Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Dev Cell ; 59(2): 244-261.e6, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38154460

ABSTRACT

WNT morphogens trigger signaling pathways fundamental for embryogenesis, regeneration, and cancer. WNTs are modified with palmitoleate, which is critical for binding Frizzled (FZD) receptors and activating signaling. However, it is unknown how WNTs are released and spread from cells, given their strong lipid-dependent membrane attachment. We demonstrate that secreted FZD-related proteins and WNT inhibitory factor 1 are WNT carriers, potently releasing lipidated WNTs and forming active soluble complexes. WNT release occurs by direct handoff from the membrane protein WNTLESS to the carriers. In turn, carriers donate WNTs to glypicans and FZDs involved in WNT reception and to the NOTUM hydrolase, which antagonizes WNTs by lipid moiety removal. WNT transfer from carriers to FZDs is greatly facilitated by glypicans that serve as essential co-receptors in Wnt signaling. Thus, an extracellular network of carriers dynamically controls secretion, posttranslational regulation, and delivery of WNT morphogens, with important practical implications for regenerative medicine.


Subject(s)
Glypicans , Wnt Proteins , Wnt Proteins/metabolism , Glypicans/metabolism , Wnt Signaling Pathway , Embryonic Development , Lipids , Frizzled Receptors/chemistry , Frizzled Receptors/metabolism
2.
Cell Rep Methods ; 3(11): 100627, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-37924815

ABSTRACT

Adrenal insufficiency is a life-threatening condition resulting from the inability to produce adrenal hormones in a dose- and time-dependent manner. Establishing a cell-based therapy would provide a physiologically responsive approach for the treatment of this condition. We report the generation of large numbers of human-induced steroidogenic cells (hiSCs) from human pluripotent stem cells (hPSCs). Directed differentiation of hPSCs into hiSCs recapitulates the initial stages of human adrenal development. Following expression of steroidogenic factor 1, activation of protein kinase A signaling drives a steroidogenic gene expression profile most comparable to human fetal adrenal cells, and leads to dynamic secretion of steroid hormones, in vitro. Moreover, expression of the adrenocorticotrophic hormone (ACTH) receptor/co-receptor (MC2R/MRAP) results in dose-dependent ACTH responsiveness. This protocol recapitulates adrenal insufficiency resulting from loss-of-function mutations in AAAS, which cause the enigmatic triple A syndrome. Our differentiation protocol generates sufficient numbers of hiSCs for cell-based therapy and offers a platform to study disorders causing adrenal insufficiency.


Subject(s)
Adrenal Insufficiency , Pluripotent Stem Cells , Humans , Glucocorticoids/pharmacology , Adrenal Insufficiency/genetics , Adrenocorticotropic Hormone/pharmacology , Pluripotent Stem Cells/metabolism , Receptors, Corticotropin
3.
Cancer Res ; 83(13): 2123-2141, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37129912

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare cancer in which tissue-specific differentiation is paradoxically associated with dismal outcomes. The differentiated ACC subtype CIMP-high is prevalent, incurable, and routinely fatal. CIMP-high ACC possess abnormal DNA methylation and frequent ß-catenin-activating mutations. Here, we demonstrated that ACC differentiation is maintained by a balance between nuclear, tissue-specific ß-catenin-containing complexes, and the epigenome. On chromatin, ß-catenin bound master adrenal transcription factor SF1 and hijacked the adrenocortical super-enhancer landscape to maintain differentiation in CIMP-high ACC; off chromatin, ß-catenin bound histone methyltransferase EZH2. SF1/ß-catenin and EZH2/ß-catenin complexes present in normal adrenals persisted through all phases of ACC evolution. Pharmacologic EZH2 inhibition in CIMP-high ACC expelled SF1/ß-catenin from chromatin and favored EZH2/ß-catenin assembly, erasing differentiation and restraining cancer growth in vitro and in vivo. These studies illustrate how tissue-specific programs shape oncogene selection, surreptitiously encoding targetable therapeutic vulnerabilities. SIGNIFICANCE: Oncogenic ß-catenin can use tissue-specific partners to regulate cellular differentiation programs that can be reversed by epigenetic therapies, identifying epigenetic control of differentiation as a viable target for ß-catenin-driven cancers.


Subject(s)
Adrenal Cortex Neoplasms , Adrenocortical Carcinoma , Humans , beta Catenin/genetics , beta Catenin/metabolism , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/metabolism , Adrenocortical Carcinoma/pathology , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Epigenesis, Genetic , Chromatin/genetics
4.
Neuro Oncol ; 25(1): 185-198, 2023 01 05.
Article in English | MEDLINE | ID: mdl-35640920

ABSTRACT

BACKGROUND: Supratentorial RELA fusion (ST-RELA) ependymomas (EPNs) are resistant tumors without an approved chemotherapeutic treatment. Unfortunately, the molecular mechanisms that lead to chemoresistance traits of ST-RELA remain elusive. The aim of this study was to assess RELA fusion-dependent signaling modules, specifically the role of the Hedgehog (Hh) pathway as a novel targetable vulnerability in ST-RELA. METHODS: Gene expression was analyzed in EPN from patient cohorts, by microarray, RNA-seq, qRT-PCR, and scRNA-seq. Inhibitors against Smoothened (SMO) (Sonidegib) and Aurora kinase A (AURKA) (Alisertib) were evaluated. Protein expression, primary cilia formation, and drug effects were assessed by immunoblot, immunofluorescence, and immunohistochemistry. RESULTS: Hh components were selectively overexpressed in EPNs induced by the RELA fusion. Single-cell analysis showed that the Hh signature was primarily confined to undifferentiated, stem-like cell subpopulations. Sonidegib exhibited potent growth-inhibitory effects on ST-RELA cells, suggesting a key role in active Hh signaling; importantly, the effect of Sonidegib was reversed by primary cilia loss. We, thus, tested the effect of AURKA inhibition by Alisertib, to induce cilia stabilization/reassembly. Strikingly, Alisertib rescued ciliogenesis and synergized with Sonidegib in killing ST-RELA cells. Using a xenograft model, we show that cilia loss is a mechanism for acquiring resistance to the inhibitory effect of Sonidegib. However, Alisertib fails to rescue cilia and highlights the need for other strategies to promote cilia reassembly, for treating ST-RELA tumors. CONCLUSION: Our study reveals a crucial role for the Hh pathway in ST-RELA tumor growth, and suggests that rescue of primary cilia represents a vulnerability of the ST-RELA EPNs.


Subject(s)
Ependymoma , Supratentorial Neoplasms , Humans , Hedgehog Proteins , Cilia/metabolism , Cilia/pathology , Aurora Kinase A/genetics , Ependymoma/pathology , Supratentorial Neoplasms/pathology , Transcription Factor RelA
5.
Med Oncol ; 39(12): 216, 2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36175721

ABSTRACT

Patients diagnosed with acute lymphoblastic leukemia (ALL) bearing t(4;11)/MLL-AF4 have aggressive clinical features, poor prognosis and there is an urgent need for new therapies to improve outcomes. Panobinostat (LBH589) has been identified as a potential therapeutic agent for ALL with t(4;11) and studies suggest that the antineoplastic effects are associated with reduced MLL-AF4 fusion protein and reduced expression of HOX genes. Here, we evaluated the in vitro effects of the combination of LBH589 with methotrexate (MTX) or 6-mercaptopurine (6MP) by cell proliferation assays and Calcusyn software in ALL cell line (RS4;11); the in vivo effects of LBH589 in xenotransplanted NOD-scid IL2Rgammanull mice measuring human lymphoblasts by flow cytometry; and the expression of HOX genes by qPCR after treatment in an adult model of ALL with t(4;11). LBH589 combination with MTX or 6MP did not promote synergistic effects in RS4;11 cell line. LBH589 treatment leads to increased overall survival and reduction of blasts in xenotransplanted mice but caused no significant changes in HOXA7, HOXA9, HOXA10, and MEIS1 expression. The LBH589, alone, showed promising antineoplastic effects in vivo and may represent a potential agent for chemotherapy in ALL patients with t(4;11).


Subject(s)
Mercaptopurine , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adult , Animals , Humans , Mercaptopurine/pharmacology , Methotrexate/pharmacology , Mice , Mice, Inbred NOD , Panobinostat/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
6.
J Cell Biochem ; 122(12): 1925-1935, 2021 12.
Article in English | MEDLINE | ID: mdl-34581457

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare, but highly aggressive cancer of the adrenal cortex with a generally poor prognosis. Despite being rare, completely resected ACCs present a high risk of recurrence. Musashi-2 (MSI2) has recently been recognized as a potential prognostic biomarker and therapeutic target in many cancers. However, no studies have evaluated the clinical significance of MSI2 expression in ACC. Here, we addressed MSI2 expression and its association with ACC prognosis and clinicopathological parameters. MSI2 expression was analyzed in TCGA, GSE12368, GSE33371, and GSE49278 ACC datasets; and its correlation with other genes and immune cell infiltration were investigated by using the R2: Genomics Analysis and Visualization Platform and TIMER databases, respectively. Enrichment analysis was performed with the DAVID Functional Annotation Tool. Kaplan-Meier curves, log-rank tests, and Cox regression analyses were used to explore the prognostic role of MSI2 in ACC. Our findings demonstrated the potential value of MSI2 overexpression as an independent predictor of poor prognosis in patients with completely resected ACC (hazard ratio 6.715, 95% confidence interval 1.266 - 35.620, p =.025). In addition, MSI2 overexpression was associated with characteristics of unfavorable prognosis, such as cortisol excess (p = .002), recurrence (p =.003), and death (p =.015); positively correlated with genes related to steroid biosynthesis (p < .05); and negatively correlated with immune-related pathways (p < .05). Our findings demonstrate that MSI2 has value as a prognostic marker for completely resected ACC and reinforce the investigation of its role as a possible therapeutic target for patients with ACC.


Subject(s)
Adrenal Cortex Neoplasms , Adrenocortical Carcinoma , Biomarkers, Tumor/immunology , Gene Expression Regulation, Neoplastic/immunology , Neoplasm Proteins/immunology , RNA-Binding Proteins/immunology , Adrenal Cortex Neoplasms/immunology , Adrenal Cortex Neoplasms/mortality , Adrenocortical Carcinoma/immunology , Adrenocortical Carcinoma/mortality , Adult , Female , Humans , Male , Middle Aged , Steroids/immunology
7.
Mol Cell Endocrinol ; 528: 111243, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33716050

ABSTRACT

Adrenocortical cancer (ACC) is a rare and aggressive type of endocrine tumor with high risk of recurrence and metastasis. The overall survival of patients diagnosed with ACC is low and treatment for metastatic stages remain limited to mitotane, which has low efficiency in advanced stages of the disease and is associated with high toxicity. Therefore, identification of new biological targets to improve ACC treatment is crucial. Blockade of the Wnt/beta-catenin pathway decreased adrenal steroidogenesis and increased apoptosis of NCI-H295 human ACC cells, in vitro and in a xenograft mouse model. Aurora kinases play important roles in cell division during the G1-M phase and their aberrant expression is correlated with a poor prognosis in different types of tumors. Hence, we hypothesized that inhibition of aurora kinases activity combined with the beta-catenin pathway blockade would improve the impairment of ACC cell growth in vitro. We studied the combinatorial effects of AMG 900, an aurora kinase inhibitor and PNU-74654, a beta-catenin pathway blocker, on proliferation, survival and tumor progression in multiple ACC cell lines: NCI-H295, CU-ACC1 and CU-ACC2. Exposure of ACC cells to the combination of AMG 900 with PNU-74654 decreased cell proliferation and viability compared to either treatment alone. In addition, AMG 900 inhibited cell invasion and clonogenesis compared to PNU-74654, and the combination showed no greater effects. In contrast, PNU-74654 was more effective in decreasing cortisol secretion. These data suggest that inhibition of aurora kinases activity combined with blockade of the beta-catenin pathway may provide a combinatorial approach for targeting ACC tumors.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Aurora Kinase A/genetics , Benzamides/pharmacology , Phthalazines/pharmacology , Wnt Signaling Pathway/drug effects , Adrenal Cortex Neoplasms/drug therapy , Aurora Kinase B/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Sequence Analysis, RNA , Up-Regulation/drug effects
8.
Sci Rep ; 10(1): 15193, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32938995

ABSTRACT

SHOC2 scaffold protein has been mainly related to oncogenic ERK signaling through the RAS-SHOC2-PP1 phosphatase complex. In leukemic cells however, SHOC2 upregulation has been previously related to an increased 5-year event-free survival of pediatric pre-B acute lymphoid leukemia, suggesting that SHOC2 could be a potential prognostic marker. To address such paradoxical function, our study investigated how SHOC2 impact leukemic cells drug response. Our transcriptome analysis has shown that SHOC2 can modulate the DNA-damage mediated by p53. Notably, upon genetic inhibition of SHOC2 we observed a significant impairment of p53 expression, which in turn, leads to the blockage of key apoptotic molecules. To confirm the specificity of DNA-damage related modulation, several anti-leukemic drugs has been tested and we did confirm that the proposed mechanism impairs cell death upon daunorubicin-induced DNA damage of human lymphoid cells. In conclusion, our study uncovers new insights into SHOC2 function and reveals that this scaffold protein may be essential to activate a novel mechanism of p53-induced cell death in pre-B lymphoid cells.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Lymphoid/metabolism , Precursor Cells, B-Lymphoid/physiology , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , DNA Damage/drug effects , Daunorubicin/therapeutic use , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Lymphoid/diagnosis , Leukemia, Lymphoid/drug therapy , MAP Kinase Signaling System , Tumor Suppressor Protein p53/genetics , ras Proteins/metabolism
9.
Mol Biol Rep ; 47(8): 6439-6443, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32671623

ABSTRACT

TP53 p.R337H germline mutation is highly prevalent in the Southern region of Brazil. We sought to investigate TP53 p.R337H mutation in pediatric tumor samples from a population settled in a geographic area of high prevalence for this variant. Mutation assessment and genetic counseling for carriers/relatives were provided. 6/57 tumor samples were heterozygous for TP53 p.R337H. As expected, a high frequency was observed within adrenocortical tumors (3/3) and choroid plexus carcinomas (2/2). Interestingly, the TP53 R337H mutation was found in one case of pediatric rhabdomyosarcoma with Li-Fraumeni pedigree. Our finding expands the spectrum of childhood cancer associated with this germline mutation.


Subject(s)
Germ-Line Mutation , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adrenal Cortex Neoplasms/epidemiology , Adrenal Cortex Neoplasms/genetics , Brazil/epidemiology , Carcinoma/epidemiology , Carcinoma/genetics , Child, Preschool , Choroid Plexus Neoplasms/epidemiology , Choroid Plexus Neoplasms/genetics , Cohort Studies , Female , Humans , Male , Mutation Rate , Neoplasms/epidemiology , Point Mutation , Rhabdomyosarcoma/epidemiology , Rhabdomyosarcoma/genetics
10.
Oncogene ; 39(30): 5282-5291, 2020 07.
Article in English | MEDLINE | ID: mdl-32561853

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare and aggressive malignancy with limited therapeutic options. The lack of mouse models that recapitulate the genetics of ACC has hampered progress in the field. We analyzed The Cancer Genome Atlas (TCGA) dataset for ACC and found that patients harboring alterations in both p53/Rb and Wnt/ß-catenin signaling pathways show a worse prognosis compared with patients that harbored alterations in only one. To model this, we utilized the Cyp11b2(AS)Cre mouse line to generate mice with adrenocortical-specific Wnt/ß-catenin activation, Trp53 deletion, or the combination of both. Mice with targeted Wnt/ß-catenin activation or Trp53 deletion showed no changes associated with tumor formation. In contrast, alterations in both pathways led to ACC with pulmonary metastases. Similar to ACCs in humans, these tumors produced increased levels of corticosterone and aldosterone and showed a high proliferation index. Gene expression analysis revealed that mouse tumors exhibited downregulation of Star and Cyp11b1 and upregulation of Ezh2, similar to ACC patients with a poor prognosis. Altogether, these data show that altering both Wnt/ß-catenin and p53/Rb signaling is sufficient to drive ACC in mouse. This autochthonous model of ACC represents a new tool to investigate the biology of ACC and to identify new treatment strategies.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Disease Models, Animal , Tumor Suppressor Protein p53/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/pathology , Animals , Cell Proliferation/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Mice, Knockout , Mice, Transgenic , Prognosis
11.
Cell Rep ; 31(3): 107524, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32320669

ABSTRACT

Activating mutations in the canonical Wnt/ß-catenin pathway are key drivers of hyperplasia, the gateway for tumor development. In a wide range of tissues, this occurs primarily through enhanced effects on cellular proliferation. Whether additional mechanisms contribute to ß-catenin-driven hyperplasia remains unknown. The adrenal cortex is an ideal system in which to explore this question, as it undergoes hyperplasia following somatic ß-catenin gain-of-function (ßcat-GOF) mutations. Targeting ßcat-GOF to zona Glomerulosa (zG) cells leads to a progressive hyperplastic expansion in the absence of increased proliferation. Instead, we find that hyperplasia results from a functional block in the ability of zG cells to transdifferentiate into zona Fasciculata (zF) cells. Mechanistically, zG cells demonstrate an upregulation of Pde2a, an inhibitor of zF-specific cAMP/PKA signaling. Hyperplasia is further exacerbated by trophic factor stimulation leading to organomegaly. Together, these data indicate that ß-catenin drives adrenal hyperplasia through both proliferation-dependent and -independent mechanisms.


Subject(s)
Adrenal Hyperplasia, Congenital/metabolism , Adrenal Hyperplasia, Congenital/pathology , beta Catenin/metabolism , Adrenal Hyperplasia, Congenital/genetics , Animals , Cell Transdifferentiation/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , beta Catenin/genetics
12.
J Mol Med (Berl) ; 98(4): 495-512, 2020 04.
Article in English | MEDLINE | ID: mdl-32219470

ABSTRACT

Glioblastoma is the most common aggressive primary brain tumor. Standard care includes maximal safe surgical resection, radiation, and chemotherapy with temozolomide. However, the impact of this therapeutic approach on patient survival is disappointing and poor outcomes are frequently observed. Therefore, new therapeutic targets are needed to treat this potentially deadly tumor. Aurora kinases are one of today's most sought-after classes of therapeutic targets to glioblastoma therapy. They are a family of proteins composed of three members: Aurora-A, Aurora-B, and Aurora-C that play different roles in the cell division through regulation of chromosome segregation. Deregulation of these genes has been reported in glioblastoma and a progressive number of studies have shown that inhibition of these proteins could be a promising strategy for the treatment of this tumor. This review discusses the preclinical and early clinical findings on the potential use of the Aurora kinases as new targets for the treatment of glioblastoma. KEY MESSAGES: GBM is a very aggressive tumor with limited therapeutic options. Aurora kinases are a family of serine/threonine kinases implicated in GBM pathology. Aurora kinases are critical for glioblastoma cell growth, apoptosis, and chemoresistance. Inhibition of Aurora kinases has a synergistic or sensitizing effect with chemotherapy drugs, radiotherapy, or with other targeted molecules in GBM. Several Aurora kinase inhibitors are currently in clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinases/antagonists & inhibitors , Glioblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Aurora Kinases/genetics , Aurora Kinases/metabolism , Biomarkers, Tumor , Glioblastoma/drug therapy , Glioblastoma/etiology , Glioblastoma/pathology , Humans , Molecular Targeted Therapy , Multigene Family , Protein Kinase Inhibitors/therapeutic use , Translational Research, Biomedical , Xenograft Model Antitumor Assays
13.
Cancer Gene Ther ; 27(6): 509-512, 2020 06.
Article in English | MEDLINE | ID: mdl-31308481

ABSTRACT

RELA-fused supratentorial (ST) ependymoma (EPN) is an aggressive subgroup with poor prognosis. Considering the putative role of Notch signaling in the maintenance of the cancer stem cells (CSC) phenotype in RELA-fused EPN, we investigated the expression of Notch pathway and its target genes in this subgroup. We also evaluated the effects of two Notch inhibitors (DAPT and RO4929097) on cell proliferation, apoptosis, colony formation, and CSCs markers gene expression on EPN cell line of the RELA-fused subgroup (BXD-1425). In addition, in silico signatures of the Notch genes and CSCs markers were analyzed on a large clinical dataset from GSE64415 study. We found that among the ST-EPN subgroups the Notch signaling (NOTCH1, JAG1, JAG2, and HES4) is specifically activated in the ST-EPN-RELA. Furthermore, treatment of the RELA-fused EPN cell line with the Notch inhibitors impaired the Notch signaling expression and revealed that Notch axis is not essential for cell proliferation and survival in this setting. NOTCH1 expression in ST-EPN was correlated with the CSCs markers VEGFA and L1CAM overexpression and JAG1 expression was correlated with the CCND1 and CDK6 overexpression. In addition, in vitro treatment with Notch inhibitors induced downregulation of CSCs markers. These findings indicate that Notch signaling can be involved in the ST-EPN-RELA CSCs maintenance by modulating the expression of genes responsible for cell phenotype and cell fate.


Subject(s)
Ependymoma/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Notch/metabolism , Supratentorial Neoplasms/metabolism , Transcription Factor RelA/metabolism , Calcium-Binding Proteins/metabolism , Ependymoma/pathology , Humans , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Supratentorial Neoplasms/pathology , Up-Regulation
14.
Neurol Sci ; 41(3): 691-694, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31728854

ABSTRACT

BACKGROUND: Ependymoma (EPN) is the third most common childhood cancer of the central nervous system. RELA fusion-positive EPN accounts for approximately 70% of all childhood supratentorial tumors and shows the worst prognosis among the supratentorial EPNs. TP53 mutation is infrequent in RELA fusions EPNs. In the population from the Southern region of Brazil, there is a high incidence of the germline TP53 p.R337H mutation that predisposes carriers to develop early-onset tumors. However, despite this high incidence, the frequency of this mutation among EPN patients remains to be determined. Here, we investigated the presence of the TP53 p.R337H mutation in a larger cohort of pediatric EPNs of three institutions located in the state of São Paulo, Brazil. METHODS: The TP53 p.R337H mutation was screened by conventional RT-PCR and Sanger sequencing in 49 pediatric EPNs diagnosed during the period from 1995 to 2016. RESULTS: We described for the first time a case of a 5-year-old girl with RELA fusion EPN with a heterozygous TP53 p.R337H mutation. CONCLUSIONS: The present finding indicates that the TP53 p.R337H germline mutation is uncommon in patients with EPN in Brazil and screening of pediatric patients RELA fusion EPN may be informative to better understand the role of TP53 germline mutations in the development and prognosis of these tumors.


Subject(s)
Ependymoma/genetics , Supratentorial Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Brazil/epidemiology , Child , Child, Preschool , Cohort Studies , Ependymoma/epidemiology , Female , Germ-Line Mutation , Humans , Male , Supratentorial Neoplasms/epidemiology , Transcription Factor RelA
15.
Cytotechnology ; 70(6): 1713-1722, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30374857

ABSTRACT

Medulloblastoma (MB) is the most common malignant childhood brain tumor. MB is currently classified into four molecular subgroups (Wnt, Shh, Group 3, and Group 4). The wingless (Wnt) pathway is responsible for embryonic development and is deregulated in MB. We analyzed the activation of the Wnt pathway in MB cell lines and its correlation with the Shh pathway, with emphasis on the importance of cellular characterization. Transient ß-catenin transfection led to an increase in the ß-catenin gene and protein expression in MB cell lines. Wnt pathway activation resulted in a reduced number of colonies in all cell lines studied and a significant increase in the G2/M cell cycle phase only in ONS-76 cells. Regarding the Shh pathway, transfection caused a reduced expression of the PTCH1 and SMO genes only in the UW473 cells. Further studies are needed to understand the mechanism underlying the molecular events associated with the effects of Wnt activation in MB.

16.
ACM arq. catarin. med ; 47(3): 60-73, jul.-set. 2018.
Article in Portuguese | LILACS | ID: biblio-915960

ABSTRACT

Avaliar as características da demanda e os determinantes de procura pelo SE por pacientes classificados como pouco urgentes e não-urgentes em hospital geral, Sul do Brasil. Estudo epidemiológico, transversal. A coleta de dados foi realizada por entrevista, com duração aproximada de 15 minutos, em 2017, por pesquisadores treinados. Desfecho do estudo: determinantes de procura. Variáveis independentes: demográficas, socioeconômicas e características gerais de procura. Também coletou-se dados sobre a auto percepção do paciente (urgência e preocupação) e escolha do local para atendimento (Escala Likert 0-10). Os dados qualitativos apresentados na forma de frequências simples e relativa, os quantitativos como média e desvio padrão. Foram analisadas associações entre variáveis independentes e o desfecho, através do teste qui-quadrado de Pearson, seguido de Razão de Prevalência e Intervalo Confiança 95%. Participaram do estudo 290 pacientes: 50 (17%) pouco urgentes e 240 (83,0%) não urgentes, sexo feminino (57,2%), faixa etária entre 15-29 anos (39,0%), sem companheiro (51,0%), ensino superior/médio (69,3%), em atividade ocupacional (61,2%). Sobre características gerais da demanda: 77,9% oriundos de suas residências e 39,3% utilizou veículo próprio para o deslocamento. Distância média deslocamento (12,47±15,8 Km) e tempo médio deslocamento (25,98±23,55 minutos). Tempo médio triagem (17,69min.±15,36) e de espera atendimento médico (1h13min±1h10min). Não houve associação com significância estatística entre características demográficas, socioeconômicas e motivos de procura pelo SE. Conclui-se que os critérios determinantes de procura pelo SE foram resolutividade (40,9%) e funcionamento inadequado das UBS (24,7%), seguidos de procura por especialista e agudização da doença crônica.


To evaluate the demand and the determinants of the search for the Emergency service by patients classified as not very urgent and not-urgent in a general hospital, South of Brazil. Cross epidemiological study. Data collect was performed by interview, with an approximated duration of 15 minutes, in 2017, by trained researchers. Study outcome: Search determinants. Independent variables: demographic, socioeconomic and general demand characteristics. It was also collected data on the patient self-perception (urgency and concern) and the choice of the medical care (Likert Scale 0-10). The qualitative data presented by simple and relative way frequencies, the quantitative ones as average and standard deviation. Associations between independent variables and the outcome were analyzed using Pearson's chi-square test, followed by a Prevalence Ratio and 95% Confidence Interval. The study included 290 patients: 50 (17%) were not very urgent and 240 (83.0%) female not-urgent were (57.2%), 15-29 years old (39.0%), without partner (51.0%), higher /college degree (69.3%) in occupational activity (61.2%). On general demand characteristics: 77.9% come from their homes and 39.3% used their own vehicle for the trip. Average displacement distance (12.47 ± 15.8 Km) and average displacement time (25.98 ± 23.55 minutes). Average screening time (17.69min ± 15.36) and waiting for medical care (1h13min ± 1h10min). There was no association with statistical significance between demographic, socioeconomic and reasons for Emergency Service search. The determinant criteria of the search for ES were resolutiveness (40.9%) and inadequate working of the BHU (24.7%), followed by specialist search and chronic disease exacerbation.

17.
Anticancer Drugs ; 28(6): 634-644, 2017 07.
Article in English | MEDLINE | ID: mdl-28410270

ABSTRACT

Adrenocortical tumor (ACT) is a malignancy with a low incidence rate and the current therapy for advanced disease has a limited impact on overall patient survival. A previous study from our group suggested that elevated expression of aurora-A and aurora-B is associated with poor outcome in childhood ACT. Similar results were also reported for adult ACTs. The present in-vitro study shows that AMG 900 inhibits aurora kinases in adrenocortical carcinoma cells. AMG 900 inhibited cell proliferation in NCI-H295 cells as well as in the ACT primary cultures and caused apoptosis in the cell line NCI-H295. Furthermore, it potentialized the mitotane, doxorubicin, and etoposide effects on apoptosis induction and acted synergistically with mitotane and doxorubicin in the inhibition of proliferation. In addition, we found that AMG 900 activated Notch signaling and rendered the cells sensitive to the combination of AMG 900 and Notch signaling inhibition. Altogether, these data show that aurora kinases inhibition using AMG 900 may be an adjuvant therapy to treat patients with invasive or recurrent adrenocortical carcinomas.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Adrenocortical Carcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Adrenal Cortex Neoplasms/enzymology , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/enzymology , Adrenocortical Carcinoma/pathology , Aurora Kinases/antagonists & inhibitors , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Histones/metabolism , Humans , Mitotane/administration & dosage , Mitotane/pharmacology , Phosphorylation/drug effects , Phthalazines/administration & dosage , Protein Kinase Inhibitors/administration & dosage
19.
Invest New Drugs ; 35(1): 26-36, 2017 02.
Article in English | MEDLINE | ID: mdl-27785591

ABSTRACT

Medulloblastoma (MB) is the most common solid tumor among pediatric patients and corresponds to 20 % of all pediatric intracranial tumors in this age group. Its treatment currently involves significant side effects. Epigenetic changes such as DNA methylation may contribute to its development and progression. DNA methyltransferase (DNMT) inhibitors have shown promising anticancer effects. The agent Zebularine acts as an inhibitor of DNA methylation and shows low toxicity and high efficacy, being a promising adjuvant agent for anti-cancer chemotherapy. Several studies have reported its effects on different types of tumors; however, there are no studies reporting its effects on MB. We analyzed its potential anticancer effects in four pediatric MB cell lines. The treatment inhibited proliferation and clonogenicity, increased the apoptosis rate and the number of cells in the S phase (p < 0.05), as well as the expression of p53, p21, and Bax, and decreased cyclin A, Survivin and Bcl-2 proteins. In addition, the combination of zebularine with the chemotherapeutic agents vincristine and cisplatin resulted in synergism and antagonism, respectively. Zebularine also modulated the activation of the SHH pathway, reducing SMO and GLI1 levels and one of its targets, PTCH1, without changing SUFU levels. A microarray analysis revealed different pathways modulated by the drug, including the Toll-Like Receptor pathway and high levels of the BATF2 gene. The low expression of this gene was associated with a worse prognosis in MB. Taken together, these data suggest that Zebularine may be a potential drug for further in vivo studies of MB treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Basic-Leucine Zipper Transcription Factors/genetics , Cerebellar Neoplasms/drug therapy , Cytidine/analogs & derivatives , DNA Modification Methylases/antagonists & inhibitors , Medulloblastoma/drug therapy , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Apoptosis/drug effects , Biomarkers , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Child , Child, Preschool , Cisplatin/pharmacology , Cytidine/pharmacology , DNA Modification Methylases/metabolism , Drug Interactions , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Infant, Newborn , Male , Medulloblastoma/genetics , Medulloblastoma/metabolism , Oligonucleotide Array Sequence Analysis , Prognosis , Vincristine/pharmacology , Young Adult
20.
BMC Complement Altern Med ; 15: 393, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26518729

ABSTRACT

BACKGROUND: Glioblastoma is the most common tumor of the central nervous system and one of the hardest tumors to treat. Consequently, the search for novel therapeutic options is imperative. 7-epiclusianone, a tetraprenylated benzophenone isolated from the epicarp of the native plant Garcinia brasiliensis, exhibits a range of biological activities but its prospect anticancer activity is underexplored. Thus, the aim of the present study was to evaluate the influence of 7-epiclusianone on proliferation, clonogenic capacity, cell cycle progression and induction of apoptosis in two glioblastoma cell lines (U251MG and U138MG). METHODS: Cell viability was measured by the MTS assay; for the clonogenic assay, colonies were stained with Giemsa and counted by direct visual inspection; For cell cycle analysis, cells were stained with propidium iodide and analyzed by cytometry; Cyclin A expression was determined by immunoblotting; Apoptotic cell death was determined by annexin V fluorescein isothiocyanate labeling and Caspase-3 activity in living cells. RESULTS: Viability of both cell lines was drastically inhibited; moreover, the colony formation capacity was significantly reduced, demonstrating long-term effects even after removal of the drug. 7-epiclusianone treatment at low concentrations also altered cell cycle progression, decreased the S and G2/M populations and at higher concentrations increased the number of cells at sub-G1, in concordance with the increase of apoptotic cells. CONCLUSION: The present study demonstrates for the first time the anticancer potential of 7-epiclusianone against glioblastoma cells, thus meriting its further investigation as a potential therapeutic agent.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzophenones/pharmacology , Benzoquinones/pharmacology , Garcinia/chemistry , Glioblastoma/physiopathology , Plant Extracts/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glioblastoma/drug therapy , Humans
SELECTION OF CITATIONS
SEARCH DETAIL