Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Pept Sci ; 30(7): e3592, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38447547

RESUMO

The CRISPR-Cas9 system has revolutionized the field of genetic engineering, but targeted cellular delivery remains a central problem. The delivery of the preformed ribonuclease-protein (RNP) complex has the advantages of fewer side effects and avoidance of potential permanent effects. We reasoned that an internalizing IgG antibody as a targeting device could address the delivery of Cas9-RNP. We opted for protein trans-splicing mediated by a split intein to facilitate posttranslational conjugation of the two large protein entities. We recently described the cysteine-less CL split intein that efficiently performs under oxidizing conditions and does not interfere with disulfide bonds or thiol bioconjugation chemistries. Using the CL split intein, we report for the first time the ligation of monoclonal IgG antibody precursors, expressed in mammalian cells, and a Cas9 precursor, obtained from bacterial expression. A purified IgG-Cas9 conjugate was loaded with sgRNA to form the active RNP complex and introduced a double-strand break in its target DNA in vitro. Furthermore, a synthetic peptide variant of the short N-terminal split intein precursor proved useful for chemical modification of Cas9. The split intein ligation procedure reported here for IgG-Cas9 provides the first step towards a novel CRISPR-Cas9 targeting approach involving the preformed RNP complex.


Assuntos
Sistemas CRISPR-Cas , Imunoglobulina G , Inteínas , Imunoglobulina G/química , Imunoglobulina G/genética , Humanos , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Proteína 9 Associada à CRISPR/química
2.
Z Gastroenterol ; 61(10): 1365-1370, 2023 Oct.
Artigo em Alemão | MEDLINE | ID: mdl-36482058

RESUMO

BACKGROUND: Pancreatic cancer is despite modern diagnostic tools and treatment regimen associated with poor outcome. Many patients show cachexia and sarcopenia. METHODS: In a retrospective analysis the SMI (cm²/m²) was measured by determining the skelettal muscle area in a computed tomography image at lumbar vertebrae 3. Further clinical parameters were measured to determine the outcome. RESULTS: The mean survival after diagnosis in the population with sarcopenia was significantly lower (14,4 vs 17,7 months, p=0,046). Significantly shorter survival was also seen for higher age (p=0,006), no tumor resection (p=0,004), metastases (p=0,002) and high CA19-9 level (p=0,002) CONCLUSION: Sarcopenia is an indipendant prognostic factor in patients with pancreatic cancer. SMI should be measured clinical practice and further studies are necessary to asses a potential therapeutic strategy.


Assuntos
Músculo Esquelético , Neoplasias Pancreáticas , Sarcopenia , Humanos , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/patologia , Neoplasias Pancreáticas/complicações , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/tratamento farmacológico , Prognóstico , Estudos Retrospectivos , Sarcopenia/diagnóstico , Sarcopenia/patologia , Tomografia Computadorizada por Raios X , Vértebras Lombares/diagnóstico por imagem , Caquexia , Neoplasias Pancreáticas
3.
Angew Chem Int Ed Engl ; 61(1): e202109769, 2022 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-34725904

RESUMO

Ibrutinib is an inhibitor of Bruton's tyrosine kinase that has been approved for the treatment of patients with chronic lymphocytic leukemia, mantle cell lymphoma and Waldenstrom's macroglobulinemia and is connected with toxicities. To minimize its toxicities, we linked ibrutinib to a cell-targeted, internalizing antibody. To this end, we synthesized a poly-anionic derivate, ibrutinib-Cy3.5, that retains full functionality. This anionic inhibitor is complexed by our anti-CD20-protamine targeting conjugate and free protamine, and thereby spontaneously assembles into an electrostatically stabilized vesicular nanocarrier. The complexation led to an accumulation of the drug driven by the CD20 antigen internalization to the intended cells and an amplification of its pharmacological effectivity. In vivo, we observed a significant enrichment of the drug in xenograft lymphoma tumors in immune-compromised mice and a significantly better response to lower doses compared to the original drug.


