Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nanomedicine ; 36: 102403, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33932594

RESUMO

Therapeutic gene silencing by RNA interference relies on the safe and efficient in vivo delivery of small interfering RNAs (siRNAs). Polyethylenimines are among the most studied cationic polymers for gene delivery. For several reasons including superior tolerability, small linear PEIs would be preferable over branched PEIs, but they show poor siRNA complexation. Their chemical modification for siRNA formulation has not been extensively explored so far. We generated a set of small linear PEIs bearing tyrosine modifications (LPxY), leading to substantially enhanced siRNA delivery and knockdown efficacy in vitro in various cell lines, including hard-to-transfect cells. The tyrosine-modified linear 10 kDa PEI (LP10Y) is particularly powerful, associated with favorable physicochemical properties and very high biocompatibility. Systemically administered LP10Y/siRNA complexes reveal antitumor effects in mouse xenograft and patient-derived xenograft (PDX) models, and their direct application into the brain achieves therapeutic inhibition of orthotopic glioma xenografts. LP10Y is particularly interesting for therapeutic siRNA delivery.


Assuntos
Terapia Genética , Neoplasias Experimentais , Polietilenoimina , RNA Interferente Pequeno , Transfecção , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Polietilenoimina/química , Polietilenoimina/farmacologia , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Int J Mol Sci ; 22(1)2020 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-33374770

RESUMO

MET-amplified gastric cancer cells are extremely sensitive to MET inhibition in vitro, whereas clinical efficacy of MET inhibitors is disappointing. The compensatory activation of other oncogenic growth factor receptors may serve as an underlying mechanism of resistance. In this study, we analyzed the role of HER receptors, in particular HER3 and its ligand heregulin, in this respect. This also included the chromatin-organizer protein SATB1, as an established regulator of HER expression in other tumor entities. In a panel of MET-amplified gastric carcinoma cell lines, cell growth under anchorage-dependent and independent conditions was studied upon inhibitor treatment or siRNA-mediated knockdown. Expression analyses were performed using RT-qPCR, FACS, and immunoblots. Signal transduction was monitored via antibody arrays and immunoblots. As expected, MET inhibition led to a growth arrest and inhibition of MAPK signaling. Strikingly, however, this was accompanied by a rapid and profound upregulation of the oncogenic receptor HER3. This finding was determined as functionally relevant, since HER3 activation by HRG led to partial MET inhibitor resistance, and MAPK/Akt signaling was even found enhanced upon HRG+MET inhibitor treatment compared to HRG alone. SATB1 was identified as mediator of HER3 upregulation. Concomitantly, SATB1 knockdown prevented upregulation of HER3, thus abrogating the HRG-promoted rescue from MET inhibition. Taken together, our results introduce the combined HER3/MET inhibition as strategy to overcome resistance towards MET inhibitors.


Assuntos
Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Receptor ErbB-3/genética , Neoplasias Gástricas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases , Proteínas de Ligação à Região de Interação com a Matriz/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Pirazinas/farmacologia , Receptor ErbB-3/metabolismo , Neoplasias Gástricas/genética , Triazóis/farmacologia , Regulação para Cima
3.
Artigo em Inglês | MEDLINE | ID: mdl-23532495

RESUMO

Acute toxic lung injury by reactive inhalational compounds is an important and still unresolved medical problem. Hazardous gases or vapors, e. g. chlorine, phosgene, sulfur mustard or methyl isocyanate, are released during occupational accidents or combustion processes and also represent a potential threat in terroristic scenarios. According to their broad-range chemical reactivity, the mechanism of lung injury evoked by these agents has long been described as rather unspecific. Consequently, therapeutic options are still restricted to symptomatic treatment. However, in recent years, ion channels of the transient receptor potential (TRP) family have been identified to act as specific sensor molecules expressed in the respiratory tract and to engage defined signaling pathways upon inhalational exposure to toxic challenges. These pulmonary receptor molecules have been primarily characterized in sensory neurons of the lung. However, chemosensory molecules are also expressed in non-neuronal cells, e.g. in the lung epithelium as well as in the pulmonary vasculature. Thus, activation of respiratory chemosensors by toxic inhalants promotes a complex signaling network directly or indirectly regulating pulmonary blood flow, the integrity of the epithelial lining, and the mucociliary clearance of the bronchial system. This review gives a synopsis on reactive lung-toxic agents and their specific target molecules in the lung and summarizes the current knowledge about the pathophysiological role of chemosensory signaling in neuronal and non-neuronal cells in toxic lung injury. Finally, we describe possible future strategies for a causal, specifically tailored treatment option based on the mechanistic understanding of molecular events ensuing inhalation of lung-toxic agents.


Assuntos
Lesão Pulmonar/induzido quimicamente , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Tosse/fisiopatologia , Humanos , Irritantes/toxicidade , Lesão Pulmonar/terapia , Depuração Mucociliar , Canais de Potencial de Receptor Transitório/química
4.
Arch Toxicol ; 89(9): 1631-43, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25395009

RESUMO

The transient receptor potential ankyrin 1 (TRPA1) cation channel is expressed in different tissues including skin, lung and neuronal tissue. Recent reports identified TRPA1 as a sensor for noxious substances, implicating a functional role in the molecular toxicology. TRPA1 is activated by various potentially harmful electrophilic substances. The chemical warfare agent sulfur mustard (SM) is a highly reactive alkylating agent that binds to numerous biological targets. Although SM is known for almost 200 years, detailed knowledge about the pathophysiology resulting from exposure is lacking. A specific therapy is not available. In this study, we investigated whether the alkylating agent 2-chloroethyl-ethylsulfide (CEES, a model substance for SM-promoted effects) and SM are able to activate TRPA1 channels. CEES induced a marked increase in the intracellular calcium concentration ([Ca(2+)]i) in TRPA1-expressing but not in TRPA1-negative cells. The TRP-channel blocker AP18 diminished the CEES-induced calcium influx. HEK293 cells permanently expressing TRPA1 were more sensitive toward cytotoxic effects of CEES compared with wild-type cells. At low CEES concentrations, CEES-induced cytotoxicity was prevented by AP18. Proof-of-concept experiments using SM resulted in a pronounced increase in [Ca(2+)]i in HEK293-A1-E cells. Human A549 lung epithelial cells, which express TRPA1 endogenously, reacted with a transient calcium influx in response to CEES exposure. The CEES-dependent calcium response was diminished by AP18. In summary, our results demonstrate that alkylating agents are able to activate TRPA1. Inhibition of TRPA1 counteracted cellular toxicity and could thus represent a feasible approach to mitigate SM-induced cell damage.


Assuntos
Alquilantes/toxicidade , Canais de Cálcio/efeitos dos fármacos , Substâncias para a Guerra Química/toxicidade , Gás de Mostarda/análogos & derivados , Proteínas do Tecido Nervoso/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/efeitos dos fármacos , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células HEK293 , Humanos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Gás de Mostarda/toxicidade , Proteínas do Tecido Nervoso/metabolismo , Oximas/farmacologia , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/metabolismo
5.
Biochem Pharmacol ; 225: 116257, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38705532

RESUMO

Gastric cancer remains among the deadliest neoplasms worldwide, with limited therapeutic options. Since efficacies of targeted therapies are unsatisfactory, drugs with broader mechanisms of action rather than a single oncogene inhibition are needed. Preclinical studies have identified histone deacetylases (HDAC) as potential therapeutic targets in gastric cancer. However, the mechanism(s) of action of HDAC inhibitors (HDACi) are only partially understood. This is particularly true with regard to ferroptosis as an emerging concept of cell death. In a panel of gastric cancer cell lines with different molecular characteristics, tumor cell inhibitory effects of different HDACi were studied. Lipid peroxidation levels were measured and proteome analysis was performed for the in-depth characterization of molecular alterations upon HDAC inhibition. HDACi effects on important ferroptosis genes were validated on the mRNA and protein level. Upon HDACi treatment, lipid peroxidation was found increased in all cell lines. Class I HDACi (VK1, entinostat) showed the same toxicity profile as the pan-HDACi vorinostat. Proteome analysis revealed significant and concordant alterations in the expression of proteins related to ferroptosis induction. Key enzymes like ACSL4, POR or SLC7A11 showed distinct alterations in their expression patterns, providing an explanation for the increased lipid peroxidation. Results were also confirmed in primary human gastric cancer tissue cultures as a relevant ex vivo model. We identify the induction of ferroptosis as new mechanism of action of class I HDACi in gastric cancer. Notably, these findings were independent of the genetic background of the cell lines, thus introducing HDAC inhibition as a more general therapeutic principle.


Assuntos
Ferroptose , Inibidores de Histona Desacetilases , Peroxidação de Lipídeos , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Ferroptose/efeitos dos fármacos , Ferroptose/fisiologia , Peroxidação de Lipídeos/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Linhagem Celular Tumoral , Sistema y+ de Transporte de Aminoácidos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Coenzima A Ligases/metabolismo , Coenzima A Ligases/genética , Coenzima A Ligases/antagonistas & inibidores , Relação Dose-Resposta a Droga
6.
ChemMedChem ; 17(9): e202100755, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35073610

RESUMO

Herein we report the structure-activity and structure-physicochemical property relationships of a series of class I selective ortho-aminoanilides targeting the "foot-pocket" in HDAC1&2. To balance the structural benefits and the physicochemical disadvantages of these substances, we started with a set of HDACi related to tacedinaline (CI-994) and evaluated their solubility, lipophilicity (log D7.4 ) and inhibition of selected HDAC isoforms. Subsequently, we selected the most promising "capless" HDACi and transferred its ZBG to our previously published scaffold featuring a peptoid-based cap group. The resulting hit compound 10 c (LSH-A54) showed favorable physicochemical properties and is a potent, selective HDAC1/2 inhibitor. The following evaluation of its slow binding properties revealed that LSH-A54 binds tightly to HDAC1 in an induced-fit mechanism. The potent HDAC1/2 inhibitory properties were reflected by attenuated cell migration in a modified wound healing assay and reduced cell viability in a clonogenic survival assay in selected breast cancer cell lines.


Assuntos
Inibidores de Histona Desacetilases , Peptoides , Histona Desacetilase 1 , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Peptoides/química
7.
Gastroenterology ; 138(3): 1189-99.e1-2, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19900447

RESUMO

BACKGROUND & AIMS: Induction of immediate early transcription factors (ITF) represents the first transcriptional program controlling mitogen-stimulated cell cycle progression in cancer. Here, we examined the transcriptional mechanisms regulating the ITF protein c-Myc and its role in pancreatic cancer growth in vitro and in vivo. METHODS: Expression of ITF proteins was examined by reverse-transcription polymerase chain reaction and immunoblotting, and its implications in cell cycle progression and growth was determined by flow cytometry and [(3)H]-thymidine incorporation. Intracellular Ca(2+) concentrations, calcineurin activity, and cellular nuclear factor of activated T cells (NFAT) distribution were analyzed. Transcription factor complex formations and promoter regulation were examined by immunoprecipitations, reporter gene assays, and chromatin immunoprecipitation. Using a combination of RNA interference knockdown technology and xenograft models, we analyzed the significance for pancreatic cancer tumor growth. RESULTS: Serum promotes pancreatic cancer growth through induction of the proproliferative NFAT/c-Myc axis. Mechanistically, serum increases intracellular Ca(2+) concentrations and activates the calcineurin/NFAT pathway to induce c-Myc transcription. NFAT binds to a serum responsive element within the proximal promoter, initiates p300-dependent histone acetylation, and creates a local chromatin structure permissive for the inducible recruitment of Ets-like gene (ELK)-1, a protein required for maximal activation of the c-Myc promoter. The functional significance of this novel pathway was emphasized by impaired c-Myc expression, G1 arrest, and reduced tumor growth upon NFAT depletion in vitro and in vivo. CONCLUSIONS: Our study uncovers a novel mechanism regulating cell growth and identifies the NFAT/ELK complex as modulators of early stages of mitogen-stimulated proliferation in pancreatic cancer cells.


Assuntos
Adenocarcinoma/metabolismo , Proliferação de Células , Montagem e Desmontagem da Cromatina , Histonas/metabolismo , Fatores de Transcrição NFATC/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Acetilação , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Sítios de Ligação , Western Blotting , Calcineurina/metabolismo , Cálcio/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Fatores de Transcrição NFATC/genética , Transplante de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Soro/metabolismo , Elemento de Resposta Sérica , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção , Proteínas Elk-1 do Domínio ets/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
8.
Cells ; 10(2)2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33572976

RESUMO

Overexpression of members of the HER/erbB transmembrane tyrosine kinase family like HER2/erbB2/neu is associated with various cancers. Some heterodimers, especially HER2/HER3 heterodimers, are particularly potent inducers of oncogenic signaling. Still, from a clinical viewpoint their inhibition has yielded only moderate success so far, despite promising data from cell cultures. This suggests acquired resistance upon inhibitor therapy as one putative issue, requiring further studies in cell culture also aiming at rational combination therapies. In this paper, we demonstrate in ovarian carcinoma cells that the RNAi-mediated single knockdown of HER2 or HER3 leads to the rapid counter-upregulation of the respective other HER family member, thus providing a rational basis for combinatorial inhibition. Concomitantly, combined knockdown of HER2/HER3 exerts stronger anti-tumor effects as compared to single inhibition. In a tumor cell line xenograft mouse model, therapeutic intervention with nanoscale complexes based on polyethylenimine (PEI) for siRNA delivery, again reveals HER3 upregulation upon HER2 single knockdown and a therapeutic benefit from combination therapy. On the mechanistic side, we demonstrate that HER2 knockdown or inhibition reduces miR-143 levels with subsequent de-repression of HER3 expression, and validates HER3 as a direct target of miR-143. HER3 knockdown or inhibition, in turn, increases HER2 expression through the upregulation of the transcriptional regulator SATB1. These counter-upregulation processes of HER family members are thus based on distinct molecular mechanisms and may provide the basis for the rational combination of inhibitors.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/antagonistas & inibidores , Regulação para Cima , Animais , Anticorpos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/metabolismo , Redes Reguladoras de Genes/efeitos dos fármacos , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cancers (Basel) ; 13(4)2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33562653

RESUMO

The increasing knowledge of molecular drivers of tumorigenesis has fueled targeted cancer therapies based on specific inhibitors. Beyond "classic" oncogene inhibitors, epigenetic therapy is an emerging field. Epigenetic alterations can occur at any time during cancer progression, altering the structure of the chromatin, the accessibility for transcription factors and thus the transcription of genes. They rely on post-translational histone modifications, particularly the acetylation of histone lysine residues, and are determined by the inverse action of histone acetyltransferases (HATs) and histone deacetylases (HDACs). Importantly, HDACs are often aberrantly overexpressed, predominantly leading to the transcriptional repression of tumor suppressor genes. Thus, histone deacetylase inhibitors (HDACis) are powerful drugs, with some already approved for certain hematological cancers. Albeit HDACis show activity in solid tumors as well, further refinement and the development of novel drugs are needed. This review describes the capability of HDACis to influence various pathways and, based on this knowledge, gives a comprehensive overview of various preclinical and clinical studies on solid tumors. A particular focus is placed on strategies for achieving higher efficacy by combination therapies, including phosphoinositide 3-kinase (PI3K)-EGFR inhibitors and hormone- or immunotherapy. This also includes new bifunctional inhibitors as well as novel approaches for HDAC degradation via PROteolysis-TArgeting Chimeras (PROTACs).

10.
Cancers (Basel) ; 13(11)2021 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-34199909

RESUMO

Chronic acid reflux causes cellular damage and inflammation in the lower esophagus. Due to these irritating insults, the squamous epithelium is replaced by metaplastic epithelium, which is a risk factor for the development of esophageal adenocarcinoma (EAC). In this study, we investigated the acid susceptibility in a Barrett's cell culture in vitro model, using six cell lines, derived from squamous epithelium (EPC1 and EPC2), metaplasia (CP-A), dysplasia (CP-B), and EAC (OE33 and OE19) cells. Cells exposed to acidic pH showed a decreased viability dependent on time, pH, and progression status in the Barrett's sequence, with the highest acid susceptibility in the squamous epithelium (EPC1 and EPC2), and the lowest in EAC cells. Acid pulsing was accompanied with an activation of the Nrf2/Keap1- and the NFκB-pathway, resulting in an increased expression of HO1-independent of the cellular context. OE33 showed a decreased responsiveness towards 5-FU, when the cells were grown in acidic conditions (pH 6 and pH 5.5). Our findings suggest a strong damage of squamous epithelium by gastroesophageal reflux, while Barrett's dysplasia and EAC cells apparently exert acid-protective features, which lead to a cellular resistance against acid reflux.

11.
J Biol Chem ; 284(39): 26411-20, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19648111

RESUMO

Melanocortin-4 receptor (MC4R)-induced anorexigenic signaling in the hypothalamus controls body weight and energy homeostasis. So far, MC4R-induced signaling has been exclusively attributed to its coupling to G(s) proteins. In line with this monogamous G protein coupling profile, most MC4R mutants isolated from obese individuals showed a reduced ability to activate G(s). However, some mutants displayed enhanced G(s) coupling, suggesting that signaling pathways independent of G(s) may be involved in MC4R-mediated anorexigenic signaling. Here we report that the G(s) signaling-deficient MC4R-D90N mutant activates G proteins in a pertussis toxin-sensitive manner, indicating that this mutant is able to selectively interact with G(i/o) proteins. Analyzing a hypothalamic cell line (GT1-7 cells), we observed activation of pertussis toxin-sensitive G proteins by the wild-type MC4R as well, reflecting multiple coupling of the MC4R to G(s) and G(i/o) proteins in an endogenous cell system. Surprisingly, the agouti-related protein, which has been classified as a MC4R antagonist, selectively activates G(i/o) signaling in GT1-7 cells. Thus, the agouti-related protein antagonizes melanocortin-dependent G(s) activation not only by competitive antagonism but additionally by initiating G(i/o) protein-induced signaling as a biased agonist.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Toxina Pertussis/farmacologia , Receptor Tipo 4 de Melanocortina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Relacionada com Agouti/genética , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Ensaio de Imunoadsorção Enzimática , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Hipotálamo/citologia , Mutação , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ligação Proteica/efeitos dos fármacos , Ensaio Radioligante , Receptor Tipo 4 de Melanocortina/genética , Transfecção , alfa-MSH/metabolismo , alfa-MSH/farmacologia
12.
J Cancer Res Clin Oncol ; 146(4): 859, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32112147

RESUMO

In the original article, the title of the article is "Restoration of MARCK enhances chemosensitivity in cancer". The authors would like to change the article title to "Restoration of MARCKS enhances chemosensitivity in cancer" by adding a letter "S" to the word MARCK.

13.
J Cancer Res Clin Oncol ; 146(4): 843-858, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32056006

RESUMO

PURPOSE: Increased ATP-binding-cassette (ABC) transporter activity is a major cause of chemotherapy resistance in cancer. The ABC transporter family member ABCB1 is often overexpressed in colorectal cancer (CRC). Phosphatidylinositol-4,5-bisphosphat (PI(4,5)P2)-dependent pathways are involved in the regulation of ABCB1 function. The protein Myristoylated Alanine-Rich C-Kinase Substrate (MARCKS) is a pivotal regulator of PI(4,5)P2 and inactivated in many CRC cancers via genetic deletion or hyperphosphorylation. Therefore, MARCKS may critically impact ABCB1. METHODS: CRC samples as well as CRC cell lines were tested for a connection between MARCKS and ABCB1 via immunofluorescence and Western-blot analysis. ABCB1 function was studied via calcein influx assay under treatment with known ABCB1 inhibitors (verapamil, tariquidar) as well as the kinase inhibitor bosutinib. ABCB1 internalization and MARCKS translocation was analyzed via confocal microscopy exploiting the endocytosis inhibitors chlorpromazine and dynasore. Abundance of PI(4,5)P2 was monitored by intramolecular fluorescence resonance energy transfer (FRET). Reproductive cell survival was studied via colorimetric WST-1 and clonogenic assays in combination with exposure to the chemotherapeutics doxorubicin and 5-fuorouracil (5-FU). RESULTS: We found increased ABCB1 expression in MARCKS negative CRC patient tumor samples and established CRC cell lines. Mechanistically, the reconstitution of MARCKS function via recombinant expression or the pharmacological inhibition of MARCKS phosphorylation led to a substantial decrease in ABCB1 activity. In CRC cells, bosutinib treatment resulted in a MARCKS translocation from the cytosol to the plasma membrane, while simultaneously, ABCB1 was relocated to intracellular compartments. Inhibition of MARCKS phosphorylation via bosutinib rendered cells more sensitive to the chemotherapeutics doxorubicin and 5-FU. CONCLUSIONS: Cells devoid of MARCKS function showed incomplete ABCB1 internalization, leading to higher ABCB1 activity enhancing chemoresistance. Vice versa our data suggest the prevention of MARCKS inhibition by reversing hyperphosphorylation or genomic restoration after deletion as two promising approaches to overcome tumor cell resistance towards chemotherapeutic ABCB1 substrates.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Substrato Quinase C Rico em Alanina Miristoilada/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Compostos de Anilina , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Fluoresceínas/metabolismo , Transferência Ressonante de Energia de Fluorescência , Células HT29 , Humanos , Microscopia Confocal , Substrato Quinase C Rico em Alanina Miristoilada/deficiência , Nitrilas , Fosforilação , Quinolinas
14.
J Control Release ; 319: 63-76, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-31866504

RESUMO

Extracellular vesicles (ECVs) are secreted cell-derived membrane particles involved in intercellular signaling and cell-cell communication. By transporting various bio-macromolecules, ECVs and in particular exosomes are relevant in various (patho-) physiological processes. ECVs are also released by cancer cells and can confer pro-tumorigenic effects. Their target cell tropism, effects on proliferation rates, natural stability in blood and immunotolerance makes ECVs particularly interesting as delivery vehicles. Polyethylenimines (PEIs) are linear or branched polymers which are capable of forming non-covalent complexes with small RNA molecules including siRNAs or antimiRs, for their delivery in vitro and in vivo. This study explores for the first time the combination of PEI-based nanoparticles with naturally occurring ECVs from different cell lines, for the delivery of small RNAs. ECV-modified PEI/siRNA complexes are analyzed by electron microscopy vs. ECV or complex alone. On the functional side, we demonstrate increased knockdown efficacy and storage stability of PEI/siRNA complexes upon their modification with ECVs. This is paralleled by enhanced tumor cell-inhibition by ECV-modified PEI/siRNA complexes targeting Survivin. Pre-treatment with various inhibitors of cellular internalization reveals alterations in cellular uptake mechanisms and biological activities of PEI/siRNA complexes upon their ECV modification. Extending our studies towards PEI-complexed antimiRs against miR-155 or miR-1246, dose-dependent cellular and molecular effects are enhanced in ECV-modified complexes, based on the de-repression of direct miRNA target genes. Differences between ECVs from different cell lines are observed regarding their capacity of enhancing PEI/siRNA efficacies, independent of the target cell line for transfection. Finally, an in vivo therapy study in mice bearing s.c. PC3 prostate carcinoma xenografts reveals marked inhibition of tumor growth upon treatment with ECVPC3-modified PEI/siSurvivin complexes, based on profound target gene knockdown. We conclude that ECV-modification enhances the activity of PEI-based complexes, by altering pivotal physicochemical and biological nanoparticle properties.


Assuntos
Vesículas Extracelulares , Polietilenoimina , Animais , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Masculino , Camundongos , RNA Interferente Pequeno , Transfecção
15.
Pharmaceuticals (Basel) ; 11(4)2018 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-30248976

RESUMO

Transient receptor potential (TRP) channels represent a large family of cation channels and many members of the TRP family have been shown to act as polymodal receptor molecules for irritative or potentially harmful substances. These chemosensory TRP channels have been extensively characterized in primary sensory and neuronal cells. However, in recent years the functional expression of these proteins in non-neuronal cells, e.g., in the epithelial lining of the respiratory tract has been confirmed. Notably, these proteins have also been described in a number of cancer types. As sensor molecules for noxious compounds, chemosensory TRP channels are involved in cell defense mechanisms and influence cell survival following exposure to toxic substances via the modulation of apoptotic signaling. Of note, a number of cytostatic drugs or drug metabolites can activate these TRP channels, which could affect the therapeutic efficacy of these cytostatics. Moreover, toxic inhalational substances with potential involvement in lung carcinogenesis are well established TRP activators. In this review, we present a synopsis of data on the expression of chemosensory TRP channels in lung cancer cells and describe TRP agonists and TRP-dependent signaling pathways with potential relevance to tumor biology. Furthermore, we discuss a possible role of TRP channels in the non-genomic, tumor-promoting effects of inhalational carcinogens such as cigarette smoke.

16.
Thromb Haemost ; 97(6): 873-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17549287

RESUMO

The role of vasa vasorum (VV) in atherosclerosis is hotly debated, and new experimental techniques have recently opened an opportunity to take a fresh look at this important topic. Although the proliferation of VV due to atherogenic stimuli is controversial, experimental and clinical evidence strongly suggest the potential of VV in vascular proliferative disorders. In the past, paradigms of atherosclerosis and restenosis have excluded the adventitia and VV in the artery wall due, in part, to a lack of i) appropriate animal models featuring adventitial VV neovascularization, ii) imaging technologies to quantitate adventitial VV and plaque neovascularization and iii) its consequences, concerning information on detectable plaque substrate in vulnerable lesions. VV proliferation is associated with increasing plaque burden and is linked to cellular processes which are critical during the development of atherosclerotic plaques such as inflammation, plaque perfusion and concomitant intraplaque hemorrhage - but the regulation and induction of VV based on pathological settings are poorly understood. This review discusses the current scientific status and its controversies and identifies open research questions.


Assuntos
Aterosclerose/fisiopatologia , Inflamação/fisiopatologia , Neovascularização Patológica/fisiopatologia , Vasa Vasorum/fisiopatologia , Proteínas Angiogênicas/metabolismo , Animais , Aterosclerose/etiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Humanos , Inflamação/complicações , Inflamação/metabolismo , Inflamação/patologia , Aneurisma Intracraniano/etiologia , Aneurisma Intracraniano/fisiopatologia , Microcirculação/fisiopatologia , Modelos Cardiovasculares , Neovascularização Patológica/complicações , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ruptura , Vasa Vasorum/metabolismo , Vasa Vasorum/patologia
17.
Eur J Radiol ; 61(2): 303-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17011152

RESUMO

PURPOSE: To present a method for calculating split renal function solely from routine triphasic helical computed tomography (CT). SUBJECTS AND METHODS: We retrospectively included 26 adult patients who received renal scintigraphy and triphasic CT within 4 weeks in the years 2003 and 2004. All scans were performed using a standard abdominal protocol. Split renal function was calculated as relative single-kidney glomerular filtration rate (GFR) using a simplified "two-point Patlak plot" technique. As a reference method, split renal function was determined from renal scintigraphy using the standard technique. RESULTS: Linear correlation between the two methods was r=0.91, split renal function (CT)=0.0266+0.9573 x split renal function (scintigraphy). CONCLUSION: Split renal function can be measured accurately by minimally extended triphasic CT.


Assuntos
Meios de Contraste/farmacocinética , Testes de Função Renal/métodos , Rim/fisiologia , Tomografia Computadorizada Espiral , Adulto , Idoso , Feminino , Taxa de Filtração Glomerular , Humanos , Rim/diagnóstico por imagem , Masculino , Matemática , Pessoa de Meia-Idade , Cintilografia , Estudos Retrospectivos
18.
Accid Anal Prev ; 105: 44-51, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27491717

RESUMO

Accidents between right-turning motor vehicles and straight-ahead cyclists are one of the most common accident types leading to cyclist injuries at signalised junctions in Denmark. A before-after safety evaluation of applying staggered stop lines in 189 arms at 123 signalised junctions is presented. The evaluation accounts for long-term accident trends and changes in motor vehicle traffic volumes. Applying staggered stop lines gives no decline in accidents between right-turning motor vehicles and straight-ahead cyclists. However, there is a statistical tendency to a decline of these right-turn accidents involving heavy vehicles. There are several questions about factors leading to right-turn accidents that cannot be answered by recorded accident data. A study of conflicting behaviour focuses on factors leading to conflicts. Video observations have been carried out in 10 arms at signalised junctions. A total of 45 situations with conflicting behaviour between right-turning motor vehicles and straight-ahead cyclists have been investigated and compared to a reference group of simultaneous arrivals. The relative risk is lowest when both parties stop on red before entering the junction. Upon simultaneous arrival of both parties at a green light, the relative risk is highest. Cyclists tend to have a higher relative risk of being involved in conflicts if they; a) ride through on yellow, b) have a time distance of at least 2seconds to other cyclists, c) wear a black jacket, and/or d) arrive at the junction at a speed of at least 25km/h. Much less can be said about the motor vehicles or their drivers on the basis of these video observations, but motor vehicles stopping in the cycle crossing in order to yield to pedestrians or cyclists have a higher relative risk of being involved in conflicts.


Assuntos
Acidentes de Trânsito/prevenção & controle , Acidentes de Trânsito/estatística & dados numéricos , Ciclismo/lesões , Planejamento Ambiental , Dinamarca , Humanos , Veículos Automotores/estatística & dados numéricos , Risco , Segurança
19.
Eur J Endocrinol ; 152(6): 851-62, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15941924

RESUMO

OBJECTIVE: The importance of the melanin-concentrating hormone (MCH) system for regulation of energy homeostasis and body weight has been demonstrated in rodents. We analysed the human MCH receptor 1 gene (MCHR1) with respect to human obesity. DESIGN: This consisted of genomic screening of 13.4 kb encompassing the MCHR1 in extremely obese German children and adolescents and association analyses for two coding single nucleotide polymorphisms (SNPs). To confirm initial positive association results, additional association studies and transmission disequilibrium tests in further German, Danish, French and American samples were conducted. Selected SNPs were investigated using functional in vitro studies and reporter gene assays. METHODS: Single-stranded conformation polymorphism analysis, re-sequencing, PCR-restriction fragment length polymorphism analyses, tetra-primer amplification refractory mutation systems, matrix-assisted laser desorption/ionization time of flight mass spectrometry and reporter gene assays were carried out as well as measuring inositol phosphate formation, inhibition of cAMP formation and activation of p42/44 MAP kinase. RESULTS: We identified 11 infrequent variations and two SNPs in the MCHR1 coding sequence and 18 SNPs (eight novel) in the flanking sequence. Association and transmission disequilibrium with obesity were detected for several SNPs in independent study groups of German obese children and adolescents and controls. In two German samples, encompassing 4056 and 295 individuals, trends towards association with obesity were detected. Findings in a second epidemiological German sample and in Danish, French and American samples were negative. Functional in vitro studies as well as reporter gene assays revealed no significant results. CONCLUSION: Our initial association of MCHR1 alleles/haplotype detected might be related to juvenile-onset obesity, conditional on a particular genetic and/or environmental background. Alternatively, we could not exclude the possibility that the initially detected association represented a false positive finding.


Assuntos
Obesidade/genética , Receptores do Hormônio Hipofisário/genética , Adolescente , Adulto , Animais , Células COS , Chlorocebus aethiops , AMP Cíclico/antagonistas & inibidores , DNA/química , DNA/genética , Feminino , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Polimorfismo de Nucleotídeo Único , Polimorfismo Conformacional de Fita Simples , Análise de Sequência de DNA
20.
J Mol Endocrinol ; 51(1): 79-90, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23613280

RESUMO

Metallothioneins (MTs) are cytoprotective proteins acting as scavengers of toxic metal ions or reactive oxygen species. MTs are upregulated in follicular thyroid carcinoma and are regarded as a marker of thyroid stress in Graves' disease. However, the mechanism of MT regulation in thyrocytes is still elusive. In other cellular systems, cAMP-, calcium-, or protein kinase C (PKC)-dependent signaling cascades have been shown to induce MT expression. Of note, all of these three pathways are activated following the stimulation of the TSH receptor (TSHR). Thus, we hypothesized that TSH represents a key regulator of MT expression in thyrocytes. In fact, TSHR stimulation induced expression of MT isoform 1X (MT1X) in human follicular carcinoma cells. In these cells, Induction of MT1X expression critically relied on intact Gq/11 signaling of the TSHR and was blocked by chelation of intracellular calcium and inhibition of PKC. TSHR-independent stimulation of cAMP formation by treating cells with forskolin also led to an upregulation of MT1X, which was completely dependent on PKA. However, inhibition of PKA did not affect the regulation of MT1X by TSH. As in follicular thyroid carcinoma cells, TSH also induced MT1 protein in primary human thyrocytes, which was PKC dependent as well. In summary, these findings indicate that TSH stimulation induces MT1X expression via Gq/11 and PKC, whereas cAMP-PKA signaling does not play a predominant role. To date, little has been known regarding cAMP-independent effects of TSHR signaling. Our findings extend the knowledge about the PKC-mediated functions of the TSHR.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Metalotioneína/metabolismo , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Glândula Tireoide/metabolismo , Tireotropina/farmacologia , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Carbacol/metabolismo , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Metalotioneína/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores da Tireotropina/genética , Receptores da Tireotropina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA