Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 14(9): e2000314, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27684370

RESUMO

Cancer research has become a global enterprise, and the number of researchers, as well as the cost for their activities, has skyrocketed. The budget for the National Cancer Institute of the United States National Institutes of Health alone was US$5.2 billion in 2015. Since most of the research is funded by public money, it is perfectly legitimate to ask if these large expenses are worth it. In this brief commentary, we recapitulate some of the breakthroughs that mark the history of breast cancer research over the past decades and emphasize the resulting benefits for afflicted women. In 1971, only 40% of women diagnosed with breast cancer would live another 10 years. Today, nearly 80% of women reach that significant milestone in most developed countries. This dramatic change has afforded breast cancer patients many productive years and a better quality of life. Progress resulted largely from advances in the understanding of the molecular details of the disease and their translation into innovative, rationally designed therapies. These developments are founded on the revolution in molecular and cellular biology, an entirely new array of methods and technologies, the enthusiasm, optimism, and diligence of scientists and clinicians, and the considerable funding efforts from public and private sources. We were lucky to be able to spend our productive years in a period of scientific upheaval in which methods and concepts were revolutionized and that allowed us to contribute, within the global scientific community, to the progress in basic science and clinical practice.

2.
Arch Toxicol ; 92(7): 2227-2243, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29845424

RESUMO

Novel therapies are required for the treatment of metastatic renal cell carcinoma (RCC), which is associated with inoperable disease and patient death. Histone deacetylases (HDACs) are epigenetic modifiers and potential drug targets. Additional information on molecular pathways that are altered by histone deacetylase inhibitors (HDACi) in RCC cells is warranted. It should equally be delineated further which individual members of the 18 mammalian HDACs determine the survival and tumor-associated gene expression programs of such cells. Most importantly, an ongoing dispute whether HDACi promote or suppress metastasis-associated epithelial-to-mesenchymal transition (EMT) has to be resolved before HDACi are considered further as clinically relevant drugs. Here we show how HDACi affect murine and primary human RCC cells. We find that these agents induce morphological alterations resembling the metastasis-associated EMT. However, individual and proteomics-based analyses of epithelial and mesenchymal marker proteins and of EMT-associated transcription factors (EMT-TFs) reveal that HDACi do not trigger EMT. Pathway deconvolution analysis identifies reduced proliferation and apoptosis induction as key effects of HDACi. Furthermore, these drugs lead to a reduction of the cell adhesion molecule E-cadherin and of the platelet-derived growth factor receptor-ß (PDGFRß), which is a key driver of RCC metastasis formation. Accordingly, HDACi reduce the pulmonary spread of syngeneic transplanted renal carcinoma cells in mice. Specific genetic elimination of the histone deacetylases HDAC1/HDAC2 reflects the effects of pharmacological HDAC inhibition regarding growth suppression, apoptosis, and the downregulation of E-cadherin and PDGFRß. Thus, these epigenetic modifiers are non-redundant gatekeepers of cell fate and precise pharmacological targets.


Assuntos
Carcinoma de Células Renais/enzimologia , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 2/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Neoplasias Renais/enzimologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Camundongos Endogâmicos BALB C , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Biochim Biophys Acta ; 1849(8): 1003-16, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26070487

RESUMO

The p53 tumor suppressor plays a critical role in cancer, and more than 50% of human tumors contain mutations or deletions of the TP53 gene. p53 can transactivate or repress target genes in response to diverse stress signals, such as transient growth arrest, DNA repair, cellular differentiation, senescence and apoptosis. Through an unbiased genome-wide ChIP-seq analysis, we have found that 5-lipoxygenase (ALOX5, 5-LO) which is a key enzyme of leukotriene (LT) biosynthesis, is a direct target gene of p53 and its expression is induced by genotoxic stress via actinomycin D (Act.D) or etoposide (Eto) treatment. 5-LO and LTs play a role in immunological diseases as well as in tumorigenesis and tumor growth. p53 binds to a specific binding site consisting of a complete p53 consensus-binding motif in ALOX5 intron G which is located about 64kbp downstream of the transcriptional start site. We confirmed the strong binding of p53 to the 5-LO target site in ChIP-qPCR experiments. Expression analyses by qRT-PCR and immunoblot further revealed that genotoxic stress induces the ALOX5 mRNA and protein expression in a p53-dependent manner. Knockdown of p53 in U2OS cells leads to a downregulation of 5-LO mRNA and protein expression. In addition, immunofluorescence and immunoprecipitation assays indicate the direct binding of 5-LO to p53 protein. Furthermore, we found that 5-LO can inhibit the transcriptional activity of p53 suggesting that 5-LO acts in a negative feedback loop to limit induction of p53 target genes.


Assuntos
Araquidonato 5-Lipoxigenase/genética , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteína Supressora de Tumor p53/fisiologia , Sítios de Ligação , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Dactinomicina/farmacologia , Etoposídeo/farmacologia , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Regiões Promotoras Genéticas/efeitos dos fármacos , Células Tumorais Cultivadas
4.
Int J Cancer ; 137(10): 2296-309, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25991553

RESUMO

Cellular transformation is initiated by the activation of oncogenes and a closely associated developmental reprogramming of the epigenetic landscape. Transcription factors, regulators of chromatin states and microRNAs influence cell fates in development and stabilize the phenotypes of normal, differentiated cells and of cancer cells. The miR-302/367 cluster, predominantly expressed in human embryonic stem cells (hESs), can promote the cellular reprogramming of human and mouse cells and contribute to the generation of iPSC. We have used the epigenetic reprogramming potential of the miR-302/367 cluster to "de-program" tumor cells, that is, hift their gene expression pattern towards an alternative program associated with more benign cellular phenotypes. Induction of the miR-302/367 cluster in extensively mutated U87MG glioblastoma cells drastically suppressed the expression of transformation related proteins, for example, the reprogramming factors OCT3/4, SOX2, KLF4 and c-MYC, and the transcription factors POU3F2, SALL2 and OLIG2, required for the maintenance of glioblastoma stem-like tumor propagating cells. It also diminished PI3K/AKT and STAT3 signaling, impeded colony formation in soft agar and cell migration and suppressed pro-inflammatory cytokine secretion. At the same time, the miR-302/367 cluster restored the expression of neuronal markers of differentiation. Most notably, miR-302/367 cluster expressing cells lose their ability to form tumors and to establish liver metastasis in nude mice. The induction of the miR-302/367 cluster in U87MG glioblastoma cells suppresses the expression of multiple transformation related genes, abolishes the tumor and metastasis formation potential of these cells and can potentially become a new approach for cancer therapy.


Assuntos
Neoplasias Encefálicas/genética , Transformação Celular Neoplásica/patologia , Citocinas/metabolismo , Glioblastoma/genética , MicroRNAs/genética , Fatores de Transcrição/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Reprogramação Celular , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos Nus , Transplante de Neoplasias
5.
Anal Biochem ; 466: 83-8, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25172132

RESUMO

The presence of endotoxins in preparations of recombinantly produced therapeutic proteins poses serious problems for patients. Endotoxins can cause fever, respiratory distress syndromes, intravascular coagulation, or endotoxic shock. A number of methods have been devised to remove endotoxins from protein preparations using separation procedures based on molecular mass or charge properties. Most of the methods are limited in their endotoxin removal capacities and lack general applicability. We are describing a biotechnological approach for endotoxin removal. This strategy exploits the observation that endotoxins form micelles that expose negative charges on their surface, leading to preferential binding of endotoxins to cationic surfaces, allowing the separation from their resident protein. Endotoxins exhibit high affinity to stretches of histidines, which are widely used tools to facilitate the purification of recombinant proteins. They bind to nickel ions and are the basis for protein purification from cellular extracts by immobilized metal affinity chromatography. We show that the thrombin-mediated cleavage of two histidine tags from the purified recombinant protein and the adsorption of these histidine tags and their associated endotoxins to a nickel affinity column result in an appreciable depletion of the endotoxins in the purified protein fraction.


Assuntos
Química Farmacêutica/métodos , Endotoxinas/isolamento & purificação , Histidina/química , Proteínas Recombinantes/química , Animais , Linhagem Celular , Células Cultivadas , Química Farmacêutica/normas , Química Farmacêutica/tendências , Histidina/metabolismo , Humanos , Lipopolissacarídeos/isolamento & purificação , Camundongos , Ligação Proteica , Proteínas Recombinantes/metabolismo
6.
Stem Cells ; 28(5): 928-38, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20235097

RESUMO

The mammary gland represents a unique model system to study gene functions in adult stem cells. Mammary stem cells (MaSCs) can regenerate a functional epithelium on transplantation into cleared fat pads. We studied the consequences of distinct genetic modifications of MaSCs on their repopulation and differentiation ability. The reconstitution of ductal trees was used as a stem cell selection procedure and the nearly quantitative lentiviral infection efficiency of the primary mammary epithelial cells (MECs) rendered the enrichment of MaSCs before their transplantation unnecessary. The repopulation frequency of transduced MaSCs was nearly 100% in immunodeficient recipients and the resulting transgenic ducts homogeneously expressed the virally encoded fluorescent marker proteins. Transplantation of a mixture of MECs, expressing different fluorescent proteins, resulted in a distinct pattern of ductal outgrowths originating from a small number of individually transduced MaSCs. We used genetically modified MECs to define multiple functions of Stat5 during mammary gland development and differentiation. Stat5-downregulation in MaSCs did not affect primary ductal outgrowth, but impaired side branching and the emergence of mature alveolar cells from luminal progenitors during pregnancy. Conversely, the expression of a constitutively active variant of Stat5 (cS5-F) caused epithelial hyperproliferation, thickening of the ducts and precocious, functional alveoli formation in virgin mice. Expression of cS5-F also prevented involution and caused the formation of estrogen and progesterone receptor positive (ER(+)PR(+)) adenocarcinomas. The tumors expressed activated Stat5 and Stat3 and contained a small fraction of CD44(+) cells, possibly indicative of cancer stem cells.


Assuntos
Adenocarcinoma/etiologia , Adenocarcinoma/metabolismo , Linhagem da Célula/genética , Células Epiteliais/metabolismo , Neoplasias Mamárias Animais/etiologia , Neoplasias Mamárias Animais/metabolismo , Fator de Transcrição STAT5/fisiologia , Células-Tronco/metabolismo , Adenocarcinoma/patologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Células Epiteliais/citologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fator de Transcrição STAT5/genética , Células-Tronco/citologia , Células Tumorais Cultivadas
7.
Mol Cancer Res ; 6(2): 267-81, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18314488

RESUMO

Signal transduction events often involve the assembly of protein complexes dependent on modular interactions. The inappropriate assembly of modular components plays a role in oncogenic transformation and can be exploited for therapeutic purposes. Selected peptides embedded in the context of a scaffold protein can serve as competitive inhibitors of intracellular protein functions in cancer cells. Therapeutic application depends on binding specificities and affinities, as well as on the production and purification characteristics of the peptide aptamers and their delivery into cells. We carried out experiments to improve the properties of the scaffold. We found that the commonly used bacterial thioredoxin scaffold is suboptimal for therapeutic purposes because it aggregates during purification and is most likely immunogenic in humans. We compared the properties of peptide aptamers embedded in three alternative scaffold structures: a coiled-coil stem-loop structure, a dimerization domain, and human thioredoxin (hTrx). We found that only the hTrx molecule can be efficiently produced in bacteria and purified with high yield. We removed five internal cysteines of hTrx to circumvent aggregation during purification, which is a prerequisite for efficient transduction. Insertion of our previously characterized peptide aptamers [e.g., specifically binding signal transducer and activator of transcription 3 (Stat3)] into the modified hTrx scaffold retained their target binding properties. Addition of a protein transduction domain, consisting of nine arginines, results in a fusion protein, which is taken up by cultured cells. We show that treatment of glioblastoma cells, expressing constitutively activated Stat3, with the purified peptide aptamers strongly inhibits Stat3 signaling, causing cell growth arrest and inducing apoptosis.


Assuntos
Aptâmeros de Peptídeos/metabolismo , Espaço Intracelular/metabolismo , Proteínas Recombinantes/metabolismo , Sequência de Aminoácidos , Animais , Apoptose , Aptâmeros de Peptídeos/química , Aptâmeros de Peptídeos/isolamento & purificação , Sequência de Bases , Linhagem Celular Tumoral , Proliferação de Células , Cisteína , Escherichia coli , Glioblastoma/patologia , Humanos , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Estrutura Quaternária de Proteína , Proteínas Recombinantes/isolamento & purificação , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Tiorredoxinas/química , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Transdução Genética
8.
Breast Cancer Res ; 10(6): R94, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19014541

RESUMO

INTRODUCTION: The prolactin-Janus-kinase-2-signal transducer and activator of transcription-5 (JAK2-STAT5) pathway is essential for the development and functional differentiation of the mammary gland. The pathway also has important roles in mammary tumourigenesis. Prolactin regulated target genes are not yet well defined in tumour cells, and we undertook, to the best of our knowledge, the first large genetic screen of breast cancer cells treated with or without exogenous prolactin. We hypothesise that the identification of these genes should yield insights into the mechanisms by which prolactin participates in cancer formation or progression, and possibly how it regulates normal mammary gland development. METHODS: We used subtractive hybridisation to identify a number of prolactin-regulated genes in the human mammary carcinoma cell line SKBR3. Northern blotting analysis and luciferase assays identified the gene encoding heat shock protein 90-alpha (HSP90A) as a prolactin-JAK2-STAT5 target gene, whose function was characterised using apoptosis assays. RESULTS: We identified a number of new prolactin-regulated genes in breast cancer cells. Focusing on HSP90A, we determined that prolactin increased HSP90A mRNA in cancerous human breast SKBR3 cells and that STAT5B preferentially activated the HSP90A promoter in reporter gene assays. Both prolactin and its downstream protein effector, HSP90alpha, promote survival, as shown by apoptosis assays and by the addition of the HSP90 inhibitor, 17-allylamino-17-demethoxygeldanamycin (17-AAG), in both untransformed HC11 mammary epithelial cells and SKBR3 breast cancer cells. The constitutive expression of HSP90A, however, sensitised differentiated HC11 cells to starvation-induced wild-type p53-independent apoptosis. Interestingly, in SKBR3 breast cancer cells, HSP90alpha promoted survival in the presence of serum but appeared to have little effect during starvation. CONCLUSIONS: In addition to identifying new prolactin-regulated genes in breast cancer cells, we found that prolactin-JAK2-STAT5 induces expression of the HSP90A gene, which encodes the master chaperone of cancer. This identifies one mechanism by which prolactin contributes to breast cancer. Increased expression of HSP90A in breast cancer is correlated with increased cell survival and poor prognosis and HSP90alpha inhibitors are being tested in clinical trials as a breast cancer treatment. Our results also indicate that HSP90alpha promotes survival depending on the cellular conditions and state of cellular transformation.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas de Choque Térmico HSP90/genética , Janus Quinase 2/genética , Prolactina/farmacologia , Fator de Transcrição STAT5/genética , Apoptose/fisiologia , Benzoquinonas/farmacologia , Northern Blotting , Western Blotting , Diferenciação Celular , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Biblioteca Gênica , Humanos , Lactamas Macrocíclicas/farmacologia , Luciferases/metabolismo , Hibridização de Ácido Nucleico , RNA Mensageiro/metabolismo , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
9.
Comb Chem High Throughput Screen ; 11(2): 135-45, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18336207

RESUMO

Peptide aptamers are molecules that bind to protein targets and are able to interfere with their functions. In the past, important achievements have been made using such peptide aptamers in different approaches and for various purposes. Peptide aptamers are comprised of a variable peptide region of 8 to 20 amino acids in length, which is displayed by a scaffold protein. An overview of the numerous scaffold proteins that have been investigated for their suitability to present peptide aptamers will be given. To identify peptide aptamers efficiently and specifically binding to a predetermined target, two eukaryotic systems have been used in multiple studies: a modified version of the Gal4 yeast-two-hybrid system and the optimized LexA interaction trap system. The two yeast systems are compared and the design of high-complexity peptide aptamer libraries for these systems is described. Although the yeast-two-hybrid system is based on intracellular interactions mammalian screens, performed in cell culture experiments, are sometimes preferred or required. We will give an overview of the mammalian selection systems available, which are based on the expression of peptide aptamers in retroviral or lentiviral vectors. We will show that the isolation and use of peptide aptamers as inhibitors of individual signaling components represents a new challenge for drug development.


Assuntos
Aptâmeros de Peptídeos/genética , Biblioteca de Peptídeos , Animais , Aptâmeros de Peptídeos/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Humanos
10.
Mol Cancer Res ; 4(12): 983-98, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17189388

RESUMO

The ErbB2 receptor tyrosine kinase is overexpressed in approximately 30% of breast tumor cases and its overexpression correlates with an unfavorable prognosis. A major contributor for this course of the disease is the insensitivity of these tumors toward chemotherapy. Monoclonal antibodies, inhibiting the ligand-induced activation of the receptor and tyrosine kinase inhibitors acting on the intrinsic enzymatic activity of the intracellular domain, have been developed as targeted drugs. Both have been shown to be beneficial for breast cancer patients. We targeted a third aspect of receptor function: its association with intracellular signaling components. For this purpose, we selected peptide aptamers, which specifically interact with defined domains of the intracellular part of the receptor. The peptide aptamers were selected from a random peptide library using a yeast two-hybrid system with the intracellular tyrosine kinase domain of ErbB2 as a bait construct. The peptide aptamer AII-7 interacts with high specificity with the ErbB2 receptor in vitro and in vivo. The aptamers colocalized with the intracellular domain of ErbB2 within cells. We investigated the functional consequences of the aptamer interaction with the ErbB2 receptor within tumor cells. The aptamer sequences were either expressed intracellularly or introduced into the cells as recombinant aptamer proteins. The phosphorylation of p42/44 mitogen-activated protein kinase was nearly unaffected and the activation of signal transducers and activators of transcription-3 was only modestly reduced. In contrast, they strongly inhibited the induction of AKT kinase in MCF7 breast cancer cells treated with heregulin, whereas AKT activation downstream of insulin-like growth factor I or epidermal growth factor receptor was not or only slightly affected. High AKT activity is responsible for the enhanced resistance of ErbB2-overexpressing cancer cells toward chemotherapeutic agents. Peptide aptamer interference with AKT activation resulted in the restoration of regular sensitivity of breast cancer cells toward Taxol.


Assuntos
Aptâmeros de Peptídeos/farmacologia , Neoplasias da Mama/patologia , Paclitaxel/farmacologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Motivos de Aminoácidos , Antineoplásicos/farmacologia , Aptâmeros de Peptídeos/química , Interações Medicamentosas , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Biblioteca de Peptídeos , Conformação Proteica , Receptor ErbB-2/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
11.
Mol Endocrinol ; 20(10): 2369-81, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16772532

RESUMO

Organ development requires the integration of multiple extracellular signals to assure a proper balance between proliferation and differentiation and to achieve and maintain specialized functions. Considerable progress has been made in the study of hormones and growth factors and in the understanding of the regulated intracellular pathways and transcriptional events that contribute to mammogenesis. Cell culture experiments have pointed out crucial pathways and components, which were subsequently validated in vivo experiments. We found that the mammalian target of rapamycin (mTOR) pathway is essential for both growth and differentiation of mammary epithelial cells and that the action of mTOR is mediated through the induction of the helix-loop-helix transcriptional regulators Id1 and Id2. Pharmacological inhibition of mTOR activity in HC11 mammary epithelial cells reduced cellular proliferation and prevented the lactogenic hormone-induced expression of milk proteins. Treatment of female mice with rapamycin impaired mammary gland differentiation and milk protein synthesis. The effects of mTOR on proliferation and differentiation require the functions of the helix-loop-helix proteins Id1 and Id2. Rapamycin treatment of HC11 cells resulted in a suppression of Id1 expression and an inhibition of proliferation. This effect of rapamycin was reversed by the forced expression of Id1. Rapamycin also prevented the induction of Id2 by lactogenic hormones and milk protein gene expression. Expression of a Id2 transgene bypassed the requirement of mTOR activity for beta-casein induction. These data suggest that mTOR activity has distinguishable functions in the proliferative and the differentiated state of mammary epithelial cells: it is a prerequisite for proliferation through the induction of Id1 and for differentiation-specific gene expression through the induction of Id2. The relative strengths of these proliferation and differentiation signals reflected by the expression levels of the individual Id proteins are crucial to the functional life cycle of mammary epithelial cells and might be disturbed in tumorigenesis.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteína 1 Inibidora de Diferenciação/metabolismo , Proteína 2 Inibidora de Diferenciação/metabolismo , Glândulas Mamárias Animais/citologia , Proteínas Quinases/metabolismo , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Primers do DNA , Células Epiteliais/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Luciferases , Glândulas Mamárias Animais/metabolismo , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirolimo/toxicidade , Serina-Treonina Quinases TOR
12.
Mol Cell Endocrinol ; 451: 1-14, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28576744

RESUMO

The effects of Jak Stat signaling and the persistent activation of Stat3 and Stat5 on tumor cell survival, proliferation and invasion have made the Jak Stat pathway a favorite target for drug development and cancer therapy. This notion was strengthened when additional biological functions of Stat signaling in cancer and their roles in the regulation of cytokine dependent inflammation and immunity in the tumor microenvironment were discovered. Stats act not only as transcriptional inducers, but affect gene expression via epigenetic modifications, induce epithelial mesenchymal transition, generate a pro-tumorigenic microenvironment, promote cancer stem cell self-renewal and differentiation, and help to establish the pre-metastatic niche formation. The effects of Jak Stat inhibition on the suppression of pro-inflammatory responses appears most promising and could become a strategy in the prevention of tumor progression. The direct and mediated mechanisms of Jak Stat signaling in and on tumors cells, the interactions with other signaling pathways and transcription factors and the targeting of the functionally crucial secondary modifications of Stat molecules suggest novel approaches to the future development of Jak Stat based cancer therapeutics.


Assuntos
Antineoplásicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Imunoterapia/métodos , Janus Quinases/genética , Neoplasias/terapia , Fatores de Transcrição STAT/genética , Proliferação de Células , Transição Epitelial-Mesenquimal , Humanos , Janus Quinases/antagonistas & inibidores , Janus Quinases/imunologia , Terapia de Alvo Molecular , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição STAT/antagonistas & inibidores , Fatores de Transcrição STAT/imunologia , Transdução de Sinais , Microambiente Tumoral/efeitos dos fármacos
13.
J Innate Immun ; 8(2): 111-20, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26845587

RESUMO

Translational research and precision medicine are based on a profound knowledge of cellular and molecular mechanisms contributing to various physiologic processes and pathologic reactions in diverse organs. Whereas specific molecular interactions and mechanisms have been identified during the past 5 decades, the underlying principles were defined much earlier and originate from to the seminal observations made by outstanding researchers between 1850 and 1915. One of the most outstanding exponents of these scientists is Paul Ehrlich. His work resulted not only in the foundation and birth of modern hematology and immunology, but also led to the development of chemotherapy and specific targeted treatment concepts. In 2015, the Medical University of Vienna organized a memorial meeting, with the aim of honoring Paul Ehrlich's contributions to science, and to commemorate the 100th anniversary of his death. The authors of the current review served as faculty members and dedicate this paper to Paul Ehrlich and his remarkable contributions to medicine.


Assuntos
Pesquisa Translacional Biomédica/história , Animais , Áustria , Congressos como Assunto , História do Século XIX , História do Século XX , Humanos
14.
Mol Cancer Res ; 1(1): 32-47, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12496367

RESUMO

Signal transducer and activator of transcription 5 (Stat5) transduces extracellular cytokine and growth factor signals to the nucleus of mammary epithelial cells and thereby regulates gene transcription during pregnancy, lactation, and weaning. Gene constructs were prepared which subject the wild-type Stat5 or a constitutively active variant of Stat5 to the control of the beta-lactoglobulin (BLG) regulatory sequences and direct it to the mammary epithelium. The integrity and functionality of these constructs were confirmed through introduction into cultured mammary epithelial cells and hormone induction experiments. Expression levels and states of activity of Stat5 in mammary gland tissue were manipulated by introducing Stat5 variants as transgenes into the pronuclei of transgenic mice. The consequences of enhanced Stat5 expression and activation on the development of alveoli, their differentiated functions, and on postlactational involution were investigated. As expected, the transgenic mouse lines expressed the wild-type Stat5 construct (BLG/STAT5) and the constitutively active Stat5 variant (BLG/STAT5ca) exclusively in mammary epithelial cells during pregnancy and lactation. BLG/STAT5 mice exhibited larger alveoli at mid-pregnancy and a delayed onset of involution. Condensed alveoli, a high degree of cellular proliferation, and delayed involution were associated with STAT5ca expression. Elevated levels of beta-casein gene expression were found in BLG/STAT5 and STAT5ca transgenic mice during late pregnancy and lactation, indicating a limiting role for Stat5 under normal physiological conditions. This was accompanied by higher levels of beta-casein secretion into the milk and enhanced growth of pups. Transgenic animals expressing the BLG/STAT5ca transgene were predisposed to tumor formation in the mammary gland. This study extends the functional observations made in cultured mammary epithelial cells and in gene knockout mice. It identifies Stat5 as a multifunctional regulator of mammary cell proliferation, milk protein gene expression, and postlactational apoptosis.


Assuntos
Apoptose , Caseínas/genética , Proteínas de Ligação a DNA/metabolismo , Lactação , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/fisiologia , Proteínas do Leite , Transativadores/metabolismo , Animais , Sítios de Ligação , Caseínas/biossíntese , Caseínas/metabolismo , Diferenciação Celular , Divisão Celular , Células Cultivadas , Células Epiteliais , Feminino , Regulação da Expressão Gênica , Genes Reporter , Variação Genética , Camundongos , Camundongos Transgênicos , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT5 , Transgenes
15.
Mol Cancer Res ; 2(3): 170-82, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15037656

RESUMO

The transcription factor signal transducer and activator of transcription (Stat) 3 is activated through the interleukin-6 family of cytokines and by binding of growth factors to the epidermal growth factor (EGF) receptor. It plays an essential role in embryonic development and assumes specialized tasks in many differentiated tissues. Constitutively activated Stat3 has been found in tumor cell lines and primary tumors and plays a crucial role in tumor cell survival and proliferation. To inhibit the oncogenic action of Stat3 in tumor cells, we have selected short peptides, so-called peptide aptamers, which specifically interact with defined functional domains of this transcription factor. The peptide aptamers were selected from a peptide library of high complexity by an adaptation of the yeast two-hybrid procedure. Peptide aptamers specifically interacting with the Stat3 dimerization domain caused inhibition of DNA binding activity and suppression of transactivation by Stat3 in EGF-responsive cells. Similarly, a peptide aptamer selected for its ability to recognize the Stat3 DNA binding domain inhibited DNA binding and transactivation by Stat3 following EGF stimulation of cells. Peptide aptamers were expressed in bacteria as fusion proteins with a protein transduction domain and introduced into human myeloma cells. This resulted in dose-dependent growth inhibition, down-regulation of Bcl-x(L) expression, and induction of apoptosis. The inhibition of Stat3 functions through the interaction with peptide aptamers counteracts the transformed phenotype and could become useful in targeted tumor therapy.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Neoplasias/patologia , Peptídeos/metabolismo , Peptídeos/farmacologia , Transativadores/química , Transativadores/metabolismo , Ativação Transcricional/efeitos dos fármacos , Animais , Caspase 3 , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dimerização , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Biblioteca de Peptídeos , Peptídeos/química , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fator de Transcrição STAT3 , Transdução de Sinais/efeitos dos fármacos , Proteína bcl-X
16.
Artigo em Inglês | MEDLINE | ID: mdl-16375697

RESUMO

STAT5 belongs to a small family of transcription factors with dual functions. The seven signal transducers and activators of transcription (STAT) act as signaling components between the plasma membrane and the nucleus, and as transcription factors with specific DNA binding ability in the nucleus. STAT5 regulates the expression of genes, which determine important cellular phenotypes. It can promote proliferation and inhibit apoptosis, but is also involved in the regulation of differentiation between specific gene expression. STAT5 can also contribute to the transformed phenotype. In many leukemias and some solid tumors, STAT5 is constitutively activated through receptors or receptor associated tyrosine kinases and contributes to the survival and the proliferation of malignant cells. STAT5 activity appears to be limiting for these phenotypes. Inhibition of STAT5 in these tumor cells results in growth arrest and apoptosis. Targeting of STATs and other downstream mediators of oncogenic tyrosine kinases provides a promising strategy for tumor therapy, which might be refractory to resistance mechanisms incapacitating tyrosine kinase inhibitors. The well-studied steps in the activation of STAT5 and its roles in different subcellular compartments suggest original interference strategies, which could be used to inhibit its function. The challenge for drug developers will be the exploitation of defined protein-protein or protein-DNA interactions as targets of inhibition.


Assuntos
Neoplasias , Fator de Transcrição STAT5 , Linfócitos T/metabolismo , Animais , Apoptose , Citocinas/metabolismo , Humanos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Fator de Transcrição STAT5/química , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Linfócitos T/imunologia , Células Tumorais Cultivadas
17.
Curr Mol Med ; 4(5): 539-47, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15267225

RESUMO

Monoclonal antibodies had the lure of drugs very much since their first description. The ability to bind to a predetermined chemical structure stimulated the imagination of drug discoverers and developers. Nevertheless it took many years before a drug was registered which started to make good on the promise. The complexity of the molecule, made up of four polypeptide chains, its large molecular weight, its multiple and versatile functional domains and its mouse origin initially were obstacles for the production and the utilisation. Also the selection of appropriate target structures on the surface of cells turned out be difficult. Many of these difficulties have been overcome. The replacement of most of the murine sequences with equivalent human sequences and the concomittant decrease in immunogenicity, and the identification of cell surface components which are causative and limiting in cellular transformation have made monoclonal antibodies valuable weapons in the fight against cancer. Multiple mechanisms of monoclonal antibody action are being exploited for this purpose. Antibodies can sequester growth factors and prevent the activation of crucial growth factor receptors. A monoclonal antibody directed against the vascular endothelial growth factor (VEGF) has been shown to be a potent neo-vascularisation inhibitor (bevacizumab). An antibody against the extracellular domain of the EGF receptor prevents the binding of the ligand to the receptor and thereby its activation (cetuximab). EGFR activity, however, is absolutely required for the survival and proliferation of certain human tumour cells. An antibody which interferes with the dimerisation of the ErbB2 and the ErbB3 members of the EGF receptor family prevents the association of a most potent signaling module (pertuxumab). The signals emenating from this dimer determine many phenotypic properties of e.g. human breast cancer cells. A monoclonal antibody also directed against ErbB2 has been most successful, clinically and commercially (trastuzumab). This antibody interferes with signals generated by the receptor and causes the arrest of the cell cycle in tumour cells. In addition, it recruits immune effector cells as cytotoxic agents. Finally, monoclonal antibody derivatives, single chain Fv fragments, have been used as a basis for the construction of recombinant tumour toxins. These molecules harness the exquisite binding specificity of the antibodies and combine them with the toxic principles of bacteria.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Região Variável de Imunoglobulina/uso terapêutico , Imunotoxinas/uso terapêutico , Neoplasias/terapia , Animais , Anticorpos Monoclonais/imunologia , Receptores ErbB/imunologia , Genes erbB-2/imunologia , Humanos , Região Variável de Imunoglobulina/imunologia , Camundongos , Neoplasias/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia
18.
Cancers (Basel) ; 7(1): 503-37, 2015 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-25809097

RESUMO

Signal transducers and activators of transcription (Stats) play central roles in the conversion of extracellular signals, e.g., cytokines, hormones and growth factors, into tissue and cell type specific gene expression patterns. In normal cells, their signaling potential is strictly limited in extent and duration. The persistent activation of Stat3 or Stat5 is found in many human tumor cells and contributes to their growth and survival. Stat5 activation plays a pivotal role in nearly all hematological malignancies and occurs downstream of oncogenic kinases, e.g., Bcr-Abl in chronic myeloid leukemias (CML) and Jak2(V617F) in other myeloproliferative diseases (MPD). We defined the mechanisms through which Stat5 affects growth and survival of K562 cells, representative of Bcr-Abl positive CML, and HEL cells, representative for Jak2(V617F) positive acute erythroid leukemia. In our experiments we suppressed the protein expression levels of Stat5a and Stat5b through shRNA mediated downregulation and demonstrated the dependence of cell survival on the presence of Stat5. Alternatively, we interfered with the functional capacities of the Stat5 protein through the interaction with a Stat5 specific peptide ligand. This ligand is a Stat5 specific peptide aptamer construct which comprises a 12mer peptide integrated into a modified thioredoxin scaffold, S5-DBD-PA. The peptide sequence specifically recognizes the DNA binding domain (DBD) of Stat5. Complex formation of S5-DBD-PA with Stat5 causes a strong reduction of P-Stat5 in the nuclear fraction of Bcr-Abl-transformed K562 cells and a suppression of Stat5 target genes. Distinct Stat5 mediated survival mechanisms were detected in K562 and Jak2(V617F)-transformed HEL cells. Stat5 is activated in the nuclear and cytosolic compartments of K562 cells and the S5-DBD-PA inhibitor most likely affects the viability of Bcr-Abl+ K562 cells through the inhibition of canonical Stat5 induced target gene transcription. In HEL cells, Stat5 is predominantly present in the cytoplasm and the survival of the Jak2(V617F)+ HEL cells is impeded through the inhibition of the cytoplasmic functions of Stat5.

19.
Cancer Res ; 75(15): 3147-54, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26239478

RESUMO

iRGD is a derivative of the integrin-binding peptide RGD, which selectively increases the penetrability of tumor tissue to various coadministered substances in several preclinical models. In this study, we investigated the ability of iRGD to improve the delivery of sorafenib and doxorubicin therapy in hepatocellular carcinoma (HCC) using established mouse models of the disease. A contrast-enhanced MRI method was developed in parallel to assess the in vivo effects of iRGD in this setting. We found that iRGD improved the delivery of marker substances to the tumors of HCC-bearing mice about three-fold without a parallel increase in normal tissues. Control peptides lacking the critical CendR motif had no effect. Similarly, iRGD also selectively increased the signal intensity from tumors in Gd-DTPA-enhanced MRI. In terms of antitumor efficacy, iRGD coadministration significantly augmented the individual inhibitory effects of sorafenib and doxorubicin without increasing systemic toxicity. Overall, our results offered a preclinical proof of concept for the use of iRGD coadministration as a strategy to widen the therapeutic window for HCC chemotherapy, as monitored by Gd-DTPA-enhanced MRI as a noninvasive, clinically applicable method to identify iRGD-reactive tumors.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Doxorrubicina/administração & dosagem , Neoplasias Hepáticas/tratamento farmacológico , Niacinamida/análogos & derivados , Oligopeptídeos/administração & dosagem , Compostos de Fenilureia/administração & dosagem , Administração Intravenosa , Motivos de Aminoácidos , Animais , Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Doxorrubicina/farmacocinética , Sistemas de Liberação de Medicamentos , Azul Evans/administração & dosagem , Gadolínio DTPA , Células Hep G2/efeitos dos fármacos , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos Nus , Camundongos Transgênicos , Niacinamida/administração & dosagem , Oligopeptídeos/química , Sorafenibe , Distribuição Tecidual
20.
J Cancer Res Clin Oncol ; 129(12): 669-75, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-13680379

RESUMO

The major aim of molecular cancer research is the development of new therapeutic strategies and compounds that target directly the genetic and biochemical causes of malignant transformation. Therapeutic genes, antibodies and their derivatives, but also small molecular weight compounds, have been used for this purpose. Small peptides might be able to complement these agents because of their ability to recognize specific protein domains and thus to interfere with enzymatic functions or protein-protein interactions. A variation of the yeast-two-hybrid procedure allows to select specifically binding peptides, so called peptide aptamers, from a peptide library of high complexity. This selection procedure can be adapted to any protein or protein fragment as a bait construct and selects for the intracellular interaction between the bait of choice and the peptide aptamer prey. Peptide aptamers thus selected can be cloned, provided with a protein transduction domain, expressed in bacteria and introduced into cancer cells. There they might disrupt protein-protein interactions crucial for cancer cell growth or survival. We introduce an example in which the Stat3 arm of EGF receptor signaling is selectively inhibited by a peptide aptamer and causes the growth arrest of EGF receptor-dependent tumor cells. The aptamer constructs can be supplemented with additional functional domains to enhance their inhibitory effects.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Substâncias de Crescimento/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transativadores/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Antineoplásicos/síntese química , Receptores ErbB/antagonistas & inibidores , Humanos , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Biossíntese Peptídica , Proteínas Recombinantes/uso terapêutico , Fator de Transcrição STAT3
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA