Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Mol Sci ; 17(1)2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26784176

RESUMO

During the last few decades, improvements in the planning and application of radiotherapy in combination with surgery and chemotherapy resulted in increased survival rates of tumor patients. However, the success of radiotherapy is impaired by two reasons: firstly, the radioresistance of tumor cells and, secondly, the radiation-induced damage of normal tissue cells located in the field of ionizing radiation. These limitations demand the development of drugs for either radiosensitization of tumor cells or radioprotection of normal tissue cells. In order to identify potential targets, a detailed understanding of the cellular pathways involved in radiation response is an absolute requirement. This review describes the most important pathways of radioresponse and several key target proteins for radiosensitization.


Assuntos
Dano ao DNA , Neoplasias/radioterapia , Tolerância a Radiação , Radiação Ionizante , Radiossensibilizantes/farmacologia , Animais , Reparo do DNA , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos da radiação
2.
J Biol Chem ; 289(18): 12896-907, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24668805

RESUMO

Cerebral ischemia frequently leads to long-term disability and death. Excitotoxicity is believed to be the main cause for ischemia-induced neuronal death. Although a role of glutamate receptors in this process has been firmly established, the contribution of metabotropic GABAB receptors, which control excitatory neurotransmission, is less clear. A prominent characteristic of ischemic insults is endoplasmic reticulum (ER) stress associated with the up-regulation of the transcription factor CCAAT/enhancer-binding protein-homologous protein (CHOP). After inducing ER stress in cultured cortical neurons by sustained Ca(2+) release from intracellular stores or by a brief episode of oxygen and glucose deprivation (in vitro model of cerebral ischemia), we observed an increased expression of CHOP accompanied by a strong reduction of cell surface GABAB receptors. Our results indicate that down-regulation of cell surface GABAB receptors is caused by the interaction of the receptors with CHOP in the ER. Binding of CHOP prevented heterodimerization of the receptor subunits GABAB1 and GABAB2 and subsequent forward trafficking of the receptors to the cell surface. The reduced level of cell surface receptors diminished GABAB receptor signaling and, thus, neuronal inhibition. These findings indicate that ischemia-mediated up-regulation of CHOP down-regulates cell surface GABAB receptors by preventing their trafficking from the ER to the plasma membrane. This mechanism leads to diminished neuronal inhibition and may contribute to excitotoxicity in cerebral ischemia.


Assuntos
Estresse do Retículo Endoplasmático , Neurônios/metabolismo , Receptores de GABA-B/metabolismo , Fator de Transcrição CHOP/metabolismo , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Regulação para Baixo , Feminino , Expressão Gênica , Glucose/metabolismo , Células HEK293 , Humanos , Microscopia Confocal , Neurônios/citologia , Oxigênio/metabolismo , Ligação Proteica , Ratos , Ratos Wistar , Receptores de GABA-B/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição CHOP/genética
3.
Strahlenther Onkol ; 190(8): 745-52, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24638269

RESUMO

Improvements of radiotherapy in combination with surgery and systemic therapy have resulted in increased survival rates of tumor patients. However, radiation-induced normal tissue toxicity is still dose limiting. Several strategies have been pursued with the goal to develop substances which may prevent or reduce damage to normal tissue. Drugs applied before radiotherapy are called radioprotectors; those given after radiotherapy to reduce long-term effects are radiomitigators. Despite more than 50 years of research, until now only two substances, amifostine and palifermin, have overcome all obstacles of clinical approval and are applied during radiotherapy of head and neck cancer or total body irradiation, respectively. However, better understanding of the cellular pathways involved in radiation response has allowed the development of several highly promising drugs functioning as scavengers of reactive oxygen species or targeting specific molecules involved in regulation of cell death pathways or cell cycle arrest. The present review describes the major targets for radioprotectors or radiomitigators currently tested in clinical trials.


Assuntos
Sobrevivência Celular/efeitos da radiação , Sequestradores de Radicais Livres/farmacologia , Lesões por Radiação/prevenção & controle , Protetores contra Radiação/farmacologia , Células Tumorais Cultivadas/efeitos da radiação , Amifostina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Ensaios Clínicos como Assunto , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Fracionamento da Dose de Radiação , Fator 7 de Crescimento de Fibroblastos/farmacologia , Humanos , Transferência Linear de Energia , Neoplasias Otorrinolaringológicas/patologia , Neoplasias Otorrinolaringológicas/radioterapia , Lesões por Radiação/patologia , Espécies Reativas de Oxigênio/efeitos da radiação , Células Tumorais Cultivadas/efeitos dos fármacos , Irradiação Corporal Total/efeitos adversos
4.
J Neurooncol ; 115(3): 323-31, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24022637

RESUMO

Epidermal growth factor receptor (EGFR) gene amplification and overexpression are commonly present in glioblastoma, and confer advantages of growth, invasiveness and radio/chemotherapy-resistance for tumour cells. Here, we assessed the role of EGFR activation for downstream mitogenic signalling in the commonly used glioblastoma cell line U251. Despite the high expression level, activation of EGFR under standard culture conditions was low. Intact EGFR function was verified by the rapid phosphorylation of EGFR and downstream mitogen-activated protein(MAP) kinase ERK1/2 upon addition of exogenous EGF to serum-starved cells. By contrast, addition of fetal bovine serum (FBS) activated downstream ERK1/2 via the MAP kinase kinase without phosphorylating EGFR. A phosphoreceptor tyrosine kinase array showed FBS-induced activation of insulin-like growth factor-1 receptor (IGF-1R),and the IGF-1R inhibitor AG1024 inhibited FBS-induced phosphorylation of ERK1/2, implying IGF-1R as the major driver of FBS-associated mitogenic signalling in the absence of exogenous EGF. These findings have important implications for in vitro drug testing in glioblastoma. Moreover, activation of ERK1/2 was also strongly influenced by growth state and cell density of U251 cultures. Re-seeding exponentially growing cultures at high cell density induced p27/CDKN1B expression and suppressed P-ERK1/2 indicating a certain regulation of proliferation by contact inhibition. Strikingly, highly activated ERK1/2 signalling and cell cycle progression occurred when cells were released from plateau phase regardless of high seeding density. This phenomenon might implicate a proliferation response in the early recurrence observed after clinical therapy in glioblastoma patients. However, whether it will recapitulate in vivo remains to be demonstrated.


Assuntos
Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor IGF Tipo 1/metabolismo , Animais , Western Blotting , Bovinos , Glioblastoma/patologia , Humanos , Fosforilação , Soro/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
5.
Artif Intell Med ; 133: 102405, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36328666

RESUMO

Antimicrobial resistance is one of the biggest threats to global health, food security, and development. Antibiotic overuse and misuse are the main drivers for the emergence of resistance. It is crucial to optimise the use of existing antibiotics in order to improve medical outcomes, decrease toxicity and reduce the emergence of resistance. We formulate the design of antibiotic dosing regimens as an optimisation problem, and use an evolutionary algorithm suited to continuous optimisation (differential evolution) to solve it. Regimens are represented as vectors of real numbers encoding daily doses, which can vary across the treatment duration. A stochastic mathematical model of bacterial infections with tuneable resistance levels is used to evaluate the effectiveness of evolved regimens. The objective is to minimise the treatment failure rate, subject to a constraint on the maximum total antibiotic used. We consider simulations with different levels of bacterial resistance, two ways of administering the drug (orally and intravenously), as well as coinfections with two strains of bacteria. Our approach produced effective dosing regimens, with an average improvement in lowering the failure rate 30%, when compared with standard fixed-daily-dose regimens with the same total amount of antibiotic.


Assuntos
Antibacterianos , Infecções Bacterianas , Humanos , Antibacterianos/uso terapêutico , Bactérias/genética , Infecções Bacterianas/tratamento farmacológico , Modelos Teóricos , Algoritmos
6.
Front Oncol ; 12: 802482, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35155239

RESUMO

In prostate cancer (PCa), a characteristic stromal-epithelial redistribution of the membrane protein caveolin 1 (CAV1) occurs upon tumor progression, where a gain of CAV1 in the malignant epithelial cells is accompanied by a loss of CAV1 in the tumor stroma, both facts that were correlated with higher Gleason scores, poor prognosis, and pronounced resistance to therapy particularly to radiotherapy (RT). However, it needs to be clarified whether inhibiting the CAV1 gain in the malignant prostate epithelium or limiting the loss of stromal CAV1 would be the better choice for improving PCa therapy, particularly for improving the response to RT; or whether ideally both processes need to be targeted. Concerning the first assumption, we investigated the RT response of LNCaP PCa cells following overexpression of different CAV1 mutants. While CAV1 overexpression generally caused an increased epithelial-to-mesenchymal phenotype in respective LNCaP cells, effects that were accompanied by increasing levels of the 5'-AMP-activated protein kinase (AMPK), a master regulator of cellular homeostasis, only wildtype CAV1 was able to increase the three-dimensional growth of LNCaP spheroids, particularly following RT. Both effects could be limited by an additional treatment with the SRC inhibitor dasatinib, finally resulting in radiosensitization. Using co-cultured (CAV1-expressing) fibroblasts as an approximation to the in vivo situation of early PCa it could be revealed that RT itself caused an activated, more tumor-promoting phenotype of stromal fibroblats with an increased an increased metabolic potential, that could not be limited by combined dasatinib treatment. Thus, targeting fibroblasts and/or limiting fibroblast activation, potentially by limiting the loss of stromal CAV1 seems to be absolute for inhibiting the resistance-promoting CAV1-dependent signals of the tumor stroma.

7.
J Biol Chem ; 285(46): 35606-14, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20826795

RESUMO

Metabotropic GABA(B) receptors are abundantly expressed at glutamatergic synapses where they control excitability of the synapse. Here, we tested the hypothesis that glutamatergic neurotransmission may regulate GABA(B) receptors. We found that application of glutamate to cultured cortical neurons led to rapid down-regulation of GABA(B) receptors via lysosomal degradation. This effect was mimicked by selective activation of AMPA receptors and further accelerated by coactivation of group I metabotropic glutamate receptors. Inhibition of NMDA receptors, blockade of L-type Ca(2+) channels, and removal of extracellular Ca(2+) prevented glutamate-induced down-regulation of GABA(B) receptors, indicating that Ca(2+) influx plays a critical role. We further established that glutamate-induced down-regulation depends on the internalization of GABA(B) receptors. Glutamate did not affect the rate of GABA(B) receptor endocytosis but led to reduced recycling of the receptors back to the plasma membrane. Blockade of lysosomal activity rescued receptor recycling, indicating that glutamate redirects GABA(B) receptors from the recycling to the degradation pathway. In conclusion, the data indicate that sustained activation of AMPA receptors down-regulates GABA(B) receptors by sorting endocytosed GABA(B) receptors preferentially to lysosomes for degradation on the expense of recycling. This mechanism may relieve glutamatergic synapses from GABA(B) receptor-mediated inhibition resulting in increased synaptic excitability.


Assuntos
Regulação para Baixo , Lisossomos/metabolismo , Receptores de GABA-B/metabolismo , Receptores de Glutamato/metabolismo , Animais , Western Blotting , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Ácido Egtázico/farmacologia , Endocitose/efeitos dos fármacos , Glutamatos/farmacologia , Imuno-Histoquímica , Transporte de Íons/efeitos dos fármacos , Microscopia Confocal , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nifedipino/farmacologia , Ratos , Ratos Wistar , Receptores de AMPA/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
8.
J Hepatol ; 55(2): 369-78, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21184784

RESUMO

BACKGROUND & AIMS: In injured liver, TGF-ß affects all hepatic cell types and participates in wound healing and fibrogenesis. TGF-ß downstream signaling is highly complex and cell type dependent, involving Smad and non-Smad signaling cascades thus requiring tight regulation. Endocytosis has gained relevance as important mechanism to control signaling initiation and termination. In this study, we investigated endocytic mechanisms for TGF-ß mediated Smad and non-Smad signaling in hepatocytes. METHODS: Endocytosis in hepatocytes was elucidated using chemical inhibitors, RNAi, viral gene transfer and caveolin-1-/- mice. TGF-ß signaling was monitored by Western blot, reporter assays and gene expression analysis. RESULTS: In hepatocytes, Smad activation is to a large degree accomplished AP-2 complex dependent on the hepatocyte surface without the necessity of clathrin coated pit formation or an endocytic step. In contrast, non-Smad/AKT pathway activation required functional dynamin mediated endocytosis and the presence of caveolin-1, an essential protein for caveolae formation. Furthermore, these two TGF-ß signaling initiation platforms discriminate distinct signaling routes that integrate at the transcriptional level as shown for TGF-ß target genes, Id1, Smad7, and CTGF. Endocytosis inhibition increased canonical Smad signaling and culminated in a superinduction of Id1 and Smad7 expression, whereas caveolin-1 mediated AKT pathway activation was required for maximal CTGF induction. CONCLUSIONS: Endocytosis is critical for TGF-ß signaling regulation in hepatocytes and determines gene expression signature and (patho)physiological outcome.


Assuntos
Endocitose/fisiologia , Hepatócitos/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Sequência de Bases , Caveolina 1/deficiência , Caveolina 1/genética , Caveolina 1/metabolismo , Invaginações Revestidas da Membrana Celular/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Transdução de Sinais
9.
Cytotherapy ; 13(3): 357-65, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20950214

RESUMO

BACKGROUND AIMS: Transplantation of allogeneic hematopoietic stem cells (HSC) within the framework of hematologic oncology or inherited diseases may be associated with complications such as engraftment failure and long-term pancytopenia. HSC engraftment can be improved, for example by co-transplantation with mesenchymal stem cells (MSC). Recently, a new multipotent MSC line from umbilical cord blood, unrestricted somatic stem cells (USSC), has been described. It was demonstrated that USSC significantly support proliferation of HSC in an in vitro feeder layer assay. METHODS: A NOD/SCID mouse model was used to assess the effect of USSC on co-transplanted CD34(+) cells and look for the fate of transplanted USSC. The migration potential of USSC was studied in a Boyden chamber migration assay and in vivo. Quantitative real-time polymerase chain reaction (qRT-PCR) for CXCR4, CD44, LFA1, CD62L, VLA4, RAC2, VLA5A and RAC1 were performed. NMR1 nu/nu mice were used for a tumorigenicity test. RESULTS: After 4 weeks, homing of human cells (CD45(+)) to the bone marrow of NOD/SCID mice was significantly increased in mice co-transplanted with CD34(+) cells and USSC (median 30.9%, range 7-50%) compared with the CD34(+) cell-only control group (median 5.9%, range 3-10%; P = 0.004). Homing of USSC could not be shown in the bone marrow. A cell-cell contact was not required for the graft enhancing effect of USSC. An in vivo tumorigenicity assay showed no tumorigenic potential of USSC. CONCLUSIONS: This pre-clinical study clearly shows that USSC have an enhancing effect on engraftment of human CD34(+) cells. USSC are a safe graft adjunct.


Assuntos
Antígenos CD34/metabolismo , Comunicação Celular/genética , Transformação Celular Neoplásica/patologia , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Humanos , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos SCID
10.
Onkologie ; 34(10): 502-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21985848

RESUMO

BACKGROUND: The development of distant metastasis is associated with poor outcome in patients with colorectal cancer (CRC). The stromal cell-derived factor-1 (SDF-1) and its receptor CXC chemokine receptor 4 (CXCR4) have pivotal roles in the chemotaxis of migrating tumor cells during metastasis. Thus, hampering the SDF-1/CXCR4 cross-talk is a promising strategy to suppress metastasis. METHODS: We investigated the invasive behavior of the lentivirally CXCR4 overexpressing CRC cell lines SW480, SW620 and RKO in chemotaxis and invasion assays toward an SDF-1α gradient. Low endogenous CXCR4 expression levels were determined by quantitative realtime polymerase chain reaction (PCR) and fluorescence-activated cell sorting (FACS) analyses. RESULTS: A lentiviral CXCR4 overexpression and knockdown model was established in these CRC cells. In transwell migration assays, CXCR4 overexpression favored chemotaxis and invasion of cells in all 3 lines depending on an SDF-1α gradient (p < 0.001 vs. untransduced cells). Functional CXCR4 knockdown using lentiviral short hairpin RNA (shRNA) vectors significantly decreased the migration behavior in CRC cell lines (p < 0.001), confirming a CXCR4-specific effect. Pharmacologic inhibition of the SDF-1α/CXCR4 interaction by the bicyclam Plerixafor(TM) at 100 µM significantly abrogated CXCR4-dependent migration and invasion through Matrigel(TM) (SW480, SW620, RKO; p < 0.05). CONCLUSION: Our results indicate that a CXCR4-antagonistic therapy might prevent tumor cell dissemination and metastasis in CRC patients, consequently improving survival.


Assuntos
Fármacos Anti-HIV/farmacologia , Quimiocina CXCL12/genética , Quimiotaxia/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica/genética , Vetores Genéticos/genética , Compostos Heterocíclicos/farmacologia , Lentivirus/genética , Invasividade Neoplásica/genética , Receptores CXCR4/genética , Benzilaminas , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Ciclamos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Invasividade Neoplásica/patologia , Receptor Cross-Talk/efeitos dos fármacos , Receptores CXCR4/antagonistas & inibidores
11.
J Inflamm Res ; 14: 2697-2712, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34188517

RESUMO

BACKGROUND: Donor-specific antibodies (DSA) against donor human leukocyte antigen after liver transplantation, which are associated with histological changes, have been widely studied with respect to their sustained impact on transplant function. However, their long-term impact after liver transplantation remains unclear. METHODS: We performed a cross-sectional analysis from June 2016 to July 2017 that included all patients who presented themselves for scheduled follow-up after receiving a liver transplantation between September 1989 and December 2016. In addition to a liver protocol biopsy, patients were screened for human leukocyte antigen antibodies (HLAab) and donor-specific antibodies. Subsequently, the association between human leukocyte antigen antibodies, donor-specific antibodies, histologic and clinical features, and immunosuppression was analyzed. RESULTS: Analysis for human leukocyte antigen antibodies and donor-specific antibodies against donor human leukocyte antigen was performed for 291 and 271 patients. A significant association between higher inflammation grades and the presence of human leukocyte antigen antibodies and donor-specific antibodies was detected, while fibrosis stages remained unaffected. These results were confirmed by multivariate logistic regression for inflammation showing a significant increase for presence of human leukocyte antigen antibodies and donor-specific antibodies (OR: 4.43; 95% CI: 1.67-12.6; p=0.0035). Furthermore, the use of everolimus in combination with tacrolimus was significantly associated with the status of negative human leukocyte antigen antibodies and donor-specific antibodies. Viral etiology for liver disease, hepatocellular carcinoma (HCC) and higher steatosis grades of the graft were significantly associated with a lower rate of human leukocyte antigen antibodies. The impact of human leukocyte antigen antibodies and donor-specific antibodies against donor human leukocyte antigen was associated with higher levels of laboratory parameters, such as transaminases and bilirubin. CONCLUSION: Donor-specific antibodies against donor human leukocyte antigen are associated with histological and biochemical graft inflammation after liver transplantation, while fibrosis seems to be unaffected. Future studies should validate these findings for longer observation periods and specific subgroups.

12.
Strahlenther Onkol ; 186(2): 99-106, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20127227

RESUMO

BACKGROUND AND PURPOSE: The transmembrane protein caveolin-1 (CAV1) is an essential component of caveolae, small membrane invaginations involved in vesicle formation. CAV1 plays a role in signal transduction, tumor suppression and oncogene transformation. Previous studies with CAV1 knockout mice and CAV1 knockdown in pancreatic tumor cells implicated CAV1 in mediating radioresistance. The aim of this work was to test the effect of CAV1 overexpression after irradiation in human cells lacking endogenous CAV1 expression. MATERIAL AND METHODS: Human CAV1 was overexpressed in lymphoblastoid TK6 cells (TK6-wt) using a eukaryotic expression plasmid, pCI-CAV1, or a lentiviral SIN (self-inactivating) vector, HR'SIN-CAV1. CAV1 expression was verified in TK6 cells with Western blot analysis or intracellular FACS (fluorescence-activated cell sorting) staining. The effect of CAV1 on proliferation kinetics after irradiation of TK6 cells was measured with a growth assay. RESULTS: TK6-wt showed no detectable endogenous CAV1 expression. Lentivirally mediated transduction with HR'SIN-CAV1 (TK6-CAV1) resulted in a considerably stronger CAV1 expression in comparison to TK6 cells electroporated with pCI-CAV1. Intracellular FACS analysis showed that 90% of transduced cells expressed CAV1. CAV1 enhanced early proliferation of TK6 cells after irradiation with a dose of 2 Gy, whereas proliferation of unirradiated cells was not affected. CAV1 also protected cells after irradiation with 4 Gy. This radioprotective effect was supported by a reduction of radiation-induced apoptosis. CONCLUSION: A model system for expression of exogenous CAV1 by stable lentiviral transduction of TK6 cells was established. Functional assays demonstrated enhanced early proliferation by CAV1 expression in TK6 cells after irradiation with clinically relevant doses supporting the role of CAV1 as a prosurvival factor.


Assuntos
Apoptose/efeitos da radiação , Caveolina 1/genética , Divisão Celular/efeitos da radiação , Expressão Gênica/efeitos da radiação , Apoptose/genética , Western Blotting , Divisão Celular/genética , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Citometria de Fluxo , Expressão Gênica/genética , Vetores Genéticos/genética , Humanos , Lentivirus/genética , Linfócitos/metabolismo , Linfócitos/efeitos da radiação , Plasmídeos , Proteção Radiológica , Transdução Genética/métodos
13.
Cytotherapy ; 12(3): 338-48, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20370349

RESUMO

BACKGROUND AIMS: The discovery of unrestricted somatic stem cells (USSC), a non-hematopoietic stem cell population, brought cord blood (CB) to the attention of regenerative medicine for defining more protocols for non-hematopoietic indications. We demonstrate that a reliable and reproducible method for good manufacturing practice (GMP)-conforming generation of USSC is possible that fulfils safety requirements as well as criteria for clinical applications, such as adherence of strict regulations on cell isolation and expansion. METHODS: In order to maintain GMP conformity, the automated cell processing system Sepax (Biosafe) was implemented for mononucleated cell (MNC) separation from fresh CB. After USSC generation, clinical-scale expansion was achieved by multi-layered CellSTACKs (Costar/Corning). Infectious disease markers, pyrogen and endotoxin levels, immunophenotype, potency, genetic stability and sterility of the cell product were evaluated. RESULTS: The MNC isolation and cell cultivation methods used led to safe and reproducible GMP-conforming USSC production while maintaining somatic stem cell character. CONCLUSIONS: Together with implemented in-process controls guaranteeing contamination-free products with adult stem cell character, USSC produced as suggested here may serve as a universal allogeneic stem cell source for future cell treatment and clinical settings.


Assuntos
Sangue Fetal/citologia , Transplante de Células-Tronco , Células-Tronco , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Linhagem da Célula , Células Cultivadas , Centrifugação com Gradiente de Concentração/instrumentação , Centrifugação com Gradiente de Concentração/métodos , Citometria de Fluxo , Humanos , Imunofenotipagem , Camundongos , Camundongos Nus , Regeneração/fisiologia , Transplante de Células-Tronco/legislação & jurisprudência , Transplante de Células-Tronco/métodos , Transplante de Células-Tronco/normas , Células-Tronco/citologia , Células-Tronco/fisiologia , Transplante Autólogo , Transplante Homólogo
14.
Cell Death Dis ; 11(4): 228, 2020 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-32273493

RESUMO

The integral membrane protein caveolin-1 (CAV1) plays a central role in radioresistance-mediating tumor-stroma interactions of advanced prostate cancer (PCa). Among the tumor-stroma, endothelial cells (EC) evolved as critical determinants of the radiation response. CAV1 deficiency in angiogenic EC was already shown to account for increased apoptosis rates of irradiated EC. This study explores the potential impact of differential CAV1 levels in EC on the acid sphingomyelinase (ASMase)/ceramide pathway as a key player in the regulation of EC apoptosis upon irradiation and cancer cell radioresistance. Enhanced apoptosis sensitivity of CAV1-deficient EC was associated with increased ASMase activity, ceramide generation, formation of large lipid platforms, and finally an altered p38 mitogen-activated protein kinase (MAPK)/heat-shock protein 27 (HSP27)/AKT (protein kinase B, PKB) signaling. CAV1-deficient EC increased the growth delay of LNCaP and PC3 PCa cells upon radiation treatment in direct 3D spheroid co-cultures. Exogenous C6 and C16 ceramide treatment in parallel increased the growth delay of PCa spheroids and induced PCa cell apoptosis. Analysis of the respective ceramide species in PCa cells with increased CAV1 levels like those typically found in radio-resistant advanced prostate tumors further revealed an upregulation of unsaturated C24:1 ceramide that might scavenge the effects of EC-derived apoptosis-inducing C16 ceramide. Higher ASMase as well as ceramide levels could be confirmed by immunohistochemistry in human advanced prostate cancer specimen bearing characteristic CAV1 tumor-stroma alterations. Conclusively, CAV1 critically regulates the generation of ceramide-dependent (re-)organization of the plasma membrane that in turn affects the radiation response of EC and adjacent PCa cells. Understanding the CAV1-dependent crosstalk between tumor cells and the host-derived tumor microvasculature and its impact on radiosensitivity may allow to define a rational strategy for overcoming tumor radiation resistance improving clinical outcomes by targeting CAV1.


Assuntos
Caveolina 1/metabolismo , Ceramidas/metabolismo , Células Endoteliais/efeitos da radiação , Neoplasias da Próstata/patologia , Neoplasias da Próstata/radioterapia , Esfingomielina Fosfodiesterase/metabolismo , Células Estromais/patologia , Caveolina 1/biossíntese , Caveolina 1/deficiência , Comunicação Celular/fisiologia , Comunicação Celular/efeitos da radiação , Linhagem Celular Tumoral , Ceramidas/biossíntese , Ceramidas/farmacologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Masculino , Células PC-3 , Neoplasias da Próstata/metabolismo , Tolerância a Radiação , Transdução de Sinais , Células Estromais/metabolismo , Microambiente Tumoral
15.
J Clin Med ; 8(3)2019 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-30871022

RESUMO

Tumour resistance to chemo- and radiotherapy, as well as molecularly targeted therapies, limits the effectiveness of current cancer treatments. We previously reported that the radiation response of human prostate tumours is critically regulated by CAV1 expression in stromal fibroblasts and that loss of stromal CAV1 expression in advanced tumour stages may contribute to tumour radiotherapy resistance. Here we investigated whether fibroblast secreted anti-apoptotic proteins could induce radiation resistance of prostate cancer cells in a CAV1-dependent manner and identified TRIAP1 (TP53 Regulated Inhibitor of Apoptosis 1) as a resistance-promoting CAV1-dependent factor. TRIAP1 expression and secretion was significantly higher in CAV1-deficient fibroblasts and secreted TRIAP1 was able to induce radiation resistance of PC3 and LNCaP prostate cancer cells in vitro, as well as of PC3 prostate xenografts derived from co-implantation of PC3 cells with TRIAP1-expressing fibroblasts in vivo. Immunohistochemical analyses of irradiated PC3 xenograft tumours, as well as of human prostate tissue specimen, confirmed that the characteristic alterations in stromal-epithelial CAV1 expression were accompanied by increased TRIAP1 levels after radiation in xenograft tumours and within advanced prostate cancer tissues, potentially mediating resistance to radiation treatment. In conclusion, we have determined the role of CAV1 alterations potentially induced by the CAV1-deficient, and more reactive, stroma in radio sensitivity of prostate carcinoma at a molecular level. We suggest that blocking TRIAP1 activity and thus avoiding drug resistance may offer a promising drug development strategy for inhibiting resistance-promoting CAV1-dependent signals.

16.
Radiat Res ; 169(3): 301-10, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18302483

RESUMO

Tumor radiotherapy with large-field irradiation results in an increase in apoptosis of the radiosensitive hematopoietic stem cells (CD34(+)). The aim of this study was to demonstrate the radioprotective potential of MDR1 overexpression in human CD34(+) cells using a lentiviral self-inactivating vector. Transduced human undifferentiated CD34(+) cells were irradiated with 0-8 Gy and held in liquid culture under myeloid-specific maturation conditions. After 12 days, MDR1 expression was determined by the rhodamine efflux assay. The proportion of MDR1-positive cells in cells from four human donors increased with increasing radiation dose (up to a 14-fold increase at 8 Gy). Determination of expression of myeloid-specific surface marker proteins revealed that myeloid differentiation was not affected by transduction and MDR1 overexpression. Irradiation after myeloid differentiation also led to an increase of MDR1-positive cells with escalating radiation doses (e.g. 12.5-16% from 0-8 Gy). Most importantly, fractionated irradiation (3 x 2 Gy; 24-h intervals) of MDR1-transduced CD34(+) cells resulted in an increase in MDR1-positive cells (e.g. 3-8% from 0-3 x 2 Gy). Our results clearly support a radioprotective effect of lentiviral MDR1 overexpression in human CD34(+) cells. Thus enhancing repopulation by surviving stem cells may increase the radiation tolerance of the hematopoietic system, which will contribute to widening the therapeutic index in radiotherapy.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Hematopoéticas/efeitos da radiação , Lentivirus/genética , Proteção Radiológica/métodos , Tolerância a Radiação/fisiologia , Receptores de Complemento 3b/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Relação Dose-Resposta à Radiação , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/citologia , Humanos , Doses de Radiação , Proteínas Recombinantes/metabolismo , Transfecção/métodos
17.
Exp Hematol ; 34(8): 1052-9, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16863911

RESUMO

OBJECTIVE: AMD3100 is a new CXCR4 antagonist that induces a rapid release of hematopoietic progenitors from the bone marrow to the peripheral blood. We conducted a clinical study where patients with multiple myeloma and non-Hodgkin's lymphoma were treated with AMD3100 (A) to increase the number of peripheral blood progenitor cells (PBPCs) when given a mobilization regimen of granulocyte colony-stimulating factor (G-CSF, G). Because experimental data suggest that A+G-mobilized PBPCs are functionally different from G-mobilized PBPCs, we were interested in an intraindividual comparison of the gene expression profile of CD34+ cells in the two different settings. METHODS: To this end peripheral blood CD34+ cells of three patients (three G, three A+G samples) were isolated by immunomagnetic followed by flow cytometric sorting to a purity of >99%. Total RNA was purified. Differentially expressed genes were analyzed by using the Affymetrix GeneChip Human Genome U133 Plus2.0 and the software package Micro Array Solutions 1.3 (SAS Institute Inc.). RESULTS: We found a pattern of unanimously higher (81 genes, log2 ratio > 0.5; p < 0.0001) or lower (29 genes, log2 ratio < -0.4; p < 0.0001) expressed genes in the A+G-mobilized vs G-mobilized CD34+ PBPCs. Significant changes of four selected genes noted in the microarray analysis were validated by quantitative real-time polymerase chain reaction. Genes were grouped according to gene function. Only increased expression was found in the categories antiapoptosis (e.g., MPO, HSPA1B), cell cycle (e.g., MS4A3, RRM2), replication/DNA repair (e.g., MPO, HSPA1B), cell motility (e.g., TNFSF4, HMMR), and oxygen transport. Decreased expression occurred in the proapoptosis gene group (e.g., MDA5, BCL10). CXCR4 receptor gene expression itself was significantly 1.5-fold higher in the A+G vs G group. CONCLUSION: We conclude that A+G-mobilized CD34+ PBPCs express significantly higher amounts of genes that potentially promote superior engraftment after myeloablative therapy than G-mobilized CD34+ PBPCs.


Assuntos
Perfilação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Compostos Heterocíclicos/farmacologia , Transplante de Células-Tronco de Sangue Periférico , Receptores CXCR4/antagonistas & inibidores , Benzilaminas , Ciclamos , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Receptores CXCR4/fisiologia
18.
Sci Rep ; 7: 41138, 2017 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-28112237

RESUMO

Despite good treatment results in localized prostate tumors, advanced disease stages usually have a pronounced resistance to chemotherapy and radiotherapy. The membrane protein caveolin-1 (Cav1) functions here as an important oncogene. Therefore we examined the impact of stromal Cav1 expression for tumor growth and sensitivity to ionizing radiation (IR). Silencing of Cav1 expression in PC3 cells resulted in increased tumor growth and a reduced growth delay after IR when compared to tumors generated by Cav1-expressing PC3 cells. The increased radiation resistance was associated with increasing amounts of reactive tumor stroma and a Cav1 re-expression in the malignant epithelial cells. Mimicking the human situation these results were confirmed using co-implantation of Cav1-silenced PC3 cells with Cav1-silenced or Cav1-expressing fibroblasts. Immunohistochemically analysis of irradiated tumors as well as human prostate tissue specimen confirmed that alterations in stromal-epithelial Cav1 expressions were accompanied by a more reactive Cav1-reduced tumor stroma after radiation and within advanced prostate cancer tissues which potentially mediates the resistance to radiation treatment. Conclusively, the radiation response of human prostate tumors is critically regulated by Cav1 expression in stromal fibroblasts. Loss of stromal Cav1 expression in advanced tumor stages may thus contribute to resistance of these tumors to radiotherapy.


Assuntos
Caveolina 1/metabolismo , Caveolina 1/efeitos da radiação , Progressão da Doença , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Neoplasias da Próstata/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Xenoenxertos , Humanos , Masculino , Camundongos , Células Estromais/metabolismo , Células Estromais/efeitos da radiação
19.
Front Neurorobot ; 11: 2, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28179882

RESUMO

Combined efforts in the fields of neuroscience, computer science, and biology allowed to design biologically realistic models of the brain based on spiking neural networks. For a proper validation of these models, an embodiment in a dynamic and rich sensory environment, where the model is exposed to a realistic sensory-motor task, is needed. Due to the complexity of these brain models that, at the current stage, cannot deal with real-time constraints, it is not possible to embed them into a real-world task. Rather, the embodiment has to be simulated as well. While adequate tools exist to simulate either complex neural networks or robots and their environments, there is so far no tool that allows to easily establish a communication between brain and body models. The Neurorobotics Platform is a new web-based environment that aims to fill this gap by offering scientists and technology developers a software infrastructure allowing them to connect brain models to detailed simulations of robot bodies and environments and to use the resulting neurorobotic systems for in silico experimentation. In order to simplify the workflow and reduce the level of the required programming skills, the platform provides editors for the specification of experimental sequences and conditions, environments, robots, and brain-body connectors. In addition to that, a variety of existing robots and environments are provided. This work presents the architecture of the first release of the Neurorobotics Platform developed in subproject 10 "Neurorobotics" of the Human Brain Project (HBP). At the current state, the Neurorobotics Platform allows researchers to design and run basic experiments in neurorobotics using simulated robots and simulated environments linked to simplified versions of brain models. We illustrate the capabilities of the platform with three example experiments: a Braitenberg task implemented on a mobile robot, a sensory-motor learning task based on a robotic controller, and a visual tracking embedding a retina model on the iCub humanoid robot. These use-cases allow to assess the applicability of the Neurorobotics Platform for robotic tasks as well as in neuroscientific experiments.

20.
Radiat Res ; 166(3): 463-73, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16953664

RESUMO

Overexpression of P-glycoprotein (P-gp), the product of the MDR1 (multidrug resistance 1) gene, might complement chemotherapy and radiotherapy in the treatment of tumors. However, for safety and mechanistic reasons, it is important to know whether MDR1 overexpression influences the expression of other genes. Therefore, we analyzed differential gene expression in cells of the human lymphoblast cell line TK6 retrovirally transduced with MDR1 using the GeneChip Human Genome U133 Plus2.0 (Affymetrix). Sixty-one annotated genes showed a significant change in expression (P < 10(-4)) in MDR1-overexpressing cells compared to untransduced cells and cells transduced with a control virus expressing the neomycin phosphotransferase gene. Several genes coding for proteins involved in detoxification and exocytosis showed approximately 1.4- 4-fold increases in transcript levels (e.g. ALDH1A, UNC13). Additionally, pro-apoptosis genes were down-regulated (e.g. twofold for CASP1, 2.5-fold for NALP7) with concomitant increased expression of the potential anti-apoptosis gene AKT3. In functional assays the influence of MDR1 overexpression on apoptosis signaling was further corroborated by showing reduced rates of apoptosis in response to irradiation in TK6 cells transduced with MDR1. In conclusion, the resistant phenotype of MDR1-mediated P-gp-overexpressing cells is associated with differential expression of genes coding for metabolic and apoptosis-related proteins. These results have important implications for understanding the mechanisms by which MDR1 gene therapy can protect normal tissues from radiation- or chemotherapy-induced damage during tumor treatment.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Linfócitos/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Apoptose/genética , Linhagem Celular , Sobrevivência Celular/genética , Relação Dose-Resposta à Radiação , Regulação da Expressão Gênica/fisiologia , Regulação da Expressão Gênica/efeitos da radiação , Vetores Genéticos/genética , Humanos , Linfócitos/citologia , Linfócitos/efeitos da radiação , Doses de Radiação , Proteção Radiológica , Retroviridae/genética , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA