Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Apoptosis ; 22(7): 898-919, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28424988

RESUMO

Inhibitors of apoptosis (IAPs) are a family of proteins that play a significant role in the control of programmed cell death (PCD). PCD is essential to maintain healthy cell turnover within tissue but also to fight disease or infection. Uninhibited, IAPs can suppress apoptosis and promote cell cycle progression. Therefore, it is unsurprising that cancer cells demonstrate significantly elevated expression levels of IAPs, resulting in improved cell survival, enhanced tumor growth and subsequent metastasis. Therapies to target IAPs in cancer has garnered substantial scientific interest and as resistance to anti-cancer agents becomes more prevalent, targeting IAPs has become an increasingly attractive strategy to re-sensitize cancer cells to chemotherapies, antibody based-therapies and TRAIL therapy. Antagonism strategies to modulate the actions of XIAP, cIAP1/2 and survivin are the central focus of current research and this review highlights advances within this field with particular emphasis upon the development and specificity of second mitochondria-derived activator of caspase (SMAC) mimetics (synthetic analogs of endogenously expressed inhibitors of IAPs SMAC/DIABLO). While we highlight the potential of SMAC mimetics as effective single agent or combinatory therapies to treat cancer we also discuss the likely clinical implications of resistance to SMAC mimetic therapy, occasionally observed in cancer cell lines.


Assuntos
Apoptose/genética , Proteínas Inibidoras de Apoptose/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Mitocondriais/genética , Neoplasias/tratamento farmacológico , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Proteínas Mitocondriais/antagonistas & inibidores , Neoplasias/genética , Neoplasias/patologia , Survivina , Ligante Indutor de Apoptose Relacionado a TNF/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética
2.
Brain ; 137(Pt 2): 433-48, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24355709

RESUMO

Thymosin beta 4 is a pleiotropic actin-sequestering polypeptide that is involved in wound healing and developmental processes. Thymosin beta 4 gene silencing promotes differentiation of neural stem cells whereas thymosin beta 4 overexpression initiates cortical folding of developing brain hemispheres. A role of thymosin beta 4 in malignant gliomas has not yet been investigated. We analysed thymosin beta 4 staining on tissue microarrays and performed interrogations of the REMBRANDT and the Cancer Genome Atlas databases. We investigated thymosin beta 4 expression in seven established glioma cell lines and seven glioma-initiating cell lines and induced or silenced thymosin beta 4 expression by lentiviral transduction in LNT-229, U87MG and GS-2 cells to study the effects of altered thymosin beta 4 expression on gene expression, growth, clonogenicity, migration, invasion, self-renewal and differentiation capacity in vitro, and tumorigenicity in vivo. Thymosin beta 4 expression increased with grade of malignancy in gliomas. Thymosin beta 4 gene silencing in LNT-229 and U87MG glioma cells inhibited migration and invasion, promoted starvation-induced cell death in vitro and enhanced survival of glioma-bearing mice. Thymosin beta 4 gene silencing in GS-2 cells inhibited self-renewal and promoted differentiation in vitro and decreased tumorigenicity in vivo. Gene expression analysis suggested a thymosin beta 4-dependent regulation of mesenchymal signature genes and modulation of TGFß and p53 signalling networks. We conclude that thymosin beta 4 should be explored as a novel molecular target for anti-glioma therapy.


Assuntos
Inativação Gênica , Glioblastoma/genética , Invasividade Neoplásica/genética , Células-Tronco Neoplásicas/patologia , Timosina/antagonistas & inibidores , Timosina/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Bases de Dados Genéticas , Feminino , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Células NIH 3T3 , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/fisiologia , Timosina/biossíntese
3.
J Neurochem ; 122(2): 444-55, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22564186

RESUMO

Temozolomide (TMZ) is an alkylating chemotherapeutic agent that prolongs the survival of patients with glioblastoma. Clinical benefit is more prominent in patients with methylation of the O(6) -methyl-guanine DNA methyltransferase (MGMT) promoter. However, all patients eventually suffer from tumor progression because their tumors become resistant to TMZ. Here, we modeled acquired TMZ resistance in glioma cells in vitro to identify underlying molecular mechanisms. To this end, the glioma cell lines LNT-229, LN-308, and LN-18 were exposed repetitively to increasing concentrations of TMZ to induce a stable resistant phenotype (R) defined by clonogenic survival assays. The molecular mechanisms mediating acquired resistance were assessed by immunoblot, PCR, and flow cytometry. Rescue experiments were performed with siRNA-mediated candidate gene silencing. We found in LN-18 cells constitutively expressing MGMT a strong up-regulation of MGMT levels in TMZ-resistant cells. TMZ resistance in the MGMT-negative cell lines LNT-229 and LN-308 was not associated with de novo expression of MGMT. Instead, we found a down-regulation of several DNA mismatch-repair proteins in resistant LNT-229 cells. A TMZ-resistant phenotype was also achieved by silencing selected DNA mismatch repair proteins in parental LNT-229 cells. No obvious mechanism of resistance was identified in the third cell line, LN-308, except for reduced methylation of LINE-1 repetitive elements. In conclusion, we demonstrate that different molecular mechanisms may contribute to the development of acquired TMZ resistance in glioma cells, indicating the need to develop distinct strategies to overcome resistance.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Western Blotting , Neoplasias Encefálicas/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular , Cromatina/efeitos dos fármacos , Reparo de Erro de Pareamento de DNA , Metilases de Modificação do DNA/biossíntese , Metilases de Modificação do DNA/genética , Análise Mutacional de DNA , Reparo do DNA , Enzimas Reparadoras do DNA/biossíntese , Enzimas Reparadoras do DNA/genética , Replicação do DNA/genética , Replicação do DNA/fisiologia , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Inativação Gênica , Genes Reporter/efeitos dos fármacos , Genes Reporter/genética , Glioblastoma/genética , Humanos , Reação em Cadeia da Polimerase , Interferência de RNA , Temozolomida , Ensaio Tumoral de Célula-Tronco , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Quinases Ativadas por p21/metabolismo
4.
Toxicol Lett ; 179(1): 34-42, 2008 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-18485628

RESUMO

While arsenic compounds are known as environmental toxicants (especially in drinking water) and as carcinogens, some arsenic compounds, like arsenic trioxide (As2O3), are clinically used in humans to treat some forms of cancer (e.g. leukemia). Although arsenic compounds have been studied intensively, their interactions with living cells are still not fully elucidated. We have previously proposed that modulation of intracellular calcium ([Ca2+]i) homeostasis induced by As2O3 could be an important mechanism to induce cytotoxicity. Here we demonstrate, using human cell models (neuroblastoma (SY-5Y) or embryonic kidney cells (HEK)) and confocal microscopy in combination with the calcium sensitive dye fluo 4-AM, that As2O3 interferes with calcium signaling at low (environmentally and clinically relevant concentrations of 100 pM to 1 microM). Within this concentration range, As2O3 had cell type specific cytotoxic effects, with neuroblastoma cells being more sensitive to As2O3 than HEK 293. In addition, by staining with Hoechst 33347 and counting micronucleated cells as well as apoptotic nuclei, As2O3 was found to increase the rate of apoptosis and DNA damage, which was also cell type specific. These results indicate that the As2O3-induced cell death could be triggered or mediated by [Ca2+]i signals and suggest that low concentrations of As2O3 are able to interfere with specific physiological processes in diverse cell models.


Assuntos
Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Rim/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Óxidos/toxicidade , Trióxido de Arsênio , Arsenicais , Cálcio/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Homeostase , Humanos , Rim/metabolismo , Rim/patologia , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Microscopia Confocal , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Azul Tripano
5.
Cancers (Basel) ; 9(5)2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28486397

RESUMO

Cancer is a main public health problem all over the world. It affects millions of humans no matter their age, gender, education, or social status. Although chemotherapy is the main strategy for the treatment of cancer, a major problem limiting its success is the intrinsic or acquired drug resistance. Therefore, cancer drug resistance is a major impediment in medical oncology resulting in a failure of a successful cancer treatment. This mini-overview focuses on the interdependent relationship between intracellular calcium ([Ca2+]i) signaling and multidrug resistance of cancer cells, acquired upon treatment of tumors with anticancer drugs. We propose that [Ca2+]i signaling modulates gene expression of multidrug resistant (MDR) genes which in turn can be modulated by epigenetic factors which in turn leads to modified protein expression in drug resistant tumor cells. A precise knowledge of these mechanisms will help to develop new therapeutic strategies for drug resistant tumors and will improve current chemotherapy.

6.
Oncotarget ; 8(14): 22876-22893, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28206967

RESUMO

Neuroblastoma (NB) is a pediatric cancer treated with poly-chemotherapy including platinum complexes (e.g. cisplatin (CDDP), carboplatin), DNA alkylating agents, and topoisomerase I inhibitors (e.g. topotecan (TOPO)). Despite aggressive treatment, NB may become resistant to chemotherapy. We investigated whether CDDP and TOPO treatment of NB cells interacts with the expression and function of proteins involved in regulating calcium signaling. Human neuroblastoma cell lines SH-SY5Y, IMR-32 and NLF were used to investigate the effects of CDDP and TOPO on cell viability, apoptosis, calcium homeostasis, and expression of selected proteins regulating intracellular calcium concentration ([Ca2+]i). In addition, the impact of pharmacological inhibition of [Ca2+]i-regulating proteins on neuroblastoma cell survival was studied. Treatment of neuroblastoma cells with increasing concentrations of CDDP (0.1-10 µM) or TOPO (0.1 nM-1 µM) induced cytotoxicity and increased apoptosis in a concentration- and time-dependent manner. Both drugs increased [Ca2+]i over time. Treatment with CDDP or TOPO also modified mRNA expression of selected genes encoding [Ca2+]i-regulating proteins. Differentially regulated genes included S100A6, ITPR1, ITPR3, RYR1 and RYR3. With FACS and confocal laser scanning microscopy experiments we validated their differential expression at the protein level. Importantly, treatment of neuroblastoma cells with pharmacological modulators of [Ca2+]i-regulating proteins in combination with CDDP or TOPO increased cytotoxicity. Thus, our results confirm an important role of calcium signaling in the response of neuroblastoma cells to chemotherapy and suggest [Ca2+]i modulation as a promising strategy for adjunctive treatment.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , Neuroblastoma/tratamento farmacológico , Neuroblastoma/metabolismo , Apoptose , Linhagem Celular Tumoral , Humanos , Prognóstico
7.
Mol Endocrinol ; 19(8): 2132-44, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15860547

RESUMO

The CTRdelta e13 splice variant of the rabbit calcitonin receptor, which lacks the 14 amino acids of the seventh transmembrane domain (TMD) that are encoded by exon 13, is poorly expressed on the cell surface, fails to mobilize intracellular calcium or activate Erk, and inhibits the cell surface expression of the full-length C1a isoform. Nuclear magnetic resonance- and fluorescence-activated cell sorter-based experiments showed that the residual seventh TMD of CTRdelta e13 fails to partition into the lipid bilayer, resulting in an extracellular C terminus. Truncating the receptor after residue 397 to delete the cytoplasmic tail resulted in reduced cell surface expression and an inability to mobilize intracellular calcium or activate Erk, but the truncated receptor did not inhibit C1a cell surface expression. In contrast, when the receptor was truncated after residue 374 to eliminate the entire seventh TMD domain and the C-terminal domain, the resulting receptor reduced the cell surface expression of C1a in a manner similar to that of CTRdelta e13. Thus, normal cell surface expression, mobilization of intracellular calcium, and Erk activation requires the cytoplasmic C-terminal tail of the CTR, whereas the absence of the seventh TMD in the transmembrane helical bundle causes the dominant-negative effect on the surface expression of C1a.


Assuntos
Receptores da Calcitonina/química , Animais , Western Blotting , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Separação Celular , AMP Cíclico/metabolismo , Citoplasma/metabolismo , DNA/metabolismo , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Genes Dominantes , Humanos , Imunoprecipitação , Espectroscopia de Ressonância Magnética , Peptídeos/química , Fosforilação , Dobramento de Proteína , Isoformas de Proteínas , Estrutura Terciária de Proteína , Coelhos , Receptores da Calcitonina/metabolismo , Transdução de Sinais , Temperatura , Transfecção
8.
Oncotarget ; 7(11): 12937-50, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26887050

RESUMO

Glioblastoma is the most aggressive brain tumor in adults with a median survival below 12 months in population-based studies. The main reason for tumor recurrence and progression is constitutive or acquired resistance to the standard of care of surgical resection followed by radiotherapy with concomitant and adjuvant temozolomide (TMZ/RT→TMZ). Here, we investigated the role of microRNA (miRNA) alterations as mediators of alkylator resistance in glioblastoma cells. Using microarray-based miRNA expression profiling of parental and TMZ-resistant cultures of three human glioma cell lines, we identified a set of differentially expressed miRNA candidates. From these, we selected miR-138 for further functional analyses as this miRNA was not only upregulated in TMZ-resistant versus parental cells, but also showed increased expression in vivo in recurrent glioblastoma tissue samples after TMZ/RT→TMZ treatment. Transient transfection of miR-138 mimics in glioma cells with low basal miR-138 expression increased glioma cell proliferation. Moreover, miR-138 overexpression increased TMZ resistance in long-term glioblastoma cell lines and glioma initiating cell cultures. The apoptosis regulator BIM was identified as a direct target of miR-138, and its silencing mediated the induced TMZ resistance phenotype. Altered sensitivity to apoptosis played only a minor role in this resistance mechanism. Instead, we identified the induction of autophagy to be regulated downstream of the miR-138/BIM axis and to promote cell survival following TMZ exposure. Our data thus define miR-138 as a glioblastoma cell survival-promoting miRNA associated with resistance to TMZ therapy in vitro and with tumor progression in vivo.


Assuntos
Neoplasias Encefálicas/genética , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/genética , MicroRNAs/genética , Proteína 11 Semelhante a Bcl-2/biossíntese , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/metabolismo , Humanos
9.
Cell Calcium ; 37(3): 251-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15670872

RESUMO

Humans are exposed to organotins, like trimethyltin (TMT) chloride via air, water and food, and intoxication might result in severe health complications. Toxic effects of organotin compounds are well documented, but possible mechanisms remain unclear and only little information is available how organometallic species interact with calcium controlling mechanisms. Therefore, the aim of this work was to investigate the effects of TMT on calcium homeostasis in HeLa S3 cells. Dynamic changes of cytosolic calcium (Ca2+(i)) were monitored using laser-scanning microscopy and fluo-4 loaded cells. Application of TMT resulted in sustained as well as in transient elevations of Ca2+(i). The number of reacting cells was directly correlated to the concentration of TMT used: with 500 microM TMT all cells reacted, with 50 microM TMT 80% and with 5 microM 74%. The fast Ca2+(i)-transients (spikes), measured in single cells, occurred even with 0.25 microM TMT and varied in size and duration. The sustained increase of Ca2+(i), measured as the average over all cells, was dose dependent with an approximately 8% increase for 5 microM TMT, approximately 12.3% for 50 microM and approximately 145% for 500 microM TMT. Moreover, this effect was partly reversible. A second application resulted in a similar sustained rise of Ca2+(i) compared to the first application of TMT, there was also no difference when no calcium was added to the external solution (151+/-10% compared to 145+/-15%; 500 microM TMT). This rise of Ca2+(i) was highly reduced (<10% increase) when the internal calcium stores were depleted before TMT (500 microM) was applied. Our data suggest that TMT influences Ca2+(i)-homeostasis of HeLa S3 cells, which might be related to its toxicity in this cell line.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Homeostase/efeitos dos fármacos , Compostos de Trimetilestanho/toxicidade , Compostos de Anilina , Células HeLa , Humanos , Microscopia Confocal , Xantenos
10.
Environ Health Perspect ; 113(6): 659-64, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15929885

RESUMO

Arsenic and its methylated derivatives are contaminants of air, water, and food and are known as toxicants and carcinogens. Arsenic compounds are also being used as cancer chemotherapeutic agents. In humans, inorganic arsenic is metabolically methylated to mono-, di-, and trimethylated forms. Recent findings suggest that the methylation reactions represent a toxification rather than a detoxification pathway. In recent years, the correlation between arsenic exposure, cytotoxicity and genotoxicity, mutagenicity, and tumor promotion has been established, as well as the association of arsenic exposure with perturbation of physiologic processes, generation of reactive oxygen species, DNA damage, and apoptosis induction. Trivalent forms of arsenic have been found to induce apoptosis in several cellular systems with involvement of membrane-bound cell death receptors, activation of caspases, release of calcium stores, and changes of the intracellular glutathione level. It is well known that calcium ion deregulation plays a critical role in apoptotic cell death. A calcium increase in the nuclei might lead to toxic effects in the cell. In this review, we highlight the relationship between induced disturbances of calcium homeostasis, genomic damage, and apoptotic cell death caused by arsenic and its organic derivatives.


Assuntos
Arsênio/toxicidade , Arsenicais/efeitos adversos , Cálcio/metabolismo , Animais , Apoptose , Arsênio/metabolismo , Arsenicais/metabolismo , Biotransformação , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Dano ao DNA , Poluentes Ambientais/metabolismo , Poluentes Ambientais/toxicidade , Humanos , Metilação
11.
Toxicology ; 216(1): 1-8, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16125831

RESUMO

Physiological modifications of intracellular Ca2+ ([Ca2+]i) levels trigger and/or regulate a diversity of cellular activities (e.g. neurotransmitter release, synaptic plasticity, muscular contraction, cell proliferation), while calcium overloads could result in cytotoxicity. Previously, we have shown that trimethyltin chloride (Me3SnCl; TMT) modulates calcium homeostasis in cervix adenocarcinoma (HeLa S3) cells [Florea, A.-M., Dopp, E., Büsselberg, D., 2005. TMT induces elevated calcium transients in HeLa cells: types and levels of response. Cell Calcium 37, 252-258]. Here we compare [Ca2+]i-changes induced by trimethyltin chloride in neuroblastoma SY5Y and HeLa S3 cells using calcium-sensitive dyes (fluo-4/AM (fluo-4) and rhod-2/AM (rhod-2)) and laser scanning microscopy (LSM). TMT-induced calcium elevations in neuroblastoma SY5Y as well as in HeLa S3 cells. [Ca2+]i rose to a sustained plateau or to transient spikes. Overall, the detected averaged increase of the maximum calcium elevation were: 0.5 microM approximately 125.6%; 5 microM approximately 130.1%; 500 microM approximately 145% in HeLa S3 cells and 0.5 microM approximately 133.3%; 5 microM approximately 136.1%; 500 microM approximately 147.1% in neuroblastoma SY5Y cells. The calcium rise derived from internal stores did not significantly depend on the presence of calcium in the external solution: approximately 109% (no calcium added) versus approximately 117% (2 mM calcium; 5 microM TMT) in HeLa cells. This difference was similar in neuroblastoma SY5Y cells, were approximately 127% versus approximately 136% increase (5 microM TMT) were measured. Staining of calcium stores with rhod-2 showed a TMT-induced [Ca2+]i-decrease in the stores followed by an increase of the calcium concentration in the nuclei of the two cell lines tested. Our results suggest that toxic effects in human tumour cells after exposure to trimethyltin compounds might be due to an elevation of [Ca2+]i.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Homeostase/efeitos dos fármacos , Espaço Intracelular/metabolismo , Compostos de Trimetilestanho/toxicidade , Adenocarcinoma/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Corantes Fluorescentes , Humanos , Espaço Intracelular/efeitos dos fármacos , Microscopia Confocal , Neuroblastoma/metabolismo
12.
PLoS One ; 10(10): e0139603, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26441059

RESUMO

Immunotherapy targeting glioblastoma initiating cells (GIC) is considered a promising strategy. However, GIC are prone to evade immune response and there is a need for potent adjuvants. IFN-ß might enhance the immune response and here we define its net effect on the innate immunogenicity of GIC. The transcriptomes of GIC treated with IFN-ß and controls were assessed by microarray-based expression profiling for altered expression of immune regulatory genes. Several genes involved in adaptive and innate immune responses were regulated by IFN-ß. We validated these results using reverse transcription (RT)-PCR and flow cytometry for corresponding protein levels. The up-regulation of the NK cell inhibitory molecules HLA-E and MHC class I was balanced by immune stimulating effects including the up-regulation of nectin-2. In 3 out of 5 GIC lines tested we found a net immune stimulating effect of IFN-ß in cytotoxicity assays using NKL cells as effectors. IFN-ß therefore warrants further investigation as an adjuvant for immunotherapy targeting GIC.


Assuntos
Glioblastoma/imunologia , Imunidade Inata/efeitos dos fármacos , Interferon beta/farmacologia , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Genes MHC Classe I/genética , Glioblastoma/genética , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Nectinas , Regulação para Cima/efeitos dos fármacos
13.
J Invest Dermatol ; 135(2): 369-377, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25264594

RESUMO

Even though aging and cellular senescence appear to be linked, the biological mechanisms interconnecting these two processes remain to be unravelled. Therefore, microRNA (miRNA/miR) profiles were analyzed ex vivo by means of gene array in fibroblasts isolated from young and old human donors. Expression of several miRNAs was positively correlated with donor age. Among them, miR-23a-3p was shown to target hyaluronan synthase 2 (HAS2). HA is a polysaccharide of the extracellular matrix that critically regulates the phenotype of fibroblasts. Indeed, both aged and senescent fibroblasts showed increased miR-23a-3p expression and secreted significantly lower amounts of HA compared with young and non-senescent fibroblasts. Ectopic overexpression of miR-23a-3p in non-senescent fibroblasts led to decreased HAS2-mediated HA synthesis, upregulation of senescence-associated markers, and decreased proliferation. In addition, siRNA-mediated downregulation of HAS2 and pharmacological inhibition of HA synthesis by 4-methylumbelliferone mimicked the effects of miR-23a-3p. In vivo, miR-23a-3p was upregulated and HAS2 was downregulated in the skin of old mice compared with young mice. Inhibition of HA synthesis by 4-methylumbelliferone in mice reduced dermal hydration and viscoelasticity, thereby mimicking an aged skin phenotype. Taken together, these findings appear to link miR-23a-3p and the HA microenvironment as effector mechanisms in both dermal aging and senescence.


Assuntos
Senescência Celular , Glucuronosiltransferase/genética , MicroRNAs/fisiologia , Envelhecimento da Pele , Regiões 3' não Traduzidas/genética , Adulto , Idoso , Animais , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Feminino , Fibroblastos/metabolismo , Glucuronosiltransferase/antagonistas & inibidores , Humanos , Hialuronan Sintases , Ácido Hialurônico/metabolismo , Himecromona/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade
14.
J Toxicol Sci ; 39(5): 765-74, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25242407

RESUMO

Breast cancer (BC) is a public health problem all over the world. Cisplatin (CDDP) is an antineoplastic agent with high rate of success in treating cancers. The down side of CDDP treatment is the development of chemo-resistance. Beside DNA damage and activation of p53 signaling pathway, CDDP induces tumor-cell death due to elevation in the intracellular calcium concentration ([Ca(2+)]i).However, the role of [Ca(2+)]i in CDDP induced apoptosis of breast cancer cells (MCF-7) is not well understood. Here we investigate the cytotoxic effects of CDDP in relation to [Ca(2+)]i homeostasis in MCF-7-sensitive and -resistant cell lines. Live-cell imaging using [Ca(2+)]i sensitive fluorescent dyes was employed to monitor [Ca(2+)]i CDDP treated MCF-7 cells (0.001-10 µM) and [Ca(2+)]i modulators i.e. Caffeine (10 mM); Nimodipine (10 µM); Ionomycin (10 µM); Thapsigargin (500 nM). A concentration-dependent increase of[Ca(2+)]i was observed in CDDP MCF-7 treated cells. From the concentration range tested 100 nM CDDP triggered the highest [Ca(2+)]i increase (120%; n = 19)while in drug resistant MCF-7 cells the effects of CDDP on [Ca(2+)]i were reduced as compared with the drug sensitive MCF-7 cells. Furthermore, the CDDP induced cell death correlates with the increase of [Ca(2+)]i, and thus, significantly lower in the CDDP desensitized cells (p < 0.05). Pre-application of the calcium channel blocker, Nimodipine reduced [Ca(2+)]i elevation significantly (46.6% increase; n = 26) as well as when a pre-application of Caffeine, Ionomycin or Thapsigargin occurred followed by the subsequent application of CDDP (n = 15; 37.8%, n = 32; 34.9%, n = 21; 53.7% increase respectively).


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Cisplatino/farmacologia , Homeostase/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Morte Celular/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Nimodipina/farmacologia
15.
Mol Cancer Ther ; 13(4): 948-61, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24526161

RESUMO

Glioblastoma is the most common malignant brain tumor in adults and characterized by a poor prognosis. Glioma cells expressing O(6)-methylguanine DNA methyltransferase (MGMT) exhibit a higher level of resistance toward alkylating agents, including the standard of care chemotherapeutic agent temozolomide. Here, we demonstrate that long-term glioma cell lines (LTL) as well as glioma-initiating cell lines (GIC) express receptors for the immune modulatory cytokine IFN-ß and respond to IFN-ß with induction of STAT-3 phosphorylation. Exposure to IFN-ß induces a minor loss of viability, but strongly interferes with sphere formation in GIC cultures. Furthermore, IFN-ß sensitizes LTL and GIC to temozolomide and irradiation. RNA interference confirmed that both IFN-ß receptors, R1 and R2, are required for IFN-ß-mediated sensitization, but that sensitization is independent of MGMT or TP53. Most GIC lines are highly temozolomide-resistant, mediated by MGMT expression, but nevertheless susceptible to IFN-ß sensitization. Gene expression profiling following IFN-ß treatment revealed strong upregulation of IFN-ß-associated genes, including a proapoptotic gene cluster, but did not alter stemness-associated expression signatures. Caspase activity and inhibition studies revealed the proapoptotic genes to mediate glioma cell sensitization to exogenous death ligands by IFN-ß, but not to temozolomide or irradiation, indicating distinct pathways of death sensitization mediated by IFN-ß. Thus, IFN-ß is a potential adjunct to glioblastoma treatment that may target the GIC population. IFN-ß operates independently of MGMT-mediated resistance, classical apoptosis-regulatory networks, and stemness-associated gene clusters.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Interferon beta/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Receptor de Interferon alfa e beta/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , Células K562 , Células MCF-7 , Dados de Sequência Molecular , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/efeitos da radiação , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Temozolomida
16.
Aging (Albany NY) ; 6(10): 856-78, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25411231

RESUMO

We analyzed an ex vivo model of in situ aged human dermal fibroblasts, obtained from 15 adult healthy donors from three different age groups using an unbiased quantitative proteome-wide approach applying label-free mass spectrometry. Thereby, we identified 2409 proteins, including 43 proteins with an age-associated abundance change. Most of the differentially abundant proteins have not been described in the context of fibroblasts' aging before, but the deduced biological processes confirmed known hallmarks of aging and led to a consistent picture of eight biological categories involved in fibroblast aging, namely proteostasis, cell cycle and proliferation, development and differentiation, cell death, cell organization and cytoskeleton, response to stress, cell communication and signal transduction, as well as RNA metabolism and translation. The exhaustive analysis of protein and mRNA data revealed that 77 % of the age-associated proteins were not linked to expression changes of the corresponding transcripts. This is in line with an associated miRNA study and led us to the conclusion that most of the age-associated alterations detected at the proteome level are likely caused post-transcriptionally rather than by differential gene expression. In summary, our findings led to the characterization of novel proteins potentially associated with fibroblast aging and revealed that primary cultures of in situ aged fibroblasts are characterized by moderate age-related proteomic changes comprising the multifactorial process of aging.


Assuntos
Envelhecimento/fisiologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Transcriptoma , Adulto , Idoso , Células Cultivadas , Feminino , Humanos , Técnicas In Vitro , Espectrometria de Massas , MicroRNAs , Pessoa de Meia-Idade , Fenótipo , Proteômica , Adulto Jovem
17.
Biomed Res Int ; 2013: 486072, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24093099

RESUMO

Exposure of cells and organisms to stressors might result in epigenetic changes. Here it is shown that investigation of DNA methylation using pyrosequencing is an alternative for in vitro and in vivo toxicological testing of epigenetic effects induced by chemicals and drugs. An in vitro evaluation of global and CpG site specific DNA methylation upon treatment of cells with chemicals/drugs is shown. Bisulfite genomic sequencing of methylation controls showed high methylation of LINE1 in methylation positive control and low methylation in the negative controls. The CpG sites within the LINE1 element are methylated at different levels. In vitro cell cultures show a methylation level ranging from 56% to 49%. Cultures of drug resistant tumor cells show significant hypomethylation as compared with the originating nonresistant tumor cells. The in vitro testing of epigenetically active chemicals (5-methyl-2'-deoxycytidine and trichostatin A) revealed a significant change of LINE1 methylation status upon treatment, while specific CpG sites were more prone to demethylation than others (focal methylation). In conclusion, DNA methylation using pyrosequencing might be used not only for testing epigenetic toxins/drugs but also in risk assessment of drugs, food, and environmental relevant pollutants.


Assuntos
Poluentes Atmosféricos , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala , Linhagem Celular Tumoral , Ilhas de CpG/efeitos dos fármacos , Metilação de DNA/genética , Desoxicitidina/análogos & derivados , Desoxicitidina/toxicidade , Epigênese Genética/genética , Humanos , Ácidos Hidroxâmicos/toxicidade , Elementos Nucleotídeos Longos e Dispersos/efeitos dos fármacos , Elementos Nucleotídeos Longos e Dispersos/genética , Regiões Promotoras Genéticas/efeitos dos fármacos
18.
J Toxicol ; 2011: 159619, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21804822

RESUMO

Metals and metal compounds are part of our environment. Several metals are essential for physiological functions (e.g., zinc or magnesium); while the beneficial effects of others are uncertain (e.g., manganese), some metals are proven to be toxic (e.g., mercury, lead). Additionally there are organic metal compounds; some of them are extremely toxic (e.g., trimethyltin, methylmercury), but there is very little knowledge available how they are handled by organisms. Scientific evidence indicates that long-term exposure to (some) metallic compounds induces different forms of cancer, including breast cancer. On the other side, several metal compounds have clinical use in treating life-threatening diseases such as cancer. In this paper we discuss the recent literature that shows a correlation between metal exposure and breast cancer.

19.
Cancers (Basel) ; 3(1): 1351-71, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24212665

RESUMO

Platinum complexes are clinically used as adjuvant therapy of cancers aiming to induce tumor cell death. Depending on cell type and concentration, cisplatin induces cytotoxicity, e.g., by interference with transcription and/or DNA replication mechanisms. Additionally, cisplatin damages tumors via induction of apoptosis, mediated by the activation of various signal transduction pathways, including calcium signaling, death receptor signaling, and the activation of mitochondrial pathways. Unfortunately, neither cytotoxicity nor apoptosis are exclusively induced in cancer cells, thus, cisplatin might also lead to diverse side-effects such as neuro- and/or renal-toxicity or bone marrow-suppression. Moreover, the binding of cisplatin to proteins and enzymes may modulate its biochemical mechanism of action. While a combination-chemotherapy with cisplatin is a cornerstone for the treatment of multiple cancers, the challenge is that cancer cells could become cisplatin-resistant. Numerous mechanisms of cisplatin resistance were described including changes in cellular uptake, drug efflux, increased detoxification, inhibition of apoptosis and increased DNA repair. To minimize cisplatin resistance, combinatorial therapies were developed and have proven more effective to defeat cancers. Thus, understanding of the biochemical mechanisms triggered by cisplatin in tumor cells may lead to the design of more efficient platinum derivates (or other drugs) and might provide new therapeutic strategies and reduce side effects.

20.
Neurotoxicology ; 30(5): 803-10, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19465052

RESUMO

(Neuro-)toxicity of metal and metal compounds is frequently highlighted. While specific metals or metal compounds are essential for cellular function, other metals are toxic and/or carcinogens. Metals can trigger accidental cell death in the form of necrosis, or activate programmed cell death in the form of apoptosis. The aim of anti-cancer therapy is induction of apoptosis in tumor cells. Therefore, there is an interesting twist in the toxicity of metals and metal compounds (e.g., arsenic trioxide, cisplatin); since they have a higher specificity to induce apoptosis in cancer cells (possibly due to the high turnover in these cells) they are used to cure some forms of cancer. A body of evidence suggests that second messengers, such as modulations in the intracellular calcium concentration, could be involved in metals induced toxicity as well as in the beneficial effects shown by anti-cancer drugs. Here we review the influence on calcium homeostasis induced by some metallic compounds: cisplatin, arsenic trioxide and trimethyltin chloride.


Assuntos
Anticarcinógenos/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Líquido Extracelular/efeitos dos fármacos , Neoplasias/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinógenos Ambientais/toxicidade , Humanos , Metais/antagonistas & inibidores , Metais/intoxicação , Modelos Biológicos , Neoplasias/induzido quimicamente , Neoplasias/tratamento farmacológico , Neoplasias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA