mutation confers growth factor-independent proliferation for hematopoietic cells and is a major contributor to the pathogenesis of myeloproliferative neoplasms (MPN). The lack of complete response in most patients treated with the JAK1/2 inhibitor ruxolitinib indicates the need for identifying novel therapeutic strategies. Metformin is a biguanide that exerts selective antineoplastic activity in hematological malignancies. In the present study, we investigate and compare effects of metformin and ruxolitinib alone and in combination on cell signaling and cellular functions in JAK2V617F-positive cells. In JAK2V617F-expressing cell lines, metformin treatment significantly reduced cell viability, cell proliferation, clonogenicity, and cellular oxygen consumption and delayed cell cycle progression. Metformin reduced cyclin D1 expression and RB, STAT3, STAT5, ERK1/2 and p70S6K phosphorylation. Metformin plus ruxolitinib demonstrated more intense reduction of cell viability and induction of apoptosis compared to monotherapy. Notably, metformin reduced Ba/F3 JAK2V617F tumor burden and splenomegaly in Jak2V617F knock-in-induced MPN mice and spontaneous erythroid colony formation in primary cells from polycythemia vera patients. In conclusion, metformin exerts multitarget antileukemia activity in MPN: downregulation of JAK2/STAT signaling and mitochondrial activity. Our exploratory study establishes novel molecular mechanisms of metformin and ruxolitinib action and provides insights for development of alternative/complementary therapeutic strategies for MPN.


Biochimica et Biophysica Acta | 2018

The U2AF homology motif kinase 1 (UHMK1) is upregulated upon hematopoietic cell differentiation

Isabella Barbutti; João Agostinho Machado-Neto; Vanessa Cristina Arfelli; Paula de Melo Campos; Fabiola Traina; Sara Teresinha Olalla Saad; Leticia Froehlich Archangelo

UHMK1 (KIS) is a nuclear serine/threonine kinase that possesses a U2AF homology motif and phosphorylates and regulates the activity of the splicing factors SF1 and SF3b155. Mutations in these components of the spliceosome machinery have been recently implicated in leukemogenesis. The fact that UHMK1 regulates these factors suggests that UHMK1 might be involved in RNA processing and perhaps leukemogenesis. Here we analyzed UHMK1 expression in normal hematopoietic and leukemic cells as well as its function in leukemia cell line. In the normal hematopoietic compartment, markedly higher levels of transcripts were observed in differentiated lymphocytes (CD4+, CD8+ and CD19+) compared to the progenitor enriched subpopulation (CD34+) or leukemia cell lines. UHMK1 expression was upregulated in megakaryocytic-, monocytic- and granulocytic-induced differentiation of established leukemia cell lines and in erythrocytic-induced differentiation of CD34+ cells. No aberrant expression was observed in patient samples of myelodysplastic syndrome (MDS), acute myeloid (AML) or lymphoblastic (ALL) leukemia. Nonetheless, in MDS patients, increased levels of UHMK1 expression positively impacted event free and overall survival. Lentivirus mediated UHMK1 knockdown did not affect proliferation, cell cycle progression, apoptosis or migration of U937 leukemia cells, although UHMK1 silencing strikingly increased clonogenicity of these cells. Thus, our results suggest that UHMK1 plays a role in hematopoietic cell differentiation and suppression of autonomous clonal growth of leukemia cells.


Applied Cancer Research | 2018

Increased levels of cyclin D1 negatively impacts on acute lymphoblastic leukemia overall survival

Jaqueline Cristina Fernandes; Ana Paula Nunes Rodrigues Alves; Juan Luiz Coelho-Silva; Renata Scopim-Ribeiro; Bruna Alves Fenerich; Belinda Pinto Simões; Eduardo M. Rego; João Agostinho Machado-Neto; Fabiola Traina

BackgroundCyclin D1 is a protein essential for transition from G1 to S phase during cell cycle progression, which has an oncogenic potential and is highly expressed in several human malignancies. However, in view of the heterogeneity of the findings in the literature, the prognostic value of cyclin D1 expression still needs to be validated in different cohorts of adult acute lymphoblastic leukemia (ALL) patients.MethodsBone marrow samples from 13 healthy donors and 45 adult patients with acute lymphoblastic leukemia were included. Cyclin D1 gene expression was evaluated by quantitative PCR. For statistical analysis, Mann–Whitney test, Fisher’s exact test, Chi-squared test and Cox regression were used, as appropriate. All p values were two-sided with a significance level of 5%.ResultsCyclin D1 mRNA levels were similar between primary cells from ALL patients and healthy donors. In ALL patients, high cyclin D1 expression was associated with older age at the diagnosis, presence of BCR-ABL1, and lower white blood cell counts. Importantly, increased cyclin D1 expression was an independent factor that predicted worse overall survival in our adult ALL cohort.ConclusionIncreased levels of cyclin D1 negatively impacted on ALL survival outcome, suggesting that this gene is involved in the malignant phenotype of ALL.


Heliyon | 2017

Paclitaxel induces Stathmin 1 phosphorylation, microtubule stability and apoptosis in acute lymphoblastic leukemia cells

João Agostinho Machado-Neto; Ana Paula Nunes Rodrigues Alves; Jaqueline Cristina Fernandes; Juan Luiz Coelho-Silva; Renata Scopim-Ribeiro; Bruna Alves Fenerich; Priscila Santos Scheucher; Belinda Pinto Simões; Eduardo M. Rego; Fabiola Traina

Acute lymphoblastic leukemia (ALL) is a hematological malignancy characterized by abnormal proliferation and accumulation of lymphoblasts in the hematopoietic system. Stathmin 1 is a proliferation marker for normal lymphocytes, which has been described as highly expressed in ALL patients and functionally important for leukemia phenotype. In the present study, we expand our previous observations and aim to investigate Stathmin 1 expression and its impact on laboratory features and clinical outcomes in an independent cohort of ALL patients, and to verify the effects of paclitaxel treatment on Stathmin 1 phosphorylation and cell viability in ALL cell lines. In ALL patients, Stathmin 1 expression was significantly increased, associated with lower age onset and positively correlated with white blood cell counts, but did not impact on clinical outcomes. Functional assays revealed that paclitaxel induces Stathmin 1 phosphorylation at serine 16 (an inhibitory site), microtubule stability and apoptosis in Jurkat and Namalwa cell lines. Paclitaxel treatment did not modulate cell viability of normal peripheral blood leukocytes. In conclusion, our data confirm increased levels of Stathmin 1 in ALL patients and that therapeutic doses of paclitaxel inhibits Stathmin 1 function and promote microtubule stability and apoptosis in ALL cells.


Applied Cancer Research | 2017

TET2 is upregulated during erythroid differentiation of CD34+ cells from healthy donors and myelodysplastic syndrome patients

Renata Scopim-Ribeiro; João Agostinho Machado-Neto; Adriana da Silva Santos Duarte; Fernando Ferreira Costa; Sara Teresinha Olalla Saad; Fabiola Traina

BackgroundTet methylcytosine dioxygenase 2 (TET2) is frequently mutated and/or downregulated in myeloid neoplasm, including myelodysplastic syndromes. Despite the extensive studies, the specific contribution of TET2 in disease phenotype of myeloid neoplasms is not fully elucidated. Recent findings have grown attention on the role of TET2 in normal and malignant erythropoiesis.MethodsIn the present study, we investigated TET2 mRNA levels by quantitative PCR during erythropoietin-induced erythroid differentiation CD34+ cells from healthy donor and myelodysplastic syndrome patients. Statistical analyses were performed using the ANOVA and Bonferroni post hoc test and a p-value <0.05 was considered statically significant.ResultsTET2 expression is upregulated during erythroid differentiation of CD34+ cells from healthy donor and myelodysplastic syndrome patients.ConclusionsOur findings corroborate that TET2 is involved in the erythrocyte differentiation.


Applied Cancer Research | 2017

IL32 expression in peripheral blood CD3+ cells from myelodysplastic syndromes patients

Matheus Rodrigues Lopes; João Kleber Novais Pereira; Fabiola Traina; Paula de Melo Campos; João Agostinho Machado-Neto; Sara Teresinha Olalla Saad; Patricia Favaro

BackgroundMyelodysplastic syndromes (MDS) are a heterogeneous group of disorders characterized by ineffective hematopoiesis and risk of leukemia transformation. There is evidence to suggest the participation of immune system deregulation in MDS pathogenesis. Interleukin-32 (IL-32) is a newly described multifunctional cytokine reported as an important mediator in autoimmune and inflammatory disorders. In the present study, we reported the expression of IL32 and IL32 transcript variants (α, β, γ and δ) in peripheral blood CD3+ cells from healthy controls and MDS patients.MethodsCD3+ cells were isolated by immunomagnetic cell sorting from thirty-nine untreated MDS patients and twenty-nine healthy donors. Gene expression was evaluated by quantitative PCR. For statistical analysis, Mann–Whitney test, Kruskal-Wallis test with Dunns post test and Log-rank (Mantel-Cox) were used, as appropriate. A p value <0.05 was considered statistically significant.ResultsIL32 expression and IL32 transcript variants IL32α, IL32β, IL32γ, and IL32δ, were similar in peripheral blood CD3+ cells from healthy donors and MDS patients. Increased IL-32α expression was an independent predictor for MDS disease progression by univariate and multivariate analysis.ConclusionsWe observed that IL32 expression is not differently expressed in CD3+ cells from MDS patients; nevertheless IL32α has a potential role in disease progression.


Revista Brasileira De Hematologia E Hemoterapia | 2016

Reactive oxygen species overload promotes apoptosis in JAK2V617F-positive cell lines

João Agostinho Machado-Neto; Fabiola Traina

Philadelphia chromosome-negative myeloproliferative neo-plasms (MPNs), including essential thrombocythemia (ET),polycythemia vera (PV) and primary myelofibrosis (PMF),are characterized by excessive myeloid proliferation, withpredominant megakaryocytic, erythroid, and megakary-ocytic/granulocytic expansion, respectively, and have apotential of transformation to acute myeloid leukemia.

Collaboration


Dive into the João Agostinho Machado-Neto's collaboration.

Top Co-Authors

Avatar

Fabiola Traina

University of São Paulo

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Irene Lorand-Metze

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patricia Favaro

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Paula de Melo Campos

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge