Network


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

Hotspot


Dive into the research topics where Paula de Melo Campos is active.

Publication


Featured researches published by Paula de Melo Campos.


PLOS ONE | 2014

CXCR7 is highly expressed in acute lymphoblastic leukemia and potentiates CXCR4 response to CXCL12.

Rita de Cássia Carvalho Melo; Ana Leda Longhini; Carolina L. Bigarella; Mariana Ozello Baratti; Fabiola Traina; Patricia Favaro; Paula de Melo Campos; Sara Teresinha Olalla Saad

Recently, a novel CXCL12-binding receptor, has been identified. This CXCL12-binding receptor commonly known as CXCR7 (CXC chemokine receptor 7), has lately, based on a novel nomenclature, has received the name ACKR3 (atypical chemokine receptor 3). In this study, we aimed to investigate the expression of CXCR7 in leukemic cells, as well as its participation in CXCL12 response. Interesting, we clearly demonstrated that CXCR7 is highly expressed in acute lymphoid leukemic cells compared with myeloid or normal hematopoietic cells and that CXCR7 contributed to T-acute lymphoid leukemic cell migration induced by CXCL12. Moreover, we showed that the cellular location of CXCR7 varied among T-lymphoid cells and this finding may be related to their migration capacity. Finally, we hypothesized that CXCR7 potentiates CXCR4 response and may contribute to the maintenance of leukemia by initiating cell recruitment to bone marrow niches that were once occupied by normal hematopoietic stem cells.


Leukemia Research | 2014

Stathmin 1 is involved in the highly proliferative phenotype of high-risk myelodysplastic syndromes and acute leukemia cells

João Agostinho Machado-Neto; Paula de Melo Campos; Patricia Favaro; Mariana Lazarini; Irene Lorand-Metze; Fernando Ferreira Costa; Sara Teresinha Olalla Saad; Fabiola Traina

Stathmin 1 is an important cytoplasmic microtubule-destabilizing protein that plays critical roles in proliferation and accurate chromosome segregation through regulation of microtubule dynamics. High levels of Stathmin 1 expression have been reported in leukemia and solid tumors. However, Stathmin 1 has not been studied in myelodysplastic syndrome cells. We, herein, report that significantly higher Stathmin 1 levels were observed in proliferating hematopoietic cells, in high-risk MDS and acute leukemia cells. In addition, Stathmin 1 silencing in U937 and Namalwa leukemia cells reduced cell proliferation and clonogenicity. Our data suggest that Stathmin 1 expression may be related to the highly proliferative phenotype of hematopoietic cells and add new insights into the participation of Stathmin 1 in hematological malignancies.


PLOS ONE | 2013

Immunophenotyping in Myelodysplastic Syndromes Can Add Prognostic Information to Well-Established and New Clinical Scores

Suiellen C. Reis-Alves; Fabiola Traina; Guilherme Harada; Paula de Melo Campos; Sara Teresinha Olalla Saad; Konradin Metze; Irene Lorand-Metze

Background myelodysplastic syndromes (MDS) are a heterogeneous group of hematopoietic clonal disorders. So, prognostic variables are important to separate patients with a similar biology and clinical outcome. We compared the importance of risk stratification in primary MDS of IPSS and WPSS with the just described revision of IPSS (IPSS-R), and examined if variables obtained by bone marrow immunophenotyping could add prognostic information to any of the scores. Methods In this prospective study of 101 cases of primary MDS we compared the relation of patients’ overall survival with WHO types, IPSS, IPSS-R, WPSS and phenotypic abnormalities of hematopoietic precursors. We examined aberrancies in myelomonocytic precursors and CD34+ cells. Patients were censored when receiving chemotherapy or BM transplantation. Survival analysis was made by Cox regressions and stability of the models was examined by bootstrap resampling. Results median age: 64 years (15–93). WHO types: 2 cases of 5q- syndrome, 7 of RA, 64 of RCDM and 28 of RAEB. In the univariate Cox analysis, increasing risk category of all scores, degree of anemia, higher percentage of BM blasts, higher number of CD34+ cells and their myeloid fractions besides increasing number of phenotypic abnormalities detected were significantly associated with a shorter survival. In the multivariate analysis comparing the three scores, IPSS-R was the only independent risk factor. Comparing WPSS with phenotypic variables (CD34+/CD13+ cells, CD34+/CD13− cells and “total alterations”) the score and “CD34+/CD13+ cells” remained in the model. When IPSS was tested together with these phenotypic variables, only “CD34+/CD13+ cells”, and “total alterations” remained in the model. Testing IPSS-R with the phenotypic variables studied, only the score and “CD34+/CD13+ cells” entered the model. Conclusions Immunophenotypic analysis of myelomonocytic progenitors provides additional prognostic information to all clinical scores studied. IPSS-R improved risk stratification in MDS compared to the former scores.


Leukemia Research | 2014

Familial systemic mastocytosis with germline KIT K509I mutation is sensitive to treatment with imatinib, dasatinib and PKC412.

Paula de Melo Campos; João Agostinho Machado-Neto; Renata Scopim-Ribeiro; Valeria Visconte; Ali Tabarroki; Adriana da Silva Santos Duarte; Flávia Fonseca de Carvalho Barra; José Vassalo; Heesun J. Rogers; Irene Lorand-Metze; Ramon V. Tiu; Fernando Ferreira Costa; Sara Teresinha Olalla Saad; Fabiola Traina

Mastocytosis are myeloproliferative neoplasms commonly related to gain-of-function mutations involving the tyrosine kinase domain of KIT. We herein report a case of familial systemic mastocytosis with the rare KIT K509I germ line mutation affecting two family members: mother and daughter. In vitro treatment with imatinib, dasatinib and PKC412 reduced cell viability of primary mast cells harboring KIT K509I mutation. However, imatinib was more effective in inducing apoptosis of neoplastic mast cells. Both patients with familial systemic mastocytosis had remarkable hematological and skin improvement after three months of imatinib treatment, suggesting that it may be an effective front line therapy for patients harboring KIT K509I mutation.


European Journal of Haematology | 2015

Ten-eleven-translocation 2 (TET2) is downregulated in myelodysplastic syndromes.

Renata Scopim-Ribeiro; João Agostinho Machado-Neto; Paula de Melo Campos; Cleide Aparecida Moreira Silva; Patricia Favaro; Irene Lorand-Metze; Fernando Ferreira Costa; Sara Teresinha Olalla Saad; Fabiola Traina

TET2, a member of the ten‐eleven‐translocation (TET) family genes that modify DNA by converting 5‐methylcytosine (5‐mC) to 5‐hydroxymethylcytosine (5‐hmC), is located in chromosome 4q24 and is frequently mutated in myeloid malignancies. The impact of TET2 mutation on survival outcomes is still controversial; however, functional studies have proved that it is a loss‐of‐function mutation that impairs myeloid cell differentiation and contributes to the phenotype of myeloid neoplasia. We, herein, aimed to investigate TET2 expression in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). A significantly decreased TET2 expression was observed in bone marrow cells from AML (n = 53) and patients with MDS (n = 64), compared to normal donors (n = 22). In MDS, TET2 expression was significantly reduced in RAEB‐1/RAEB‐2 compared to other WHO 2008 classifications, and a lower TET2 expression was observed at the time of MDS disease progression in four of five patients. In multivariate analysis, low TET2 expression (P = 0.03), male gender (P = 0.02), and WHO 2008 classification (P < 0.0001) were independent predictors of poorer overall survival. These results suggest that defective TET2 expression plays a role in the MDS pathophysiology and predicts survival outcomes in this disease.


Leukemia Research | 2013

IL10 inversely correlates with the percentage of CD8 + cells in MDS patients

Matheus Rodrigues Lopes; Fabiola Traina; Paula de Melo Campos; João Kleber Novais Pereira; João Agostinho Machado-Neto; Helymar da Costa Machado; Simone Cristina Olenscki Gilli; Sara Teresinha Olalla Saad; Patricia Favaro

The role of the immune system in myelodysplastic syndrome (MDS) progression has been widely accepted, although mechanisms underlying this immune dysfunction are not clear. CD4(+) and CD8(+) lymphocyte profiles in the peripheral blood of MDS patients were evaluated and correlated with clinical characteristics, the expression of FOXP3 and the anti-inflammatory cytokines IL10, TGFβ1 and CTLA4. IL10 expression inversely correlated with the percentage of CD8(+) cells and was higher in high-risk MDS. Our findings provide further evidence for the role of T cell-mediated IL10 production in MDS and strengthen the idea of distinct cytokine profiles in low and high-risk MDS.


Biochimica et Biophysica Acta | 2015

ANKHD1 silencing inhibits Stathmin 1 activity, cell proliferation and migration of leukemia cells

João Agostinho Machado-Neto; Mariana Lazarini; Patricia Favaro; Paula de Melo Campos; Renata Scopim-Ribeiro; Gilberto Carlos Franchi Junior; Alexandre E. Nowill; Paulo Roberto Moura Lima; Fernando Ferreira Costa; Serge Benichou; Sara Teresinha Olalla Saad; Fabiola Traina

ANKHD1 is highly expressed in human acute leukemia cells and potentially regulates multiple cellular functions through its ankyrin-repeat domains. In order to identify interaction partners of the ANKHD1 protein and its role in leukemia cells, we performed a yeast two-hybrid system screen and identified SIVA, a cellular protein known to be involved in proapoptotic signaling pathways. The interaction between ANKHD1 and SIVA was confirmed by co-imunoprecipitation assays. Using human leukemia cell models and lentivirus-mediated shRNA approaches, we showed that ANKHD1 and SIVA proteins have opposing effects. While it is known that SIVA silencing promotes Stathmin 1 activation, increased cell migration and xenograft tumor growth, we showed that ANKHD1 silencing leads to Stathmin 1 inactivation, reduced cell migration and xenograft tumor growth, likely through the inhibition of SIVA/Stathmin 1 association. In addition, we observed that ANKHD1 knockdown decreases cell proliferation, without modulating apoptosis of leukemia cells, while SIVA has a proapoptotic function in U937 cells, but does not modulate proliferation in vitro. Results indicate that ANKHD1 binds to SIVA and has an important role in inducing leukemia cell proliferation and migration via the Stathmin 1 pathway. ANKHD1 may be an oncogene and participate in the leukemia cell phenotype.


Leukemia & Lymphoma | 2014

YAP1 expression in myelodysplastic syndromes and acute leukemias.

João Agostinho Machado-Neto; Paula de Melo Campos; Sara Teresinha Olalla Saad; Fabiola Traina

We read with great interest the article by Safari and colleagues [1], which investigated the gene expression of the Hippo signaling pathway members, mammalian STE-20 kinase 1 and 2 (MST1/2) and Yes...


Leukemia Research | 2013

MDR-1 and GST polymorphisms are involved in myelodysplasia progression.

Bruna Palodetto; Paula de Melo Campos; Bruno Deltreggia Benites; Maria de Lourdes Lopes Ferrari Chauffaille; Elvira Deolinda Rodrigues Pereira Velloso; Fabiola Traina; Sara Teresinha Olalla Saad

Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal stem cell disorders characterized by abnormal hematopoietic differentiation and maturation, which progress toward acute leukemia in approximately 30% of the cases. Drug metabolism polymorphisms in Cytochrome P450 2B6 (CYP2B6), Glutathione S-transferase (GST) and Dehydrogenase Quinone 1 (NQO1) enzymes and P-glycoprotein (MDR-1) could modify enzyme activity. Thus, the aim of this study was to identify the influence of CYP2B6 G15631T, GSTT1, GSTM1, NQO1 C609T and MDR-1 C3435T polymorphisms on MDS progression. We analyzed 78 MDS patients using the PCR-RFLP and multiplex method. The frequency of GST deletions and MDR-1 CC genotype was lower in progression-free patients compared to patients with progression; GST: 17% vs. 35% (P=0.018); MDR-1 gene: 19% vs. 48% (P=0.012). We also verified the influence of GST deletions and MDR-1 C3435T on patient overall survival and found no significant difference (RR=0.75; P=0.599 and RR=0.79; P=0.594 respectively). We concluded that GSTM1 deletion may contribute toward MDS progression probably due to toxic metabolite accumulation which generates cell toxicity and DNA damage. Moreover, MDR-1 C3435T may have a protective effect against MDS progression because the expected lower expression of P-glycoprotein would lead to a higher degree of cell death. To the best of our knowledge, this is the first study showing the relationship of these polymorphisms with MDS progression.


Oncotarget | 2015

Stathmin 1 inhibition amplifies ruxolitinib-induced apoptosis in JAK2 V617F cells

João Agostinho Machado-Neto; Paula de Melo Campos; Patricia Favaro; Mariana Lazarini; Adriana da Silva Santos Duarte; Irene Lorand-Metze; Fernando Costa; Sara Teresinha Olalla Saad; Fabiola Traina

The JAK/STAT pathway is constitutively activated in myeloproliferative neoplasms and can be inhibited by ruxolitinib, a selective JAK1/2 inhibitor. The JAK2V617F mutation leads to constitutive STAT3 phosphorylation and potentially leads to inhibition of Stathmin 1 activity via STAT3. In support of this hypothesis, we found that, in HEL JAK2V617F cells, ruxolitinib treatment decreased STAT3 and Stathmin 1 association, induced Stathmin 1 activation and microtubule instability. Silencing of Stathmin 1 significantly reduced cell proliferation and clonal growth, and increased apoptosis induced by ruxolitinib. Stathmin 1 silencing also prevented ruxolitinib-induced microtubule instability. To phenocopy the effect of Stathmin 1 inhibition, cells were treated with paclitaxel, a microtubule-stabilizing drug, in association or not with ruxolitinib; combined treatment significantly increased apoptosis, when compared to monotherapy. Notably, Stathmin 1 mRNA levels were highly expressed in CD34+ cells from primary myelofibrosis patients. We then proposed that an undesired effect of ruxolitinib treatment may constitute Stathmin 1 activation and microtubule instability in JAK2V617F cells. Induction of microtubule stability, through Stathmin 1 silencing or paclitaxel treatment, combined with ruxolitinib could be an effective strategy for promoting apoptosis in JAK2V617F cells.

Collaboration


Dive into the Paula de Melo Campos'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

Irene Lorand-Metze

State University of Campinas

View shared research outputs
Top Co-Authors

Avatar

Patricia Favaro

Federal University of São Paulo

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mariana Lazarini

State University of Campinas

View shared research outputs
Researchain Logo
Decentralizing Knowledge