Assuntos
Adenina/análogos & derivados , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Carbocianinas/farmacologia , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Piperidinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Adenina/química , Adenina/farmacologia , Animais , Anticorpos Monoclonais/química , Antineoplásicos/química , Carbocianinas/química , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Piperidinas/química , Engenharia de Proteínas , Inibidores de Proteínas Quinases/química , Eletricidade Estática
4.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33255664

RESUMO

A microfluidic assay for the detection of promyelocytic leukemia (PML)-retinoic acid receptor α (RARα) fusion protein was developed. This microfluidic-based system can be used for rapid personalized differential diagnosis of acute promyelocyte leukemia (APL) with the aim of early initiation of individualized therapy. The fusion protein PML-RARα occurs in 95% of acute promyelocytic leukemia cases and is considered as diagnostically relevant. The fusion protein is formed as a result of translocation t(15,17) and is detected in the laboratory by fluorescence in situ hybridization (FISH) or reverse transcriptase polymerase chain reaction (RT-PCR). Diagnostic methods require many laboratory steps with specialized staff. The developed microfluidic assay includes a sandwich enzyme-linked immunosorbent assay (ELISA) system for PML-RARα on surface of magnetic microparticles in a microfluidic chip. A rapid detection of PML-RARα in cell lysates is achieved in less than one hour. A biotinylated PML-antibody on the surface of magnetic streptavidin coated microparticles is used as capture antibody. The bound translocation product is detected by a RARα antibody conjugated with horseradish peroxidase and the substrate QuantaRed. The analysis is performed in microfluidic channels which involves automated liquid processing with stringent washing and short incubation times. The results of the developed assay show that cell lysates of PML-RARα-positive cells (NB-4) can be clearly distinguished from PML-RARα-negative cells (HL-60, MV4-11).


Assuntos
Leucemia Promielocítica Aguda/diagnóstico , Proteínas de Fusão Oncogênica/genética , Proteína da Leucemia Promielocítica/genética , Receptor alfa de Ácido Retinoico/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/isolamento & purificação , Células Precursoras de Granulócitos/metabolismo , Células Precursoras de Granulócitos/patologia , Humanos , Hibridização in Situ Fluorescente/métodos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patologia , Microfluídica/métodos , Proteínas de Fusão Oncogênica/isolamento & purificação , Medicina de Precisão , Translocação Genética/genética
5.
Mol Pharm ; 14(5): 1339-1351, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28170265

RESUMO

RNA interference strategies offer an alternative to small molecular drug targeting. Small interfering RNA (siRNA) constitutes a class of molecules that allows the effective and specific inhibition of the biosynthesis of any protein. Indeed, siRNA have emerged as a major tool in molecular biology techniques and an important approach to identify suitable therapy targets in cancer. However, siRNA therapy approaches in vivo are scarce. Two major problems hinder siRNA as a therapeutic tool: (1) delivery through the bloodstream leads to degradation or rapid renal clearance (stabilization) and (2) specific uptake by the desired cell type (specificity). This review summarizes the ongoing attempts to use RNAi against disease-causing factors. We compare methods to stabilize siRNA in different conjugates and that decorate these complexes with targeting molecules such as antibodies, single-chain Fv or Fab fragments, to enable specific uptake of the carriers by the respective cells. We propose the development of antibody-coupled siRNA complexes, which have shown to allow stabilization as well as targeted uptake of siRNA to cancer cells.


Assuntos
Anticorpos/química , RNA Interferente Pequeno/administração & dosagem , Fragmentos Fab das Imunoglobulinas/química , Interferência de RNA , RNA Interferente Pequeno/química
6.
Am J Hum Genet ; 86(1): 83-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20085714

RESUMO

Autosomal-dominant striatal degeneration (ADSD) is an autosomal-dominant movement disorder affecting the striatal part of the basal ganglia. ADSD is characterized by bradykinesia, dysarthria, and muscle rigidity. These symptoms resemble idiopathic Parkinson disease, but tremor is not present. Using genetic linkage analysis, we have mapped the causative genetic defect to a 3.25 megabase candidate region on chromosome 5q13.3-q14.1. A maximum LOD score of 4.1 (Theta = 0) was obtained at marker D5S1962. Here we show that ADSD is caused by a complex frameshift mutation (c.94G>C+c.95delT) in the phosphodiesterase 8B (PDE8B) gene, which results in a loss of enzymatic phosphodiesterase activity. We found that PDE8B is highly expressed in the brain, especially in the putamen, which is affected by ADSD. PDE8B degrades cyclic AMP, a second messenger implied in dopamine signaling. Dopamine is one of the main neurotransmitters involved in movement control and is deficient in Parkinson disease. We believe that the functional analysis of PDE8B will help to further elucidate the pathomechanism of ADSD as well as contribute to a better understanding of movement disorders.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/genética , Regulação da Expressão Gênica , Mutação , Doenças Neurodegenerativas/metabolismo , Encéfalo/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Feminino , Mutação da Fase de Leitura , Genes Dominantes , Ligação Genética , Humanos , Escore Lod , Masculino , Doença de Parkinson/genética , Sistemas do Segundo Mensageiro , Transdução de Sinais
7.
Endosc Int Open ; 11(9): E818-E828, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37727511

RESUMO

Background and study aims Artificial intelligence (AI) in gastrointestinal endoscopy is developing very fast. Computer-aided detection of polyps and computer-aided diagnosis (CADx) for polyp characterization are available now. This study was performed to evaluate the diagnostic performance of a new commercially available CADx system in clinical practice. Patients and methods This prospective, non-randomized study was performed at a tertiary academic endoscopy center from March to August 2022. We included patients receiving a colonoscopy. Polypectomy had to be performed in all polyps. Every patient was examined concurrently by an endoscopist and AI using two opposing screens. The AI system, overseen by a second observer, was not visible to the endoscopist. The primary outcome was accuracy of the AI classifying the polyps into "neoplastic" and "non-neoplastic." The secondary outcome was accuracy of the classification by the endoscopists. Sessile serrated lesions were classified as neoplastic. Results We included 156 patients (mean age 65; 57 women) with 262 polyps ≤10 mm. Eighty-four were hyperplastic polyps (32.1%), 158 adenomas (60.3%), seven sessile serrated lesions (2.7%) and 13 other entities (normal/inflammatory colonmucosa, lymphoidic polyp) (4.9%) on histological diagnosis. Sensitivity, specificity and accuracy of AI were 89.70% (95% confidence interval [CI]: 84.02%-93.88%), 75.26% (95% CI: 65.46%-83.46%) and 84.35% (95% CI:79.38%-88.53%), respectively. Sensitivity, specificity and accuracy for less experienced endoscopists (2-5 years of endoscopy) were 95.56% (95% CI: 84.85%-99.46%), 61.54% (95% CI: 40.57%-79.77%) and 83.10% (95% CI: 72.34%-90.95%) and for experienced endoscopists 90.83% (95% CI: 84.19%-95.33%), 71.83% (95% CI: 59.90%-81.87%) and 83.77% (95% CI: 77.76%-88.70%), respectively. Conclusion Accuracy for polyp characterization by a new commercially available AI system is high, but does not fulfill the criteria for a "resect-and-discard" strategy.

8.
Cell Stem Cell ; 30(5): 706-721.e8, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37098346

RESUMO

Inter-patient variability and the similarity of healthy and leukemic stem cells (LSCs) have impeded the characterization of LSCs in acute myeloid leukemia (AML) and their differentiation landscape. Here, we introduce CloneTracer, a novel method that adds clonal resolution to single-cell RNA-seq datasets. Applied to samples from 19 AML patients, CloneTracer revealed routes of leukemic differentiation. Although residual healthy and preleukemic cells dominated the dormant stem cell compartment, active LSCs resembled their healthy counterpart and retained erythroid capacity. By contrast, downstream myeloid progenitors constituted a highly aberrant, disease-defining compartment: their gene expression and differentiation state affected both the chemotherapy response and leukemia's ability to differentiate into transcriptomically normal monocytes. Finally, we demonstrated the potential of CloneTracer to identify surface markers misregulated specifically in leukemic cells. Taken together, CloneTracer reveals a differentiation landscape that mimics its healthy counterpart and may determine biology and therapy response in AML.


Assuntos
Leucemia Mieloide Aguda , Multiômica , Humanos , Leucemia Mieloide Aguda/genética , Diferenciação Celular , Células-Tronco Neoplásicas/metabolismo
9.
J Biol Chem ; 286(32): 28210-22, 2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21540187

RESUMO

The cell cycle is driven by the kinase activity of cyclin·cyclin-dependent kinase (CDK) complexes, which is negatively regulated by CDK inhibitor proteins. Recently, we identified INCA1 as an interaction partner and a substrate of cyclin A1 in complex with CDK2. On a functional level, we identified a novel cyclin-binding site in the INCA1 protein. INCA1 inhibited CDK2 activity and cell proliferation. The inhibitory effects depended on the cyclin-interacting domain. Mitogenic and oncogenic signals suppressed INCA1 expression, whereas it was induced by cell cycle arrest. We established a deletional mouse model that showed increased CDK2 activity in spleen with altered spleen architecture in Inca1(-/-) mice. Inca1(-/-) embryonic fibroblasts showed an increase in the fraction of S-phase cells. Furthermore, blasts from acute lymphoid leukemia and acute myeloid leukemia patients expressed significantly reduced INCA1 levels highlighting its relevance for growth control in vivo. Taken together, this study identifies a novel CDK inhibitor with reduced expression in acute myeloid and lymphoid leukemia. The molecular events that control the cell cycle occur in a sequential process to ensure a tight regulation, which is important for the survival of a cell and includes the detection and repair of genetic damage and the prevention of uncontrolled cell division.


Assuntos
Crise Blástica/metabolismo , Proteínas de Transporte/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Fase S , Transdução de Sinais , Animais , Crise Blástica/genética , Crise Blástica/patologia , Proteínas de Transporte/genética , Linhagem Celular , Sobrevivência Celular/genética , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Fosfoproteínas/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia
10.
Oncogene ; 41(15): 2210-2224, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35220407

RESUMO

The small arginine-rich protein protamine condenses complete genomic DNA into the sperm head. Here, we applied its high RNA binding capacity for spontaneous electrostatic assembly of therapeutic nanoparticles decorated with tumour-cell-specific antibodies for efficiently targeting siRNA. Fluorescence microscopy and DLS measurements of these nanocarriers revealed the formation of a vesicular architecture that requires presence of antibody-protamine, defined excess of free SMCC-protamine, and anionic siRNA to form. Only these complex nanoparticles were efficient in the treatment of non-small-cell lung cancer (NSCLC) xenograft models, when the oncogene KRAS was targeted via EGFR-mediated delivery. To show general applicability, we used the modular platform for IGF1R-positive Ewing sarcomas. Anti-IGR1R-antibodies were integrated into an antibody-protamine nanoparticle with an siRNA specifically against the oncogenic translocation product EWS/FLI1. Using these nanoparticles, EWS/FLI1 knockdown blocked in vitro and in vivo growth of Ewing sarcoma cells. We conclude that these antibody-protamine-siRNA nanocarriers provide a novel platform technology to specifically target different cell types and yet undruggable targets in cancer therapy by RNAi.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Masculino , Proteínas de Fusão Oncogênica/genética , Protaminas/genética , Protaminas/metabolismo , Proteína Proto-Oncogênica c-fli-1/genética , RNA Interferente Pequeno/genética , Proteína EWS de Ligação a RNA/genética , Tecnologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Hematol Oncol ; 15(1): 171, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36457063

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is a fatal clonal hematopoietic malignancy, which results from the accumulation of several genetic aberrations in myeloid progenitor cells, with a worldwide 5-year survival prognosis of about 30%. Therefore, the development of more effective therapeutics with novel mode of action is urgently demanded. One common mutated gene in the AML is the DNA-methyltransferase DNMT3A whose function in the development and maintenance of AML is still unclear. To specifically target "undruggable" oncogenes, we initially invented an RNAi-based targeted therapy option that uses the internalization capacity of a colorectal cancer specific anti-EGFR-antibody bound to cationic protamine and the anionic siRNA. Here, we present a new experimental platform technology of molecular oncogene targeting in AML. METHODS: Our AML-targeting system consists of an internalizing anti-CD33-antibody-protamine conjugate, which together with anionic molecules such as siRNA or ibrutinib-Cy3.5 and cationic free protamine spontaneously assembles into vesicular nanocarriers in aqueous solution. These nanocarriers were analyzed concerning their physical properties and relevant characteristics in vitro in cell lines and in vivo in xenograft tumor models and patient-derived xenograft leukemia models with the aim to prepare them for translation into clinical application. RESULTS: The nanocarriers formed depend on a balanced electrostatic combination of the positively charged cationic protamine-conjugated anti-CD33 antibody, unbound cationic protamine and the anionic cargo. This nanocarrier transports its cargo safely into the AML target cells and has therapeutic activity against AML in vitro and in vivo. siRNAs directed specifically against two common mutated genes in the AML, the DNA-methyltransferase DNMT3A and FLT3-ITD lead to a reduction of clonal growth in vitro in AML cell lines and inhibit tumor growth in vivo in xenotransplanted cell lines. Moreover, oncogene knockdown of DNMT3A leads to increased survival of mice carrying leukemia patient-derived xenografts. Furthermore, an anionic derivative of the approved Bruton's kinase (BTK) inhibitor ibrutinib, ibrutinib-Cy3.5, is also transported by this nanocarrier into AML cells and decreases colony formation. CONCLUSIONS: We report important results toward innovative personalized, targeted treatment options via electrostatic nanocarrier therapy in AML.


Assuntos
Leucemia Mieloide Aguda , Protaminas , Humanos , Camundongos , Animais , Eletricidade Estática , RNA Interferente Pequeno/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Metiltransferases , DNA
12.
PLoS One ; 13(7): e0200163, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30001368

RESUMO

Precision cancer therapy requires on the one hand detailed knowledge about a tumor's driver oncogenes and on the other hand an effective targeted therapy that specifically inhibits these oncogenes. While the determination of genomic landscape of a tumor has reached a very precise level, the respective therapy options are scarce. The application of small inhibitory (si) RNAs is a promising field of investigation. Here, we present the effective in vivo-treatment of colorectal cancer (CRC) xenograft tumors with antibody-complexed, endoribonuclease-prepared small inhibitory (esi)RNAs. We chose heterogeneous endoribonuclease-prepared siRNA pools (esiRNAs) against the frequently mutated genes PIK3CA and KRAS and coupled them to the anti-EGFR antibody cetuximab, which was internalized specifically into the tumor cells. esiRNA pools have been shown to exhibit superior specificity in target gene knockdown compared to classic siRNAs. We identified a significant decrease in tumor growth upon this treatment due to decreased tumor cell proliferation. The ex vivo-analysis of the xenograft CRC tumors revealed the expected downregulation of the intended direct targets PIK3CA and KRAS on protein level. Moreover, known downstream targets for EGFR signaling such as p-ERK, p-AKT, and c-MYC were decreased as well. We therefore propose the use of antibody-esiRNA complexes as a novel experimental treatment option against key components of the EGFR signaling cascade.


Assuntos
Cetuximab/uso terapêutico , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Classe I de Fosfatidilinositol 3-Quinases/genética , Neoplasias Colorretais/terapia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Animais , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Terapia Combinada , Regulação para Baixo , Feminino , Células HT29 , Humanos , Camundongos , Camundongos Nus , Mutação , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Nat Protoc ; 11(1): 22-36, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26633129

RESUMO

Knockdown of genes by RNA interference (RNAi) in vitro requires methods of transfection or transduction, both of which have limited impact in vivo. As a virus-free approach, we chemically coupled cell surface receptors internalizing antibodies to the short interfering RNA (siRNA) carrier peptide protamine using the bispecific cross-linker sulfo-SMCC (sulfosuccinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate). First, protamine was conjugated amino-terminally to sulfo-SMCC, and then this conjugate was coupled via cysteine residues to the IgG backbone to carry siRNA. This complex can efficiently find, bind and internalize into receptor-positive cells in vitro and in vivo, which can be checked by flow cytometry, fluorescence microscopy and western blotting. This method obtains results similar to those of siRNA targeting molecules engineered by genetic fusions between receptor-binding and siRNA carrier units, with the advantage of using readily available purified proteins without the need for engineering, expression and purification of respective constructs. The procedure for coupling the complex takes ∼ 2 d, and the functional assays take ∼ 2 weeks.


Assuntos
Anticorpos/metabolismo , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Técnicas de Silenciamento de Genes/métodos , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Animais , Anticorpos/química , Anticorpos/imunologia , Linhagem Celular Tumoral , Receptores ErbB/imunologia , Humanos , Maleimidas/química , Camundongos , Protaminas/química , Protaminas/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/química , RNA Interferente Pequeno/metabolismo
14.
Oncotarget ; 7(6): 6774-89, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26735180

RESUMO

tTF-TAA and tTF-LTL are fusion proteins consisting of the extracellular domain of tissue factor (TF) and the peptides TAASGVRSMH and LTLRWVGLMS, respectively. These peptides represent ligands of NG2, a surface proteoglycan expressed on angiogenic pericytes and some tumor cells. Here we have expressed the model compound tTF-NGR, tTF-TAA, and tTF-LTL with different lengths in the TF domain in E. coli and used these fusion proteins for functional studies in anticancer therapy. We aimed to retarget TF to tumor vessels leading to tumor vessel infarction with two barriers of selectivity, a) the leaky endothelial lining in tumor vessels with the target NG2 being expressed on pericytes on the abluminal side of the endothelial cell barrier and b) the preferential expression of NG2 on angiogenic vessels such as in tumors. Chromatography-purified tTF-TAA showed identical Factor X (FX)-activating procoagulatory activity as the model compound tTF-NGR with Km values of approx. 0.15 nM in Michaelis-Menten kinetics. The procoagulatory activity of tTF-LTL varied with the chosen length of the TF part of the fusion protein. Flow cytometry revealed specific binding of tTF-TAA to NG2-expressing pericytes and tumor cells with low affinity and dissociation KD in the high nM range. In vivo and ex vivo fluorescence imaging of tumor xenograft-carrying animals and of the explanted tumors showed reduction of tumor blood flow upon tTF-TAA application. Therapeutic experiments showed a reproducible antitumor activity of tTF-TAA against NG2-expressing A549-tumor xenografts, however, with a rather small therapeutic window (active/toxic dose in mg/kg body weight).


Assuntos
Antígenos/metabolismo , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Oligopeptídeos/farmacologia , Proteoglicanas/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Tromboplastina/farmacologia , Animais , Antígenos/biossíntese , Linhagem Celular Tumoral , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Proteoglicanas/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
15.
PLoS One ; 10(6): e0129147, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26080083

RESUMO

The quest for novel therapeutic targets in acute myeloid leukemia (AML) is still ongoing. One of such targets, cyclin A1, was shown to be overexpressed in AML including AML stem cells. However, the function of cyclin A1 in AML is largely unknown, and the data on its impact on patients' survival remain controversial. Therefore, we developed a transgenic mouse model of stem cell-directed inducible cyclin A1 overexpression and crossed these mice with PML-RARα-knockin mice, which develop an AML M3-like phenotype. To observe the effects of cyclin A1 loss-of-function, we also crossed PML-RARα-knockin mice to cyclin A1-knockout mice. Neither overexpression nor loss of cyclin A1 significantly altered leukemogenesis in PML-RARα-knockin mice. These findings imply that upregulation of cyclin A1 is not essential for leukemogenesis. Our data suggest that cyclin A1 does not represent a suitable target for AML therapy.


Assuntos
Ciclina A1/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Animais , Regulação Leucêmica da Expressão Gênica , Técnicas de Introdução de Genes , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas de Fusão Oncogênica/genética , Regulação para Cima
16.
Clin Cancer Res ; 21(6): 1383-94, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25589625

RESUMO

PURPOSE: KRAS mutations are frequent driver mutations in multiple cancers. KRAS mutations also induce anti-EGFR antibody resistance in adenocarcinoma such as colon cancer. The aim of this study was to overcome anti-EGFR antibody resistance by coupling the antibody to KRAS-specific siRNA. EXPERIMENTAL DESIGN: The anti-EGFR antibody was chemically coupled to siRNA. The resulting complex was tested for antibody binding efficiency, serum stability and ability to deliver siRNA to EGFR-expressing cells. Western blotting, viability, apoptosis, and colony formation assays were performed for efficacy evaluation in vitro. Furthermore, therapeutic activity of the antibody-KRAS-siRNA complexes was examined in in vivo xenograft mouse tumor models. RESULTS: Antibody-siRNA complexes were targeted and internalized via the EGFR receptor. Upon internalization, target gene expression was strongly and specifically repressed, followed by a reduced proliferation and viability, and induced apoptosis of the cells in vitro. Clonogenic growth of mutant KRAS-bearing cells was suppressed by KRAS-siRNA-anti-EGFR antibody complexes. In xenograft mouse models, anti-EGFR antibody-KRAS-siRNA complexes significantly slowed tumor growth in anti-EGFR-resistant cells. CONCLUSIONS: The coupling of siRNA against KRAS to anti-EGFR antibodies provides a novel therapy approach for KRAS-mutated EGFR-positive cancer cells in vitro and in vivo. These findings provide an innovative approach for cancer-specific siRNA application and for enhanced therapeutic potential of monoclonal antibody therapy and personalized treatment of cancer entities.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , RNA Interferente Pequeno/uso terapêutico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Animais , Anticorpos Monoclonais/imunologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cetuximab/uso terapêutico , Neoplasias do Colo/genética , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Células HT29 , Humanos , Camundongos , Camundongos Nus , Protaminas/uso terapêutico , Proteínas Proto-Oncogênicas p21(ras)/imunologia , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto
17.
PLoS One ; 9(12): e115578, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25525809

RESUMO

Functional differences between healthy progenitor and cancer initiating cells may provide unique opportunities for targeted therapy approaches. Hematopoietic stem cells are tightly controlled by a network of CDK inhibitors that govern proliferation and prevent stem cell exhaustion. Loss of Inca1 led to an increased number of short-term hematopoietic stem cells in older mice, but Inca1 seems largely dispensable for normal hematopoiesis. On the other hand, Inca1-deficiency enhanced cell cycling upon cytotoxic stress and accelerated bone marrow exhaustion. Moreover, AML1-ETO9a-induced proliferation was not sustained in Inca1-deficient cells in vivo. As a consequence, leukemia induction and leukemia maintenance were severely impaired in Inca1-/- bone marrow cells. The re-initiation of leukemia was also significantly inhibited in absence of Inca1-/- in MLL-AF9- and c-myc/BCL2-positive leukemia mouse models. These findings indicate distinct functional properties of Inca1 in normal hematopoietic cells compared to leukemia initiating cells. Such functional differences might be used to design specific therapy approaches in leukemia.


Assuntos
Proteínas de Transporte/genética , Células-Tronco Hematopoéticas/metabolismo , Leucemia Experimental/patologia , Células-Tronco Neoplásicas/metabolismo , Animais , Proteínas de Transporte/metabolismo , Ciclo Celular , Células Cultivadas , Modelos Animais de Doenças , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Camundongos , Camundongos Knockout
18.
PLoS One ; 9(4): e94993, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24740120

RESUMO

External signals that are mediated by specific receptors determine stem cell fate. The thrombin receptor PAR1 plays an important role in haemostasis, thrombosis and vascular biology, but also in tumor biology and angiogenesis. Its expression and function in hematopoietic stem cells is largely unknown. Here, we analyzed expression and function of PAR1 in primary hematopoietic cells and their leukemic counterparts. AML patients' blast cells expressed much lower levels of PAR1 mRNA and protein than CD34+ progenitor cells. Constitutive Par1-deficiency in adult mice did not affect engraftment or stem cell potential of hematopoietic cells. To model an AML with Par1-deficiency, we retrovirally introduced the oncogene MLL-AF9 in wild type and Par1-/- hematopoietic progenitor cells. Par1-deficiency did not alter initial leukemia development. However, the loss of Par1 enhanced leukemic stem cell function in vitro and in vivo. Re-expression of PAR1 in Par1-/- leukemic stem cells delayed leukemogenesis in vivo. These data indicate that Par1 contributes to leukemic stem cell maintenance.


Assuntos
Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Leucemia/genética , Receptor PAR-1/genética , Doença Aguda , Animais , Hematopoese/genética , Humanos , Imuno-Histoquímica , Leucemia/metabolismo , Leucemia/patologia , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Receptor PAR-1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Blood ; 107(12): 4754-62, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16514057

RESUMO

VE-PTP, a receptor-type phosphotyrosine phosphatase, associates with the tyrosine kinase receptor Tie-2 and VE-cadherin and enhances the adhesive function of the latter. Here, VE-PTP was found to be restricted to endothelial cells, with a preference for arterial endothelium. Mutant mice expressing a truncated, secreted form of VE-PTP lacking the cytoplasmic and transmembrane domains and the most membrane-proximal extracellular fibronectin type III repeat, showed severe vascular malformations causing lethality at 10 days of gestation. Although blood vessels were initially formed, the intraembryonic vascular system soon deteriorated. Blood vessels in the yolk sac developed into dramatically enlarged cavities. In explant cultures of mutant allantoides, endothelial cells were found next to vessel structures growing as cell layers. No signs for enhanced endothelial apoptosis or proliferation were observed. Thus, the activity of VE-PTP is not required for the initial formation of blood vessels, yet it is essential for their maintenance and remodeling.


Assuntos
Vasos Sanguíneos/embriologia , Células Endoteliais/enzimologia , Neovascularização Fisiológica/fisiologia , Proteínas Tirosina Fosfatases/metabolismo , Saco Vitelino/irrigação sanguínea , Sequência de Aminoácidos/genética , Animais , Antígenos CD , Apoptose/genética , Vasos Sanguíneos/anormalidades , Caderinas/metabolismo , Proliferação de Células , Perda do Embrião/genética , Camundongos , Camundongos Mutantes , Estrutura Terciária de Proteína/genética , Proteínas Tirosina Fosfatases/genética , Receptor TIE-2/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores , Deleção de Sequência/genética , Saco Vitelino/anormalidades
20.
Archaea ; 1(1): 1-7, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15803653

RESUMO

Analysis of genome sequence data from the methanogenic archaeon Methanosarcina mazei Gö1 revealed the existence of two open reading frames encoding proton-translocating pyrophosphatases (PPases). These open reading frames are linked by a 750-bp intergenic region containing TC-rich stretches and are transcribed in opposite directions. The corresponding polypeptides are referred to as Mvp1 and Mvp2 and consist of 671 and 676 amino acids, respectively. Both enzymes represent extremely hydrophobic, integral membrane proteins with 15 predicted transmembrane segments and an overall amino acid sequence similarity of 50.1%. Multiple sequence alignments revealed that Mvp1 is closely related to eukaryotic PPases, whereas Mvp2 shows highest homologies to bacterial PPases. Northern blot experiments with RNA from methanol-grown cells harvested in the mid-log growth phase indicated that only Mvp2 was produced under these conditions. Analysis of washed membranes showed that Mvp2 had a specific activity of 0.34 U mg (protein)(-1). Proton translocation experiments with inverted membrane vesicles prepared from methanol-grown cells showed that hydrolysis of 1 mol of pyrophosphate was coupled to the translocation of about 1 mol of protons across the cytoplasmic membrane. Appropriate conditions for mvp1 expression could not be determined yet. The pyrophosphatases of M. mazei Gö1 represent the first examples of this enzyme class in methanogenic archaea and may be part of their energy-conserving system.


Assuntos
Pirofosfatase Inorgânica/metabolismo , Methanosarcina/enzimologia , Sequência de Aminoácidos , Proteínas Arqueais/isolamento & purificação , Proteínas Arqueais/metabolismo , Sequência de Bases , Membrana Celular/enzimologia , Sequência Conservada , DNA Arqueal/genética , DNA Arqueal/isolamento & purificação , Pirofosfatase Inorgânica/genética , Pirofosfatase Inorgânica/isolamento & purificação , Dados de Sequência Molecular , Conformação Proteica , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa