Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Front Immunol ; 14: 1133225, 2023.
Article in English | MEDLINE | ID: mdl-37388738

ABSTRACT

Humanity did surprisingly well so far, considering how unprepared it was to respond to the coronavirus disease 2019 (COVID-19) threat. By blending old and ingenious new technology in the context of the accumulated knowledge on other human coronaviruses, several vaccine candidates were produced and tested in clinical trials in record time. Today, five vaccines account for the bulk of the more than 13 billion doses administered worldwide. The ability to elicit biding and neutralizing antibodies most often against the spike protein is a major component of the protection conferred by immunization but alone it is not enough to limit virus transmission. Thus, the surge in numbers of infected individuals by newer variants of concern (VOCs) was not accompanied by a proportional increase in severe disease and death rate. This is likely due to antiviral T-cell responses, whose evasion is more difficult to achieve. The present review helps navigating the very large literature on T cell immunity induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and vaccination. We examine the successes and shortcomings of the vaccinal protection in the light of the emergence of VOCs with breakthrough potential. SARS-CoV-2 and human beings will likely coexist for a long while: it will be necessary to update existing vaccines to improve T-cell responses and attain better protection against COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , T-Lymphocytes , Humanities , Vaccination
2.
Blood Adv ; 5(18): 3633-3646, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34438446

ABSTRACT

Insulin and insulin-like growth factors (IGFs) are mitogenic and prosurvival factors to many different cell types, including acute lymphoblastic leukemia (ALL). Circulating IGFs are bound by IGF binding proteins (IGFBPs) that regulate their action. IGFBP7 is an IGFBP-related protein (IGFBP-rP) that in contrast to other IGFBPs/IGFBP-rPs features higher affinity for insulin than IGFs and was shown to bind the IGF1 receptor (IGF1R) as well. The role of IGFBP7 in cancer is controversial: on some tumors, it functions as an oncogene, whereas in others, it functions as a tumor suppressor. In childhood ALL, higher IGFBP7 expression levels were associated with worse prognosis. Here we show that IGFBP7 exerts mitogenic and prosurvival autocrine effects on ALL cells that were dependent on insulin/IGF. IGFBP7 knockdown or antibody-mediated neutralization resulted in significant attenuation of ALL cell viability in vitro and leukemia progression in vivo. IGFBP7 was shown to prolong the surface retention of the IGF1R under insulin/IGF1 stimulation, resulting in sustained IGF1R, insulin receptor substrate 1 (IRS-1), protein kinase B (AKT), and extracellular signal-regulated kinase (ERK) phosphorylation. Conversely, the insulin receptor was readily internalized and dephosphorylated on insulin stimulation, despite IGFBP7 addition. The affinity of homodimeric IGF1R for insulin is reportedly >100 times lower than for IGF1. In the presence of IGFBP7, however, 25 ng/mL insulin resulted in IGF1R activation levels equivalent to that of 5 ng/mL IGF1. In conclusion, IGFBP7 plays an oncogenic role in ALL by promoting the perdurance of IGF1R at the cell surface, prolonging insulin/IGF stimulation. Preclinical data demonstrate that IGFBP7 is a valid target for antibody-based therapeutic interventions in ALL.


Subject(s)
Insulin-Like Growth Factor Binding Proteins , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Cell Proliferation , Cell Survival , Humans , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Phosphorylation , Receptor, IGF Type 1/genetics
3.
J Immunol ; 205(10): 2566-2575, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32958687

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which rapidly became a pandemic of global proportions. Sepsis is commonly present with high lethality in the severe forms of the disease. The virus-induced cytokine storm puts the immune system in overdrive at the expense of the pathogen-specific immune response and is likely to underlie the most advanced COVID-19 clinical features, including sepsis-related multiple organ dysfunction as well as the pathophysiological changes found in the lungs. We review the major therapeutic strategies that have been considered for sepsis and might be amenable to repurposing for COVID-19. We also discuss two different immunization strategies that have the potential to confer antiviral heterologous protection: innate-induced trained immunity and adaptive-induced immune response resetting.


Subject(s)
Adaptive Immunity , Betacoronavirus/immunology , Coronavirus Infections , Cytokines/immunology , Immunity, Innate , Multiple Organ Failure , Pandemics , Pneumonia, Viral , COVID-19 , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Coronavirus Infections/therapy , Humans , Multiple Organ Failure/immunology , Multiple Organ Failure/pathology , Multiple Organ Failure/therapy , Pneumonia, Viral/immunology , Pneumonia, Viral/pathology , Pneumonia, Viral/therapy , SARS-CoV-2
4.
Mol Genet Genomic Med ; 8(9): e1168, 2020 09.
Article in English | MEDLINE | ID: mdl-32592449

ABSTRACT

BACKGROUND: The p.Arg337His mutation of the TP53 is the most frequent germline missense variant associated with cancer described so far in this gene. It is mainly found in the South and Southeastern regions of Brazil, where it has been associated with a high incidence of pediatric adrenocortical (ACT) and choroid plexus tumors. The frequency and geographic distribution of this mutation is largely unknown, except for the Parana State, where a mean prevalence of 0.27% was reported. In the present study, we developed a high-throughput method for p.Arg337His genotyping, what allowed us to determine the frequency and geographic distribution of this mutation in a cohort from the most populous state in Brazil. METHODS: Consecutive samples from 31,612 newborns from São Paulo State were screened for p.Arg337His. The allelic discrimination was done by real-time polymerase chain reaction (PCR) and the presence of haplotype A3 in carriers was examined by using allele-specific oligonucleotide PCR, followed by nested-PCR to detect the SNP rs9894946. RESULTS: We found 67 (0.21%) samples positive for this mutation. The highest p.Arg337His frequencies were found in the cities close to the boundary between São Paulo and Minas Gerais State. No association could be found between p.Arg337His and gender, ethnicity, premature birth or twinning. Remarkably, a trend was found between the geographic distribution of p.Arg337His carriers and occurrence of ACT. CONCLUSION: We presented for the first time the p.Arg337His frequency among individuals unselected for any disease from a subset of the São Paulo State, the most populous in Brazil. The allele discrimination assay we presented here has proven to be a reliable and efficient method for high-throughput genotyping. ACT was found to be a good sentinel cancer to suppose p.Arg337His presence in our region.


Subject(s)
Adrenal Cortex Neoplasms/epidemiology , Gene Frequency , Mutation, Missense , Population/genetics , Tumor Suppressor Protein p53/genetics , Adrenal Cortex Neoplasms/genetics , Brazil , Female , Genotyping Techniques/methods , Humans , Male , Prevalence
5.
J Immunol ; 203(5): 1298-1312, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31358659

ABSTRACT

Cure of severe infections, sepsis, and septic shock with antimicrobial drugs is a challenge because morbidity and mortality in these conditions are essentially caused by improper immune response. We have tested the hypothesis that repeated reactivation of established memory to pathogens may reset unfavorable immune responses. We have chosen for this purpose a highly stringent mouse model of polymicrobial sepsis by cecum ligation and puncture. Five weeks after priming with a diverse Ag pool, high-grade sepsis was induced in C57BL/6j mice that was lethal in 24 h if left untreated. Antimicrobial drug (imipenem) alone rescued 9.7% of the animals from death, but >5-fold higher cure rate could be achieved by combining imipenem and two rechallenges with the Ag pool (p < 0.0001). Antigenic stimulation fine-tuned the immune response in sepsis by contracting the total CD3+ T cell compartment in the spleen and disengaging the hyperactivation state in the memory T subsets, most notably CD8+ T cells, while preserving the recovery of naive subsets. Quantitative proteomics/lipidomics analyses revealed that the combined treatment reverted the molecular signature of sepsis for cytokine storm, and deregulated inflammatory reaction and proapoptotic environment, as well as the lysophosphatidylcholine/phosphatidylcholine ratio. Our results showed the feasibility of resetting uncontrolled hyperinflammatory reactions into ordered hypoinflammatory responses by memory reactivation, thereby reducing morbidity and mortality in antibiotic-treated sepsis. This beneficial effect was not dependent on the generation of a pathogen-driven immune response itself but rather on the reactivation of memory to a diverse Ag pool that modulates the ongoing response.


Subject(s)
Sepsis/immunology , Animals , Apoptosis/immunology , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Cecum/immunology , Cytokines/immunology , Disease Models, Animal , Female , Immunologic Memory/immunology , Inflammation/immunology , Lipidomics/methods , Lysophosphatidylcholines/immunology , Mice , Mice, Inbred C57BL , Phosphatidylcholines/immunology , Proteomics/methods , Shock, Septic/immunology , Spleen/immunology
6.
Hum Gene Ther ; 30(4): 511-522, 2019 04.
Article in English | MEDLINE | ID: mdl-30793967

ABSTRACT

Chimeric antigen receptor (CAR) T cell immunotherapy for the treatment of cancer is now an approved treatment for B cell malignancies. However, the use of viral vectors to provide long-term CAR expression is associated with high production costs and cumbersome quality controls, impacting the final cost of CAR T cell therapies. Nonviral integrative vectors, such as Sleeping Beauty (SB) transposons, provide an alternative to modify primary T cells. Therefore, we developed a protocol to expand SB-transfected 19BBζ CAR T cells using a lymphoblastoid cell line, and evaluated T cell phenotype as well as function along the T cell expansion. Electroporation of PBMCs with transposon plasmid decreased cell viability on day 1 but had a minor impact on the frequency of memory subpopulations when compared to mock condition. CAR+ lymphocytes showed increased proliferation compared to mock control and high cytotoxic activity towards CD19+ cells without significant differences in exhaustion markers expression. Moreover, CAR+ lymphocytes showed an increased frequency by the end of the stimulation cycle compared with day 1, suggesting that CAR expression confers a selective proliferation advantage. Immunodeficient NOD scid gamma chain knockout (NSG) mice engrafted with the human pre-B leukemic cell line RS4;11 and treated with 19BBζ CAR T cells showed improved overall survival when compared to mock T cells treated animals. The results showed that electroporation using the SB system is a simple and affordable method for inducing long-term CAR expression in T lymphocytes. Expansion of gene-modified T cells with the lymphoblastoid cell line provided up to 2 cycles of stimulations, generating effective T cells against leukemia in vitro and in vivo.


Subject(s)
DNA Transposable Elements , Genetic Vectors/genetics , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Line, Transformed , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression , Genetic Vectors/administration & dosage , Humans , Immunologic Memory , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Xenograft Model Antitumor Assays
7.
Biomed Pharmacother ; 103: 1498-1506, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29864935

ABSTRACT

Stem barks of Drimys brasiliensis (Winteraceae) are consumed by the population in the form of a condiment. It is widely used to treat gastric and stomach problems and also to treat cancer. The extracts have demonstrated antiproliferative, antileishmanial and antimicrobial activities assigned to drimane sesquiterpenes. This study aimed to optimize the extraction conditions of the drimanes sesquiterpenes identified as 1-ß-(p-coumaroyloxy)-polygodial 1, drimanial 2 and 1-ß-(p-methoxycinnamoyl)-polygodial 3 in stem bark extracts. The HPLC-DAD method was developed and validated for the quantification of drimanes 1-3. The cytotoxic activity of these drimanes in human cancer cells, and the toxicological effects of the hydroethanolic extract, were determined. The extracts were prepared using different extractive conditions (solvents, plant: solvent ratio and time). The cytotoxicity effect was evaluated against leukemia, lymphomas, carcinomas and sarcomas cells using the tetrazolium assay (MTT). Furthermore, the acute toxicity was determined by measuring the biochemical parameters and by histopathological analysis. The hemolytic activity and micronucleus test were also performed. The method was linear, sensitive, precise and accurate for both drimanes 1-3. The best condition for extraction was using dichloromethane with plant: solvent proportion 1:10 (w/v) for six hours under dynamic maceration. Isolated drimanes exhibited cytotoxic effects with IC50 values ​​ranging from 0.13 to 112.67 µM. Compound 1 demonstrated significant results for acute promyelocytic leukemia (NB4) and Burkitt's lymphoma (RAMOS) cells while driamane 3 for Burkitt's lymphoma (RAJI) and acute T cell leukemia (MOLT4) cells. No signs of toxicity was observed and neither was mutagenicity or hemolytic activity.


Subject(s)
Drimys/chemistry , Plant Bark/chemistry , Plant Stems/chemistry , Sesquiterpenes/pharmacology , Sesquiterpenes/toxicity , Toxicity Tests, Acute , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Ethanol/chemistry , Hemolysis/drug effects , Humans , Limit of Detection , Micronucleus Tests , Organ Size/drug effects , Plant Extracts/pharmacology , Plant Extracts/toxicity , Polycyclic Sesquiterpenes , Rats, Wistar , Reproducibility of Results , Sesquiterpenes/chemistry
8.
Sci Rep ; 8(1): 3459, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29472583

ABSTRACT

Quercetin is one of the most abundant flavonoids, present in fruits and vegetables and has been shown to have multiple properties capable of reducing cell growth in cancer cells. Green tea is a widely consumed beverage, known for a potential source of free radical scavenging and anti-cancer activities. Herein, we investigate the in vivo antitumor efficacy of quercetin and green tea in human leukemia. Human tumors were xenografted into NOD/SCID mice. Quercetin and green tea reduced tumor growth in HL-60 xenografts accompanied by decreased expression of anti-apoptotic proteins, BCL-2, BCL-XL and MCL-1 and increased expression of BAX, a pro-apoptotic protein. Moreover, caspase-3 was activated to a greater extent after quercetin and green tea treatment. Quercetin and green tea also mediated G1 phase cell cycle arrest in HL-60 xenografts. Treatment with quercetin and green tea induced conversion of LC3-I to LC3-II as well as activation of autophagy proteins, suggesting that quercetin and green tea initiate the autophagic progression. We have provided evidence that quercetin and green tea induces signaling at the level of apoptosis, cell cycle and autophagy which converge to antigrowth effects in HL-60 xenograft mice suggesting that these compounds may be a compelling ally in cancer treatment.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Autophagy/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Leukemia/drug therapy , Quercetin/therapeutic use , Tea , Animals , Cyclin D1/metabolism , Female , HL-60 Cells , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Microtubule-Associated Proteins/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
9.
Naunyn Schmiedebergs Arch Pharmacol ; 389(7): 791-7, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27095358

ABSTRACT

Drimys brasiliensis Miers (Winteraceae) is used in folk medicine for the treatment of cancer. Its anti-tumor activity has been demonstrated in vitro models using extracts and isolated compounds. This study investigates the cytotoxic effects of stem bark extracts of D. brasiliensis as well as isolated compounds that may be responsible for the activitys and evaluates them in leukemia cells. The stem bark extract were subjected to column chromatography, and the structures of compounds were elucidated based on spectroscopic methods by using NMR and infrared spectroscopy and GC/MS. The cytotoxicity of the isolated compounds was evaluated in chronic myeloid (K562) and acute B lymphoblastic (Nalm6) leukemia cells using tetrazolium assay (MTT). Two new compounds were isolated 1ß-O-p-methoxy-E-cinnamoyl-5α-keto-11α-enol-albicanol (1a) and the isomer 1ß-O-p-methoxy-E-cinnamoyl-5α-keto-11ß-enol-albicanol (1b) and 1ß-O-p-methoxy-E-cinnamoyl-isodrimeninol (2). The known compounds polygonal acid (3a) and the isomer isopolygonal acid (3b), fuegin (4a) and the isomer epifuegin (4b), the mixture drimanial (5) and 1ß-O-(p-methoxy-E-cinnamoyl)-6α-hydroxypolygodial (6) were also isolated. The drimanes (1-4) and drimanial (5), 1ß-(p-coumaroyloxy)-polygodial (7), 1ß-(p-methoxycinnamoyl)-polygodial (8), and polygodial (9) isolated previously were assessed in tumor cells. The IC50 values were between 3.56 and 128.91 µM. 1-ß-(p-cumaroiloxi)-polygodial showed the best result with IC50 8.18 and 3.56 µM by K562 and Nalm6, respectively. The chloroform extract of the stem bark of D. brasiliensis is a great source of drimane sesquiterpenes. Our experimental data suggest that drimanes are responsible for cytotoxicity activity demonstrated by this species, especially those with the aldehyde group linked to carbons C-11 and C-12.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drimys/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Sesquiterpenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gas Chromatography-Mass Spectrometry , Humans , Inhibitory Concentration 50 , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Magnetic Resonance Spectroscopy , Molecular Structure , Phytotherapy , Plant Bark/chemistry , Plant Stems/chemistry , Plants, Medicinal , Polycyclic Sesquiterpenes , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification , Spectrophotometry, Infrared , Structure-Activity Relationship
10.
Rev. bras. farmacogn ; 26(1): 56-61, Jan.-Feb. 2016. tab, graf
Article in English | LILACS | ID: lil-772641

ABSTRACT

Abstract The essential oil of Chenopodium ambrosioides L., Amaranthaceae, was obtained by steam distillation in a Clevenger apparatus and characterization was performed using chromatographic and spectroscopic assays (GC-FID, GC/MS, 1H NMR). Two major compounds were identified: p-cymene (42.32%) and ascaridole (49.77%). The ethanolic extract and hydrolate were fractionated by liquid–liquid partitioning and the compounds were characterized by GC/MS. The essential oil, ethanol extract and fractions by partitioning with dicloromethane, ethyl acetate and butanol were tested in tumor cell lines (K562, NALM6, B15, and RAJI). Significant cytotoxic activity was found for essential oil (IC50 = 1.0 µg/ml) for RAJI cells and fraction dicloromethane (IC50 = 34.0 µg/ml) and ethanol extract (IC50 = 47.0 µg/ml) for K562 cells. The activity of the essential oil of C. ambrosioides is probably related to the large amount of ascaridol, since the other major compound, p-cymene, is recognized as a potent anti-inflammatory and has low cytotoxic activity.

11.
PLoS One ; 10(8): e0134783, 2015.
Article in English | MEDLINE | ID: mdl-26302043

ABSTRACT

Acute Lymphoblastic Leukemia (ALL) is the most frequent childhood malignancy. In the effort to find new anti-leukemic agents, we evaluated the small drug SB225002 (N-(2-hydroxy-4-nitrophenyl)-N'-(2-bromophenyl)urea). Although initially described as a selective antagonist of CXCR2, later studies have identified other cellular targets for SB225002, with potential medicinal use in cancer. We found that SB225002 has a significant pro-apoptotic effect against both B- and T-ALL cell lines. Cell cycle analysis demonstrated that treatment with SB225002 induces G2-M cell cycle arrest. Transcriptional profiling revealed that SB225002-mediated apoptosis triggered a transcriptional program typical of tubulin binding agents. Network analysis revealed the activation of genes linked to the JUN and p53 pathways and inhibition of genes linked to the TNF pathway. Early cellular effects activated by SB225002 included the up-regulation of GLIPR1, a p53-target gene shown to have pro-apoptotic activities in prostate and bladder cancer. Silencing of GLIPR1 in B- and T-ALL cell lines resulted in increased resistance to SB225002. Although SB225002 promoted ROS increase in ALL cells, antioxidant N-Acetyl Cysteine pre-treatment only modestly attenuated cell death, implying that the pro-apoptotic effects of SB225002 are not exclusively mediated by ROS. Moreover, GLIPR1 silencing resulted in increased ROS levels both in untreated and SB225002-treated cells. In conclusion, SB225002 induces cell cycle arrest and apoptosis in different B- and T-ALL cell lines. Inhibition of tubulin function with concurrent activation of the p53 pathway, in particular, its downstream target GLIPR1, seems to underlie the anti-leukemic effect of SB225002.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Neoplasm Proteins/drug effects , Nerve Tissue Proteins/drug effects , Phenylurea Compounds/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Jurkat Cells , Membrane Proteins , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
13.
PLoS One ; 10(6): e0129298, 2015.
Article in English | MEDLINE | ID: mdl-26068922

ABSTRACT

Current monitoring of acute lymphoblastic leukemia (ALL) in living mice is based on FACS analysis of blood hCD45+ cells. In this work, we evaluated the use of human IGFBP2, B2M or Hsp90 as soluble markers of leukemia. ELISA for B2M and IGFBP2 resulted in high background levels in healthy animals, precluding its use. Conversely, plasma levels of Hsp90 showed low background and linear correlation to FACS results. In another experiment, we compared Hsp90 levels with percentage of hCD45+ cells in blood, bone marrow, liver and spleen of animals weekly sacrificed. Hsp90 levels proved to be a superior method for the earlier detection of ALL engraftment and correlated linearly to ALL burden and progression in all compartments, even at minimal residual disease levels. Importantly, the Hsp90/hCD45+ ratio was not altered when animals were treated with dexamethasone or a PI3K inhibitor, indicating that chemotherapy does not directly interfere with leukemia production of Hsp90. In conclusion, plasma Hsp90 was validated as a soluble biomarker of ALL, useful for earlier detection of leukemia engraftment, monitoring leukemia kinetics at residual disease levels, and pre-clinical or mouse avatar evaluations of anti-leukemic drugs.


Subject(s)
HSP90 Heat-Shock Proteins/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/blood , Dexamethasone/therapeutic use , Disease Progression , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Insulin-Like Growth Factor Binding Protein 2/blood , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Transplantation, Heterologous , Tumor Cells, Cultured
14.
Biochim Biophys Acta ; 1853(3): 583-93, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25523139

ABSTRACT

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.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Leukemia/pathology , RNA-Binding Proteins/genetics , Stathmin/metabolism , Amino Acid Sequence , Animals , Female , Gene Silencing , HEK293 Cells , Humans , Jurkat Cells , Leukemia/genetics , Leukemia/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , Stathmin/antagonists & inhibitors , U937 Cells
15.
Cancer Prev Res (Phila) ; 7(12): 1240-50, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25293876

ABSTRACT

This study proposes to investigate quercetin antitumor efficacy in vitro and in vivo, using the P39 cell line as a model. The experimental design comprised leukemic cells or xenografts of P39 cells, treated in vitro or in vivo, respectively, with quercetin; apoptosis, cell-cycle and autophagy activation were then evaluated. Quercetin caused pronounced apoptosis in P39 leukemia cells, followed by Bcl-2, Bcl-xL, Mcl-1 downregulation, Bax upregulation, and mitochondrial translocation, triggering cytochrome c release and caspases activation. Quercetin also induced the expression of FasL protein. Furthermore, our results demonstrated an antioxidant activity of quercetin. Quercetin treatment resulted in an increased cell arrest in G1 phase of the cell cycle, with pronounced decrease in CDK2, CDK6, cyclin D, cyclin E, and cyclin A proteins, decreased Rb phosphorylation and increased p21 and p27 expression. Quercetin induced autophagosome formation in the P39 cell line. Autophagy inhibition induced by quercetin with chloroquine triggered apoptosis but did not alter quercetin modulation in the G1 phase. P39 cell treatment with a combination of quercetin and selective inhibitors of ERK1/2 and/or JNK (PD184352 or SP600125, respectively), significantly decreased cells in G1 phase, this treatment, however, did not change the apoptotic cell number. Furthermore, in vivo administration of quercetin significantly reduced tumor volume in P39 xenografts and confirmed in vitro results regarding apoptosis, autophagy, and cell-cycle arrest. The antitumor activity of quercetin both in vitro and in vivo revealed in this study, point to quercetin as an attractive antitumor agent for hematologic malignancies.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Leukemia, Myelomonocytic, Chronic/drug therapy , Leukemia, Myelomonocytic, Chronic/pathology , Quercetin/pharmacology , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation , Female , Humans , Immunoenzyme Techniques , Leukemia, Myelomonocytic, Chronic/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred NOD , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Arch Pharm (Weinheim) ; 347(12): 885-95, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25283529

ABSTRACT

A novel class of benzo[d][1,3]dioxol-5-ylmethyl alkyl/aryl amide and ester analogues of capsaicin were designed, synthesized, and evaluated for their cytotoxic activity against human and murine cancer cell lines (B16F10, SK-MEL-28, NCI-H1299, NCI-H460, SK-BR-3, and MDA-MB-231) and human lung fibroblasts (MRC-5). Three compounds (5f, 6c, and 6e) selectively inhibited the growth of aggressive cancer cells in the micromolar (µM) range. Furthermore, an exploratory data analysis pointed at the topological and electronic molecular properties as responsible for the discrimination process regarding the set of investigated compounds. The findings suggest that the applied designing strategy, besides providing more potent analogues, indicates the aryl amides and esters as well as the alkyl esters as interesting scaffolds to design and develop novel anticancer agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Capsaicin/chemical synthesis , Capsaicin/pharmacology , Computer-Aided Design , Drug Design , Molecular Dynamics Simulation , Animals , Capsaicin/analogs & derivatives , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cluster Analysis , Humans , Inhibitory Concentration 50 , Mice , Molecular Structure , Principal Component Analysis , Structure-Activity Relationship
17.
Rev. bras. farmacogn ; 24(5): 545-552, Sep-Oct/2014. tab, graf
Article in English | LILACS | ID: lil-730562

ABSTRACT

The aim of this research was to evaluate the fractions obtained from the leaf, stem and roots of Allamanda schottii Pohl, Apocynaceae, responsible for the cytotoxicity, using several cell lines. Cytotoxicity was correlated with the season the part of the plant, and the major compounds were assessed. The ethanol extracts of leaves, stems and roots obtained at different seasons were evaluated in the human erythromyeloblastoid leukemia cell line (K562). Subsequently the ethanol extracts and dichloromethane fractions collected in winter were evaluated in mouse fibroblast cell line (Mus musculus) (L929), cervix adenocarcinoma (HeLa), human pre-B leukemia (Nalm6), as well as K562 cell line. The compounds plumericin, plumieride and ursolic acid isolated from ethanol extracts of the stems were evaluated in the same cell lines, as well as on breast adenocarcinoma cell line (MCF-7), and Mus musculus skin melanoma cell line (B16F10). The chromatographic profiles of the dichloromethane fractions were obtained by high performance liquid chromatography. The results revealed that the season during which A. schottii was collected, and the part of the plant analyzed, influence the cytotoxicity on the K562 cells tested. On the other hand the dichloromethane fractions, mainly from the stems and roots, are responsible for the cytoxicity on the cells tested. These results may be associated with the seasonal variation of plumericin in these parts of the plant. This information is in accordance with the HPLC analysis. The results clearly show the potential for the phytotherapeutic use of this species, and suggest that the cytotoxic activity observed may be due to the presence of plumericin, or to minor compounds not yet identified. The seasonal influence on the production of secondary metabolites was verified.

18.
J Nat Prod ; 77(6): 1377-82, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24905499

ABSTRACT

Bioactivity-guided fractionation of the separate CH2Cl2 extracts from the aerial parts of Peperomia alata and P. trineura yielded seven polyketides: alatanone A [3-hydroxy-2-(5'-phenylpent-4'E-enoyl)cyclohex-2-en-1-one, 1a] and alatanone B [3-hydroxy-2-(3'-phenyl-6'-methylenedioxypropanoyl)cyclohex-2-en-1-one, 2a] from P. alata and trineurone A [3-hydroxy-2-(11'-phenylundec-10'E-enoyl)cyclohex-2-en-1-one, 1b], trineurone B [3-hydroxy-2-(15'-phenyl-18'-methylenedioxypentadecanoyl)cyclohex-2-en-1-one, 2b], trineurone C [3-hydroxy-2-(17'-phenyl-20'-methylenedioxyheptadecanoyl)cyclohex-2-en-1-one, 2c], trineurone D [3-hydroxy-2-(hexadec-10'Z-enoyl)cyclohex-2-en-1-one, 3a], and trineurone E [(6R)-(+)-3,6-dihydroxy-2-(hexadec-10'Z-enoyl)cyclohex-2-en-1-one, 3b] from P. trineura. The isolated compounds were evaluated for antifungal activity against Cladosporium cladosporioides and C. sphaeospermum and for cytotoxicity against the K562 and Nalm-6 leukemia cell lines.


Subject(s)
Antifungal Agents/isolation & purification , Antifungal Agents/pharmacology , Piperaceae/chemistry , Polyketides/isolation & purification , Polyketides/pharmacology , Antifungal Agents/chemistry , Brazil , Cladosporium/drug effects , Drug Screening Assays, Antitumor , Humans , K562 Cells , Microbial Sensitivity Tests , Nuclear Magnetic Resonance, Biomolecular , Polyketides/chemistry
19.
Exp Cell Res ; 324(2): 137-45, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24726915

ABSTRACT

ANKHD1 is a multiple ankyrin repeat containing protein, recently identified as a novel member of the Hippo signaling pathway. The present study aimed to investigate the role of ANKHD1 in DU145 and LNCaP prostate cancer cells. ANKHD1 and YAP1 were found to be highly expressed in prostate cancer cells, and ANKHD1 silencing decreased cell growth, delayed cell cycle progression at the S phase, and reduced tumor xenograft growth. Moreover, ANKHD1 knockdown downregulated YAP1 expression and activation, and reduced the expression of CCNA2, a YAP1 target gene. These findings indicate that ANKHD1 is a positive regulator of YAP1 and promotes cell growth and cell cycle progression through Cyclin A upregulation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Cycle/genetics , Gene Expression Regulation, Neoplastic , Phosphoproteins/genetics , Prostatic Neoplasms/genetics , RNA-Binding Proteins/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cyclin A/genetics , Cyclin A/metabolism , HeLa Cells , Hippo Signaling Pathway , Humans , K562 Cells , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Phosphoproteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Transcription Factors , Transcriptional Activation , YAP-Signaling Proteins
20.
Phytochemistry ; 97: 81-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24252268

ABSTRACT

The EtOAc extract from the leaves of Piper carniconnectivum C. DC. was subjected to chromatographic separation to afford two non-aromatic B-ring flavanone compounds: 5-hydroxy-2-(1'-hydroxy-4'-oxo-cyclohex-2'-en-1'-yl)-6,7-dimethoxy-2,3-dihydro-4H-chromen-4-one (1) and 5-hydroxy-2-(1',2'-dihydroxy-4'-oxo-cyclohexyl)-6,7-dimethoxy-2,3-dihydro-4H-chromen-4-one (2). The absolute configuration of (+)-1 was unambiguously determined as 2S,1'R by electronic circular dichroism (ECD) spectroscopy and comparison to simulated spectra that were calculated using time-dependent density functional theory (TDDFT). This methodology allowed the assignment of the absolute configuration of (+)-2 also as 2S,1'R, except for the stereogenic center at C-2', which was assigned as R because of the evidence drawn from high resolution NMR experiments. The cytotoxic activity of both compounds and 3 (hydrogenated B-ring derivative of 1) was evaluated on twelve human leukemia cell lines, and the IC50 values (<10 µM) indicated the activity of 1 against seven cell lines.


Subject(s)
Flavanones/isolation & purification , Flavanones/pharmacology , Piper/chemistry , Circular Dichroism , Drug Screening Assays, Antitumor , Flavanones/chemistry , Humans , Inhibitory Concentration 50 , Nuclear Magnetic Resonance, Biomolecular , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...