Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Materials (Basel) ; 16(8)2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-37110056

RESUMO

The influence of plasma-reduction treatment on iron and copper compounds at different oxidation states was investigated in this study. For this purpose, reduction experiments were carried out with artificially generated patina on metal sheets and with metal salt crystals of iron(II) sulfate (FeSO4), iron(III) chloride (FeCl3), and copper(II) chloride (CuCl2), as well as with the metal salt thin films of these compounds. All the experiments were carried out under cold low-pressure microwave plasma conditions; the main focus was on plasma reduction at a low pressure in order to evaluate an implementable process in a parylene-coating device. Usually, plasma is used within the parylene-coating process as a supporting tool for adhesion improvement and micro-cleaning efforts. This article offers another useful application for implementing plasma treatment as a reactive medium in order to apply different functionalities by an alteration in the oxidation state. The effect of microwave plasmas on metal surfaces and metal composite materials has been widely studied. In contrast, this work deals with metal salt surfaces generated from a solution and the influence of microwave plasma on metal chlorides and sulfates. While the plasma reduction of metal compounds commonly succeeds with hydrogen-containing plasmas at high temperatures, this study shows a new reduction process that reduces iron salts at temperatures between 30 and 50 °C. A novelty of this study is the alteration in the redox state of the base and noble metal materials within a parylene-coating device with the help of an implemented microwave generator. Another novelty of this study is treating metal salt thin layers for reduction purposes in order to provide the opportunity to include subsequent coating experiments to create parylene metal multilayers. Another new aspect of this study is the adapted reduction process of thin metal salt layers consisting of either noble or base metals, with an air plasma pre-treatment prior to the hydrogen-containing plasma-reduction procedure.

2.
Science ; 376(6599): 1343-1347, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35709278

RESUMO

Effective tissue repair after myocardial infarction entails a vigorous angiogenic response, guided by incompletely defined immune cell-endothelial cell interactions. We identify the monocyte- and macrophage-derived cytokine METRNL (meteorin-like) as a driver of postinfarction angiogenesis and high-affinity ligand for the stem cell factor receptor KIT (KIT receptor tyrosine kinase). METRNL mediated angiogenic effects in cultured human endothelial cells through KIT-dependent signaling pathways. In a mouse model of myocardial infarction, METRNL promoted infarct repair by selectively expanding the KIT-expressing endothelial cell population in the infarct border zone. Metrnl-deficient mice failed to mount this KIT-dependent angiogenic response and developed severe postinfarction heart failure. Our data establish METRNL as a KIT receptor ligand in the context of ischemic tissue repair.


Assuntos
Adipocinas , Citocinas , Infarto do Miocárdio , Neovascularização Fisiológica , Fatores de Crescimento Neural , Proteínas Proto-Oncogênicas c-kit , Animais , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Células Endoteliais/metabolismo , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/genética , Ligantes , Macrófagos/metabolismo , Camundongos , Camundongos Mutantes , Infarto do Miocárdio/complicações , Infarto do Miocárdio/fisiopatologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo
3.
Cells ; 11(6)2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35326445

RESUMO

Chimeric antigen receptor (CAR) T-cell therapies have shown impressive results in patients with hematological malignancies; however, little success has been achieved in the treatment of solid tumors. Recently, macrophages (MΦs) were identified as an additional candidate for the CAR approach, and initial proof of concept studies using peripheral blood-derived monocytes showed antigen-redirected activation of CAR MΦs. However, some patients may not be suitable for monocyte-apheresis, and prior cancer treatment regimens may negatively affect immune cell number and functionality. To address this problem, we here introduce primary human hematopoietic stem and progenitor cells (HSPCs) as a cell source to generate functional CAR MΦs ex vivo. Our data showed successful CAR expression in cord blood (CB)-derived HSPCs, with considerable cell expansion during differentiation to CAR MΦs. HSPC-derived MΦs showed typical MΦ morphology, phenotype, and basic anti-bacterial functionality. CAR MΦs targeting the carcinoembryonic antigen (CEA) and containing either a DAP12- or a CD3ζ-derived signaling domain showed antigen redirected activation as they secreted pro-inflammatory cytokines specifically upon contact with CEA+ target cells. In addition, CD3ζ-expressing CAR MΦs exhibited significantly enhanced phagocytosis of CEA+ HT1080 cells. Our data establish human HSPCs as a suitable cell source to generate functional CAR MΦs and further support the use of CAR MΦs in the context of solid tumor therapy.


Assuntos
Antígeno Carcinoembrionário , Neoplasias , Antígeno Carcinoembrionário/metabolismo , Citocinas/metabolismo , Humanos , Imunoterapia Adotiva/métodos , Macrófagos/metabolismo , Neoplasias/metabolismo , Células-Tronco/metabolismo
4.
Mol Ther Nucleic Acids ; 27: 810-823, 2022 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-35141043

RESUMO

DNA-modifying technologies, such as the CRISPR-Cas9 system, are promising tools in the field of gene and cell therapies. However, high and prolonged expression of DNA-modifying enzymes may cause cytotoxic and genotoxic side effects and is therefore unwanted in therapeutic approaches. Consequently, development of new and potent short-term delivery methods is of utmost importance. Recently, we developed non-integrating gammaretrovirus- and MS2 bacteriophage-based Gag.MS2 (g.Gag.MS2) particles for transient transfer of non-retroviral CRISPR-Cas9 RNA into target cells. In the present study, we further improved the technique by transferring the system to the alpharetroviral vector platform (a.Gag.MS2), which significantly increased CRISPR-Cas9 delivery into target cells and allowed efficient targeted knockout of endogenous TP53/Trp53 genes in primary murine fibroblasts as well as primary human fibroblasts, hepatocytes, and cord-blood-derived CD34+ stem and progenitor cells. Strikingly, co-packaging of Cas9 mRNA and multiple single guide RNAs (sgRNAs) into a.Gag.MS2 chimera displayed efficient targeted knockout of up to three genes. Co-transfection of single-stranded DNA donor oligonucleotides during CRISPR-Cas9 particle production generated all-in-one particles, which mediated up to 12.5% of homology-directed repair in primary cell cultures. In summary, optimized a.Gag.MS2 particles represent a versatile tool for short-term delivery of DNA-modifying enzymes into a variety of target cells, including primary murine and human cells.

5.
Women Health ; 62(2): 144-156, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35068353

RESUMO

Invasive lobular carcinoma (ILC) of the breast is known to have typical molecular, clinical, and pathological characteristics that differ from invasive cancer of no special type (NST). In the German mammography screening program (MSP), we evaluated clinical differences between these tumor types at the time of their detection. Clinical features of NSTs (n = 785) and ILCs (n = 141) diagnosed in the MSP between 2009 and 2016 were compared. Compared to NST, ILC was significantly correlated with advanced age (59.1 years versus 60.6 years) and larger tumor size (1.5 cm versus 2.3 cm). ILC was significantly more frequently associated with moderate tumor differentiation (G2), whereas NST was associated with a higher rate of poorly differentiated tumors (p < .001). Furthermore, ILC presented more often as multifocal tumors (36% versus 11%, p < .001), and mastectomies were performed more often in the ILC group (27% versus 12%, p < .001). ILCs and NSTs had different clinical features at the time of detection. The pathological profile of ILC may explain some of these features. Specialists should be aware of the fact that ILC may escape detection by conventional imaging modalities for a long time, and may present later in life as more advanced multifocal disease.


Assuntos
Neoplasias da Mama , Carcinoma Lobular , Mama/diagnóstico por imagem , Neoplasias da Mama/diagnóstico por imagem , Carcinoma Lobular/diagnóstico por imagem , Carcinoma Lobular/patologia , Detecção Precoce de Câncer , Feminino , Humanos , Mamografia , Pessoa de Meia-Idade , Estudos Retrospectivos
6.
Front Immunol ; 12: 751138, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34804035

RESUMO

Immune cell therapeutics are increasingly applied in oncology. Especially chimeric antigen receptor (CAR) T cells are successfully used to treat several B cell malignancies. Efforts to engineer CAR T cells for improved activity against solid tumors include co-delivery of pro-inflammatory cytokines in addition to CARs, via either constitutive cytokine expression or inducible cytokine expression triggered by CAR recognition of its target antigen-so-called "T cells redirected for universal cytokine-mediated killing" (TRUCKs) or fourth-generation CARs. Here, we tested the hypothesis that TRUCK principles could be expanded to improve anticancer functions of NK cells. A comparison of the functionality of inducible promoters responsive to NFAT or NFκB in NK cells showed that, in contrast to T cells, the inclusion of NFκB-responsive elements within the inducible promoter construct was essential for CAR-inducible expression of the transgene. We demonstrated that GD2CAR-specific activation induced a tight NFκB-promoter-driven cytokine release in NK-92 and primary NK cells together with an enhanced cytotoxic capacity against GD2+ target cells, also shown by increased secretion of cytolytic cytokines. The data demonstrate biologically relevant differences between T and NK cells that are important when clinically translating the TRUCK concept to NK cells for the treatment of solid malignancies.


Assuntos
Imunoterapia Adotiva , Células Matadoras Naturais/imunologia , NF-kappa B/genética , Alpharetrovirus/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Linhagem Celular , Movimento Celular , Técnicas de Cocultura , Citocinas/imunologia , Vetores Genéticos , Glioblastoma/imunologia , Glioblastoma/terapia , Humanos , NF-kappa B/imunologia , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/imunologia
7.
Rofo ; 192(7): 678-685, 2020 Jul.
Artigo em Inglês, Alemão | MEDLINE | ID: mdl-32106324

RESUMO

PURPOSE: Analysis of the influence of the singular risk factors age and breast density on the 2-year incidence of breast cancer among participants in the German mammography screening program. MATERIALS AND METHODS: The multicenter study includes 111 456 subsequent round digital mammographic screening examinations from four screening units with prospective visual categorization of breast density. Based on detection in screening and during the 2-year interval after negative screening participation (interval cancers), 2-year breast cancer incidences (2 YBCI) (‰) were calculated in the 5-year age groups (5 YAG) of the target group 50-69 years and in the BI-RADS density categories ACR 1-4. Multivariate statistical evaluations were carried out using logistic regression models. RESULTS: With an increase in the 5 YAG, the 2 YBCI increased by 5.0 ‰, 6.7 ‰, 8.5 ‰ to 9.7 ‰, and was significantly different among 55-59, 60-64 and 65-69-year-old women compared to the youngest reference group 50-54 years (odds ratio (OR): 1.34; 1.68; and 1.93; p-value < 0.0001). With an increase in density categories 1-4, the 2 YBCI increased from 2.6 ‰, to 5.8 ‰, 9.6 ‰, and 9.7 ‰. The 2 YBCI differed significantly in breast density categories 2, 3, 4 from reference group 1 (OR: 2.17; 3.65; and 3.76; p-value < 0.0001). Only within the two main breast density groups 2 (frequency 44.3 %) and 3 (44.7 %), a significant increase in the 2 YBCI was observed across the 5 YAG (category 2: 3.7-8.9 ‰; category 3: 5.8-11.7 ‰; p-value < 0.001 each). The 2 YBCI was above the median of 7.5 ‰ in women with breast density category 2 and aged 65-69 years, as well as in women with breast density categories 3 and 4 aged 55-69 years. A 2 YBCI below the median was seen in women between 50-54 years regardless of breast density, as well as women in category 1 in all age groups. CONCLUSION: Within the main breast density categories 2 and 3 (almost 90 % of participants), incidences increase with age to double. A consistently low incidence is found regardless of breast density at a young screening age and in women with the lowest breast density. KEY POINTS: · The risk of breast cancer is modified by age in density categories.. · Women aged 50-54 years have a low risk in all density categories.. · Women in category ACR 1 of any age group have a low risk.. CITATION FORMAT: · Weigel S, Heindel W, Dietz C et al. Stratifizierung des Brustkrebsrisikos hinsichtlich der Einflüsse von Alter und mammografischer Dichte. Fortschr Röntgenstr 2020; 192: 678 - 685.


Assuntos
Densidade da Mama , Neoplasias da Mama/diagnóstico por imagem , Medição de Risco , Fatores Etários , Idoso , Neoplasias da Mama/classificação , Feminino , Humanos , Pessoa de Meia-Idade , Estudos Prospectivos
8.
Cancers (Basel) ; 12(2)2020 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-32041222

RESUMO

Genetically modified T cells expressing chimeric antigen receptors (CARs) so far have mostly failed in the treatment of solid tumors owing to a number of limitations, including an immunosuppressive tumor microenvironment and insufficient CAR T cell activation and persistence. Next-generation approaches using CAR T cells that secrete transgenic immunomodulatory cytokines upon CAR signaling, known as TRUCKs ("T cells redirected for universal cytokine-mediated killing"), are currently being explored. As TRUCKs were engineered by the transduction of T cells with two separate vectors, we developed a lentiviral modular "all-in-one" vector system that combines constitutive CAR expression and inducible nuclear factor of activated T cells (NFAT)-driven transgene expression for more efficient production of TRUCKs. Activation of the GD2-specific CAR via GD2+ target cells induced NFAT promoter-driven cytokine release in primary human T cells, and indicated a tight linkage of CAR-specific activation and transgene expression that was further improved by a modified NFATsyn promoter. As proof-of-concept, we showed that T cells containing the "all-in-one" vector system secrete the immunomodulatory cytokines interleukin (IL)12 or IL18 upon co-cultivation with primary human GD2+ tumor cells, resulting in enhanced effector cell properties and increased monocyte recruitment. This highlights the potential of our system to simplify application of TRUCK-modified T cells in solid tumor therapy.

9.
J Clin Invest ; 129(12): 5108-5122, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31479431

RESUMO

The transcription factor B cell CLL/lymphoma 11B (BCL11B) is indispensable for T lineage development of lymphoid progenitors. Here, we show that chimeric antigen receptor (CAR) expression during early phases of ex vivo generation of lymphoid progenitors suppressed BCL11B, leading to suppression of T cell-associated gene expression and acquisition of NK cell-like properties. Upon adoptive transfer into hematopoietic stem cell transplant recipients, CAR-expressing lymphoid progenitors differentiated into CAR-induced killer (CARiK) cells that mediated potent antigen-directed antileukemic activity even across MHC barriers. CD28 and active immunoreceptor tyrosine-based activation motifs were critical for a functional CARiK phenotype. These results give important insights into differentiation of murine and human lymphoid progenitors driven by synthetic CAR transgene expression and encourage further evaluation of ex vivo-generated CARiK cells for targeted immunotherapy.


Assuntos
Antígenos CD28/metabolismo , Células Matadoras Naturais/citologia , Linfócitos/citologia , Receptores de Antígenos Quiméricos/metabolismo , Proteínas Repressoras/metabolismo , Linfócitos T/citologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Antígenos CD19/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Separação Celular , Citotoxicidade Imunológica , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunoterapia , Imunoterapia Adotiva , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Engenharia de Proteínas , Células-Tronco/citologia , Transgenes
10.
Mol Ther ; 27(9): 1597-1611, 2019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31326401

RESUMO

Hereditary pulmonary alveolar proteinosis (PAP) is a genetic lung disease characterized by surfactant accumulation and respiratory failure arising from disruption of GM-CSF signaling. While mutations in either CSF2RA or CSF2RB (encoding GM-CSF receptor α or ß chains, respectively) can cause PAP, α chain mutations are responsible in most patients. Pulmonary macrophage transplantation (PMT) is a promising new cell therapy in development; however, no studies have evaluated this approach for hereditary PAP (hPAP) caused by Csf2ra mutations. Here, we report on the preclinical safety, tolerability, and efficacy of lentiviral-vector (LV)-mediated Csf2ra expression in macrophages and PMT of gene-corrected macrophages (gene-PMT therapy) in Csf2ra gene-ablated (Csf2ra-/-) mice. Gene-PMT therapy resulted in a stable transgene integration and correction of GM-CSF signaling and functions in Csf2ra-/- macrophages in vitro and in vivo and resulted in engraftment and long-term persistence of gene-corrected macrophages in alveoli; restoration of pulmonary surfactant homeostasis; correction of PAP-specific cytologic, histologic, and biomarker abnormalities; and reduced inflammation associated with disease progression in untreated mice. No adverse consequences of gene-PMT therapy in Csf2ra-/- mice were observed. Results demonstrate that gene-PMT therapy of hPAP in Csf2ra-/- mice was highly efficacious, durable, safe, and well tolerated.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/transplante , Proteinose Alveolar Pulmonar/genética , Proteinose Alveolar Pulmonar/terapia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Animais , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos/métodos , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/genética , Imunofenotipagem , Lentivirus/genética , Camundongos , Camundongos Knockout , Proteinose Alveolar Pulmonar/diagnóstico , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Transdução de Sinais , Transdução Genética
11.
Hum Gene Ther ; 30(4): 381-401, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30734584

RESUMO

The introduction of chimeric antigen receptors (CARs) to augment the anticancer activity of immune cells represents one of the major clinical advances in recent years. This work demonstrates that sorted CAR natural killer (NK) cells have improved antileukemia activity compared to control NK cells that lack a functional CAR. However, in terms of viability, effectiveness, risk of side effects, and clinical practicality and applicability, an important question is whether gene-modified NK cell lines represent better CAR effector cells than primary human donor CAR-NK (CAR-dNK) cells. Comparison of the functional activities of sorted CAR-NK cells generated using the NK-92 cell line with those generated from primary human dNK cells demonstrated that CAR-NK-92 cells had stronger cytotoxic activity against leukemia cells compared to CAR-dNK cells. CAR-NK-92 and CAR-dNK cells had similar CD107a surface expression upon co-incubation with leukemia cells. However, CAR-NK-92 cells secreted higher granzyme A and interleukin-17A levels, while CAR-dNK cells secreted more tumor necrosis factor alpha, interferon gamma, and granulysin. In addition, CAR-NK-92 cells revealed a significantly higher potential for adverse side effects against nonmalignant cells. In short, this work shows the feasibility for further development of CAR-NK strategies to treat leukemia.


Assuntos
Imunoterapia Adotiva , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Leucemia Mieloide Aguda/etiologia , Leucemia Mieloide Aguda/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Alpharetrovirus/genética , Animais , Biomarcadores , Biomarcadores Tumorais , Comunicação Celular/imunologia , Degranulação Celular/imunologia , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Citotoxicidade Imunológica , Modelos Animais de Doenças , Expressão Gênica , Vetores Genéticos/genética , Humanos , Imunofenotipagem , Imunoterapia Adotiva/métodos , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/terapia , Masculino , Camundongos , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Especificidade do Receptor de Antígeno de Linfócitos T , Transgenes
12.
Mol Ther Nucleic Acids ; 13: 256-274, 2018 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-30317165

RESUMO

The recently discovered CRISPR/Cas9 system is widely used in basic research and is a useful tool for disease modeling and gene editing therapies. However, long-term expression of DNA-modifying enzymes can be associated with cytotoxicity and is particularly unwanted in clinical gene editing strategies. Because current transient expression methods may still suffer from cytotoxicity and/or low efficiency, we developed non-integrating retrovirus-based CRISPR/Cas9 all-in-one particles for targeted gene knockout. By redirecting the gammaretroviral packaging machinery, we transiently delivered Streptococcus pyogenes Cas9 (SpCas9) mRNA and single-guide RNA transcripts into various (including primary) cell types. Spatiotemporal co-delivery of CRISPR/Cas9 components resulted in efficient disruption of a surrogate reporter gene, as well as functional knockout of endogenous human genes CXCR4 and TP53. Although acting in a hit-and-run fashion, knockout efficiencies of our transient particles corresponded to 52%-80% of those obtained from constitutively active integrating vectors. Stable SpCas9 overexpression at high doses in murine NIH3T3 cells caused a substantial G0/G1 arrest accompanied by reduced cell growth and metabolic activity, which was prevented by transient SpCas9 transfer. In summary, the non-integrating retrovirus-based vector particles introduced here allow efficient and dose-controlled delivery of CRISPR/Cas9 components into target cells.

13.
Hum Gene Ther Methods ; 28(6): 318-329, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28854814

RESUMO

Hereditary pulmonary alveolar proteinosis (hPAP) is a rare disorder of pulmonary surfactant accumulation and hypoxemic respiratory failure caused by mutations in CSF2RA (encoding the granulocyte/macrophage colony-stimulating factor [GM-CSF] receptor α-chain [CD116]), which results in reduced GM-CSF-dependent pulmonary surfactant clearance by alveolar macrophages. While no pharmacologic therapy currently exists for hPAP, it was recently demonstrated that endotracheal instillation of wild-type or gene-corrected mononuclear phagocytes (pulmonary macrophage transplantation [PMT]) results in a significant and durable therapeutic efficacy in a validated murine model of hPAP. To facilitate the translation of PMT therapy to human hPAP patients, a self-inactivating (SIN) lentiviral vector was generated expressing a codon-optimized human CSF2RA-cDNA driven from an EF1α short promoter (Lv.EFS.CSF2RAcoop), and a series of nonclinical efficacy and safety studies were performed in cultured macrophage cell lines and primary human cells. Studies in cytokine-dependent Ba/F3 cells demonstrated efficient transduction, vector-derived CD116 expression proportional to vector copy number, and GM-CSF-dependent cell survival and proliferation. Using a novel cell line constructed to express a normal GM-CSF receptor ß subunit and a dysfunctional α subunit (due to a function-altering CSF2RAG196R mutation) that reflects the macrophage disease phenotype of hPAP patients, it was demonstrated that Lv.EFS.CSF2RAcoop transduction restored GM-CSF receptor function. Further, Lv.EFS.CSF2RAcoop transduction of healthy primary CD34+ cells did not adversely affect cell proliferation or affect the cell differentiation program. Results demonstrate Lv.EFS.CSF2RAcoop reconstituted GM-CSF receptor α expression, restoring GM-CSF signaling in hPAP macrophages, and had no adverse effects in the intended target cells, thus supporting testing of PMT therapy of hPAP in humans.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/genética , Lentivirus/genética , Proteinose Alveolar Pulmonar/congênito , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Transdução Genética/métodos , Animais , Células Cultivadas , Terapia Genética/efeitos adversos , Células HEK293 , Humanos , Macrófagos/metabolismo , Camundongos , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Proteinose Alveolar Pulmonar/terapia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo
14.
Biochim Biophys Acta ; 1849(9): 1145-54, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26170143

RESUMO

The heteromeric transcription factor GA-binding protein (GABP) consists of two subunits, the alpha subunit (GABPA) carrying the DNA-binding ETS domain, and the beta subunit (GABPB1) harbouring the transcriptional activation domain. GABP is involved in haematopoietic stem cell maintenance and differentiation of myeloid and lymphoid lineages in mice. To elucidate the molecular function of GABP in human haematopoiesis, the present study addressed effects of ectopic overexpression of GABP focussing on the myeloid compartment. Combined overexpression of GABPA and GABPB1 caused a proliferation block in cell lines and drastically reduced the colony-forming capacity of murine lineage-negative cells. Impaired proliferation resulted from perturbed cellular cycling and induction of myeloid differentiation shown by surface markers and myelomonocytic morphology of U937 cells. Depending on the dosage and functional integrity of GABP, ITGAM expression was induced. ITGAM encodes CD11b, the alpha subunit of integrin Mac-1, whose beta subunit, ITGB2/CD18, was already described to be regulated by GABP. Finally, Shield1-dependent proteotuning, luciferase reporter assays and chromatin immunoprecipitation showed that GABP activates the ITGAM/CD11b promoter via three binding sites close to the translational start site. In conclusion, the present study supports the crucial role of GABP in myeloid cell differentiation and identified ITGAM/CD11b as a novel GABP target gene.


Assuntos
Antígeno CD11b/genética , Diferenciação Celular/fisiologia , Fator de Transcrição de Proteínas de Ligação GA/fisiologia , Células Mieloides/citologia , Regiões Promotoras Genéticas , Animais , Linhagem Celular , Fator de Transcrição de Proteínas de Ligação GA/genética , Dosagem de Genes , Humanos , Camundongos
15.
Stem Cell Reports ; 2(1): 107-18, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-24678453

RESUMO

Genetic engineering of human induced pluripotent stem cells (hiPSCs) via customized designer nucleases has been shown to be significantly more efficient than conventional gene targeting, but still typically depends on the introduction of additional genetic selection elements. In our study, we demonstrate the efficient nonviral and selection-independent gene targeting in human pluripotent stem cells (hPSCs). Our high efficiencies of up to 1.6% of gene-targeted hiPSCs, accompanied by a low background of randomly inserted transgenes, eliminated the need for antibiotic or fluorescence-activated cell sorting selection, and allowed the use of short donor oligonucleotides for footprintless gene editing. Gene-targeted hiPSC clones were established simply by direct PCR screening. This optimized approach allows targeted transgene integration into safe harbor sites for more predictable and robust expression and enables the straightforward generation of disease-corrected, patient-derived iPSC lines for research purposes and, ultimately, for future clinical applications.


Assuntos
Endonucleases/metabolismo , Recombinação Homóloga , Células-Tronco Pluripotentes/metabolismo , Células Cultivadas , Reparo do DNA por Junção de Extremidades , Técnicas de Inativação de Genes , Marcação de Genes , Loci Gênicos , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Oligodesoxirribonucleotídeos/metabolismo , Células-Tronco Pluripotentes/citologia , Reação em Cadeia da Polimerase
16.
Blood ; 123(16): 2550-61, 2014 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-24394665

RESUMO

The transcription factor lymphoid enhancer-binding factor 1 (LEF-1), which plays a definitive role in granulocyte colony-stimulating factor (G-CSF) receptor-triggered granulopoiesis, is downregulated in granulocytic progenitors of severe congenital neutropenia (CN) patients. However, the exact mechanism of LEF-1 downregulation is unclear. CN patients are responsive to therapeutically high doses of G-CSF and are at increased risk of developing acute myeloid leukemia. The normal expression of LEF-1 in monocytes and lymphocytes, whose differentiation is unaffected in CN, suggests the presence of a granulopoiesis-specific mechanism downstream of G-CSF receptor signaling that leads to LEF-1 downregulation. Signal transducer and activator of transcription 5 (STAT5) is activated by G-CSF and is hyperactivated in acute myeloid leukemia. Here, we investigated the effects of activated STAT5 on LEF-1 expression and functions in hematopoietic progenitor cells. We demonstrated that constitutively active STAT5a (caSTAT5a) inhibited LEF-1-dependent autoregulation of the LEF-1 gene promoter by binding to the LEF-1 protein, recruiting Nemo-like kinase and the E3 ubiquitin-ligase NARF to LEF-1, leading to LEF-1 ubiquitination and a reduction in LEF-1 protein levels. The proteasome inhibitor bortezomib reversed the defective G-CSF-triggered granulocytic differentiation of CD34(+) cells from CN patients in vitro, an effect that was accompanied by restoration of LEF-1 protein levels and LEF-1 messenger RNA autoregulation. Taken together, our data define a novel mechanism of LEF-1 downregulation in CN patients via enhanced ubiquitination and degradation of LEF-1 protein by hyperactivated STAT5.


Assuntos
Ácidos Borônicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Granulócitos/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Neutropenia/congênito , Proteólise/efeitos dos fármacos , Pirazinas/farmacologia , Antígenos CD34/metabolismo , Bortezomib , Diferenciação Celular/genética , Células Cultivadas , Síndrome Congênita de Insuficiência da Medula Óssea , Granulócitos/patologia , Granulócitos/fisiologia , Células HEK293 , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Humanos , Fator 1 de Ligação ao Facilitador Linfoide/genética , Neutropenia/genética , Neutropenia/metabolismo , Neutropenia/patologia , Fator de Transcrição STAT5/fisiologia
17.
Blood ; 122(16): 2877-87, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-23954893

RESUMO

Mutations in the metabolic enzymes isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) are frequently found in glioma, acute myeloid leukemia (AML), melanoma, thyroid cancer, and chondrosarcoma patients. Mutant IDH produces 2-hydroxyglutarate (2HG), which induces histone- and DNA-hypermethylation through inhibition of epigenetic regulators. We investigated the role of mutant IDH1 using the mouse transplantation assay. Mutant IDH1 alone did not transform hematopoietic cells during 5 months of observation. However, mutant IDH1 greatly accelerated onset of myeloproliferative disease-like myeloid leukemia in mice in cooperation with HoxA9 with a mean latency of 83 days compared with cells expressing HoxA9 and wild-type IDH1 or a control vector (167 and 210 days, respectively, P = .001). Mutant IDH1 accelerated cell-cycle transition through repression of cyclin-dependent kinase inhibitors Cdkn2a and Cdkn2b, and activated mitogen-activated protein kinase signaling. By computational screening, we identified an inhibitor of mutant IDH1, which inhibited mutant IDH1 cells and lowered 2HG levels in vitro, and efficiently blocked colony formation of AML cells from IDH1-mutated patients but not of normal CD34(+) bone marrow cells. These data demonstrate that mutant IDH1 has oncogenic activity in vivo and suggest that it is a promising therapeutic target in human AML cells.


Assuntos
Regulação Leucêmica da Expressão Gênica , Isocitrato Desidrogenase/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutação , Adolescente , Adulto , Animais , Antígenos CD34/metabolismo , Apoptose , Transplante de Medula Óssea , Ciclo Celular , Feminino , Humanos , Isocitrato Desidrogenase/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Adulto Jovem
18.
J Biol Chem ; 286(26): 22716-29, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21558273

RESUMO

Monocytic differentiation is orchestrated by complex networks that are not fully understood. This study further elucidates the involvement of transcription factor CCAAT/enhancer-binding protein ß (C/EBPß). Initially, we demonstrated a marked increase in nuclear C/EBPß-liver-enriched activating protein* (LAP*)/liver-enriched activating protein (LAP) levels and LAP/liver-enriched inhibiting protein (LIP) ratios in phorbol 12-myristate 13-acetate (PMA)-treated differentiating THP-1 premonocytic cells accompanied by reduced proliferation. To directly study C/EBPß effects on monocytic cells, we generated novel THP-1-derived (low endogenous C/EBPß) cell lines stably overexpressing C/EBPß isoforms. Most importantly, cells predominantly overexpressing LAP* (C/EBPß-long), but not those overexpressing LIP (C/EBPß-short), exhibited a reduced proliferation, with no effect on morphology. PMA-induced inhibition of proliferation was attenuated in C/EBPß-short cells. In C/EBPß(WT) macrophage-like cells (high endogenous C/EBPß), we measured a reduced proliferation/cycling index compared with C/EBPß(KO). The typical macrophage morphology was only observed in C/EBPß(WT), whereas C/EBPß(KO) stayed round. C/EBPα did not compensate for C/EBPß effects on proliferation/morphology. Serum reduction, an independent approach known to inhibit proliferation, induced macrophage morphology in C/EBPß(KO) macrophage-like cells but not THP-1. In PMA-treated THP-1 and C/EBPß-long cells, a reduced phosphorylation of cell cycle repressor retinoblastoma was found. In addition, C/EBPß-long cells showed reduced c-Myc expression accompanied by increased CDK inhibitor p27 and reduced cyclin D1 levels. Finally, C/EBPß-long and C/EBPß(WT) cells exhibited low E2F1 and cyclin E levels, and C/EBPß overexpression was found to inhibit cyclin E1 promoter-dependent transcription. Our results suggest that C/EBPß reduces monocytic proliferation by affecting the retinoblastoma/E2F/cyclin E pathway and that it may contribute to, but is not directly required for, macrophage morphology. Inhibition of proliferation by C/EBPß may be important for coordinated monocytic differentiation.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proliferação de Células , Ciclina E/metabolismo , Fator de Transcrição E2F1/metabolismo , Monócitos/metabolismo , Proteínas Oncogênicas/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Carcinógenos/farmacologia , Linhagem Celular , Ciclina E/genética , Fator de Transcrição E2F1/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Macrófagos , Camundongos , Monócitos/citologia , Proteínas Oncogênicas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína do Retinoblastoma/genética , Acetato de Tetradecanoilforbol/farmacologia
19.
Blood ; 117(14): 3737-47, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21289307

RESUMO

Thpo/Mpl signaling plays an important role in the maintenance of hematopoietic stem cells (HSCs) in addition to its role in megakaryopoiesis. Patients with inactivating mutations in Mpl develop thrombocytopenia and aplastic anemia because of progressive loss of HSCs. Yet, it is unknown whether this loss of HSCs is an irreversible process. In this study, we used the Mpl knockout (Mpl(-/-)) mouse model and expressed Mpl from newly developed lentiviral vectors specifically in the physiologic Mpl target populations, namely, HSCs and megakaryocytes. After validating lineage-specific expression in vivo using lentiviral eGFP reporter vectors, we performed bone marrow transplantation of transduced Mpl(-/-) bone marrow cells into Mpl(-/-) mice. We show that restoration of Mpl expression from transcriptionally targeted vectors prevents lethal adverse reactions of ectopic Mpl expression, replenishes the HSC pool, restores stem cell properties, and corrects platelet production. In some mice, megakaryocyte counts were atypically high, accompanied by bone neo-formation and marrow fibrosis. Gene-corrected Mpl(-/-) cells had increased long-term repopulating potential, with a marked increase in lineage(-)Sca1(+)cKit(+) cells and early progenitor populations in reconstituted mice. Transcriptome analysis of lineage(-)Sca1(+)cKit(+) cells in Mpl-corrected mice showed functional adjustment of genes involved in HSC self-renewal.


Assuntos
Anemia Aplástica/genética , Anemia Aplástica/terapia , Técnicas de Transferência de Genes , Células-Tronco Hematopoéticas/fisiologia , Lentivirus/genética , Receptores de Trombopoetina/genética , Regeneração/genética , Anemia Aplástica/patologia , Anemia Aplástica/fisiopatologia , Animais , Linhagem da Célula/genética , Células Cultivadas , Modelos Animais de Doenças , Terapia Genética/métodos , Vetores Genéticos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Receptores de Trombopoetina/metabolismo , Receptores de Trombopoetina/fisiologia
20.
J Biotechnol ; 152(1-2): 1-8, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21262286

RESUMO

Stem cell factor (SCF) known as the c-kit ligand, plays important roles in spermatogenesis, melanogenesis and early stages of hematopoiesis. As for the latter, SCF is essential for growth and expansion of hematopoietic stem and progenitor cells. We herein describe the production of recombinant murine SCF from Escherichia coli as soluble thioredoxin-fusion protein. The formation of insoluble and inactive inclusion bodies, usually observed when SCF is expressed in E. coli, was almost entirely prevented. After purification based on membrane adsorber technology, the fusion protein was subsequently cleaved by TEV protease in order to release mature mSCF. Following dialysis and a final purification step, the target protein was isolated in high purity. Bioactivity of mSCF was proven by different tests (MTT analogous assay, long-term proliferation assay) applying a human megakaryocytic cell line. Furthermore, the biological activity of the uncleaved fusion protein was tested as well. We observed a significant activity, even though it was less than the activity displayed by the purified mSCF. In summary, avoiding inclusion body formation we present an efficient production procedure for mSCF, one of the most important stem cell cytokines.


Assuntos
Escherichia coli/metabolismo , Fator de Células-Tronco/metabolismo , Tiorredoxinas/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Vetores Genéticos/genética , Humanos , Camundongos , Reação em Cadeia da Polimerase , Fator de Células-Tronco/genética , Tiorredoxinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...