Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Genome Biol ; 24(1): 254, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37932818

RESUMEN

We introduce DEQSeq, a nanopore sequencing approach that rationalizes the selection of favorable genome editing enzymes from directed molecular evolution experiments. With the ability to capture full-length sequences, editing efficiencies, and specificities from thousands of evolved enzymes simultaneously, DEQSeq streamlines the process of identifying the most valuable variants for further study and application. We apply DEQSeq to evolved libraries of Cas12f-ABEs and designer-recombinases, identifying variants with improved properties for future applications. Our results demonstrate that DEQSeq is a powerful tool for accelerating enzyme discovery and advancing genome editing research.


Asunto(s)
Evolución Molecular Dirigida , Recombinasas , Recombinasas/genética , Recombinasas/metabolismo , Evolución Molecular Dirigida/métodos , Edición Génica/métodos , ADN , Sistemas CRISPR-Cas
2.
Cell Chem Biol ; 30(10): 1277-1294.e12, 2023 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-37473760

RESUMEN

Nitro-fatty acids (NFAs) are endogenous lipid mediators causing a spectrum of anti-inflammatory effects by covalent modification of key proteins within inflammatory signaling pathways. Recent animal models of solid tumors have helped demonstrate their potential as anti-tumorigenic therapeutics. This study evaluated the anti-tumorigenic effects of NFAs in colon carcinoma cells and other solid and leukemic tumor cell lines. NFAs inhibited the ubiquitin-proteasome system (UPS) by directly targeting the 26S proteasome, leading to polyubiquitination and inhibition of the proteasome activities. UPS suppression induced the unfolded protein response, resulting in tumor cell death. The NFA-mediated effects were substantial, specific, and enduring, representing a unique mode of action for UPS suppression. This study provides mechanistic insights into the biological actions of NFAs as possible endogenous tumor-suppressive factors, indicating that NFAs might be key structures for designing a novel class of direct proteasome inhibitors.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Ubiquitina , Animales , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Ácidos Grasos/farmacología , Inhibidores de Proteasoma/farmacología
3.
Nucleic Acids Res ; 2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37158248

RESUMEN

Tyrosine-type site-specific recombinases (Y-SSRs) are versatile tools for genome engineering due to their ability to mediate excision, integration, inversion and exchange of genomic DNA with single nucleotide precision. The ever-increasing need for sophisticated genome engineering is driving efforts to identify novel SSR systems with intrinsic properties more suitable for particular applications. In this work, we develop a systematic computational workflow for annotation of putative Y-SSR systems and apply this pipeline to identify and characterize eight new naturally occurring Cre-type SSR systems. We test their activity in bacterial and mammalian cells and establish selectivity profiles for the new and already established Cre-type SSRs with regard to their ability to mutually recombine their target sites. These data form the basis for sophisticated genome engineering experiments using combinations of Y-SSRs in research fields including advanced genomics and synthetic biology. Finally, we identify putative pseudo-sites and potential off-targets for Y-SSRs in the human and mouse genome. Together with established methods for altering the DNA-binding specificity of this class of enzymes, this work should facilitate the use of Y-SSRs for future genome surgery applications.

4.
Cancer Gene Ther ; 30(1): 108-123, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36114329

RESUMEN

5-Lipoxygenase (5-LO), the central enzyme in the biosynthesis of leukotrienes, is frequently expressed in human solid malignancies even though the enzyme is not present in the corresponding healthy tissues. There is little knowledge on the consequences of this expression for the tumor cells regarding gene expression and cellular function. We established a knockout (KO) of 5-LO in different cancer cell lines (HCT-116, HT-29, U-2 OS) and studied the consequences on global gene expression using next generation sequencing. Furthermore, cell viability, proliferation, migration and multicellular tumor spheroid (MCTS) formation were studied in these cells. Our results show that 5-LO influences the gene expression and cancer cell function in a cell type-dependent manner. The enzyme affected genes involved in cell adhesion, extracellular matrix formation, G protein signaling and cytoskeleton organization. Furthermore, absence of 5-LO elevated TGFß2 expression in HCT-116 cells while MCP-1, fractalkine and platelet-derived growth factor expression was attenuated in U-2 OS cells suggesting that tumor cell-derived 5-LO shapes the tumor microenvironment. In line with the gene expression data, KO of 5-LO had an impact on cell proliferation, motility and MCTS formation. Interestingly, pharmacological inhibition of 5-LO only partly mimicked the KO suggesting that also noncanonical functions are involved.


Asunto(s)
Araquidonato 5-Lipooxigenasa , Neoplasias , Humanos , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/metabolismo , Línea Celular , Transducción de Señal , Neoplasias/genética , Expresión Génica , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Microambiente Tumoral
5.
Biochem Pharmacol ; 203: 115187, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35878796

RESUMEN

Human 5-lipoxygenase (5-LO) is the key enzyme of leukotriene biosynthesis, mostly expressed in leukocytes and thus a crucial component of the innate immune system. In this study, we show that 5-LO, besides its canonical function as an arachidonic acid metabolizing enzyme, is a regulator of gene expression associated with euchromatin. By Crispr-Cas9-mediated 5-LO knockout (KO) in MonoMac6 (MM6) cells and subsequent RNA-Seq analysis, we identified 5-LO regulated genes which could be clustered to immune/defense response, cell adhesion, transcription and growth/developmental processes. Analysis of differentially expressed genes identified cyclooxygenase-2 (COX2, PTGS2) and kynureninase (KYNU) as strongly regulated 5-LO target genes. 5-LO knockout affected MM6 cell adhesion and tryptophan metabolism via inhibition of the degradation of the immunoregulator kynurenine. By subsequent FAIRE-Seq and 5-LO ChIP-Seq analyses, we found an association of 5-LO with euchromatin, with prominent 5-LO binding to promoter regions in actively transcribed genes. By enrichment analysis of the ChIP-Seq results, we identified potential 5-LO interaction partners. Furthermore, 5-LO ChIP-Seq peaks resemble patterns of H3K27ac histone marks, suggesting that 5-LO recruitment mainly takes place at acetylated histones. In summary, we demonstrate a noncanonical function of 5-LO as transcriptional regulator in monocytic cells.


Asunto(s)
Araquidonato 5-Lipooxigenasa , Eucromatina , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/metabolismo , Eucromatina/genética , Histonas/metabolismo , Humanos , Metabolismo de los Lípidos , Lipooxigenasa/genética , Lipooxigenasa/metabolismo
6.
Cancer Res ; 82(17): 3002-3015, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35802645

RESUMEN

KRAS is the most frequently mutated oncogene in human cancer, and its activating mutations represent long-sought therapeutic targets. Programmable nucleases, particularly the CRISPR-Cas9 system, provide an attractive tool for genetically targeting KRAS mutations in cancer cells. Here, we show that cleavage of a panel of KRAS driver mutations suppresses growth in various human cancer cell lines, revealing their dependence on mutant KRAS. However, analysis of the remaining cell population after long-term Cas9 expression unmasked the occurence of oncogenic KRAS escape variants that were resistant to Cas9-cleavage. In contrast, the use of an adenine base editor to correct oncogenic KRAS mutations progressively depleted the targeted cells without the appearance of escape variants and allowed efficient and simultaneous correction of a cancer-associated TP53 mutation. Oncogenic KRAS and TP53 base editing was possible in patient-derived cancer organoids, suggesting that base editor approaches to correct oncogenic mutations could be developed for functional interrogation of vulnerabilities in a personalized manner for future precision oncology applications. SIGNIFICANCE: Repairing KRAS mutations with base editors can be used for providing a better understanding of RAS biology and may lay the foundation for improved treatments for KRAS-mutant cancers.


Asunto(s)
Neoplasias , Proteínas Proto-Oncogénicas p21(ras) , Sistemas CRISPR-Cas , Carcinogénesis/genética , Edición Génica , Humanos , Mutación , Neoplasias/genética , Oncogenes , Medicina de Precisión , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética
7.
Methods Mol Biol ; 2508: 235-260, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35737245

RESUMEN

The CRISPR-Cas9 technology has revolutionized the scope and pace of biomedical research, enabling the targeting of specific genomic sequences for a wide spectrum of applications. Here we describe assays to functionally interrogate mutations identified in cancer cells utilizing both CRISPR-Cas9 nuclease and base editors. We provide guidelines to interrogate known cancer driver mutations or functionally screen for novel vulnerability mutations with these systems in characterized human cancer cell lines. The proposed platform should be transferable to primary cancer cells, opening up a path for precision oncology on a functional level.


Asunto(s)
Sistemas CRISPR-Cas , Neoplasias , Sistemas CRISPR-Cas/genética , Línea Celular , Edición Génica , Humanos , Mutación , Neoplasias/genética , Medicina de Precisión
8.
Life Sci Alliance ; 5(6)2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35288457

RESUMEN

The programmable CRISPR/Cas9 DNA nuclease is a versatile genome editing tool, but it requires the host cell DNA repair machinery to alter genomic sequences. This fact leads to unpredictable changes of the genome at the cut sites. Genome editing tools that can alter the genome without causing DNA double-strand breaks are therefore in high demand. Here, we show that expression of promoter-associated short guide (sg)RNAs together with dead Cas9 (dCas9) fused to a Krüppel-associated box domains (KRABd) in combination with the transcription repression domain of methyl CpG-binding protein 2 (MeCP2) can lead to persistent gene silencing in mouse embryonic stem cells and in human embryonic kidney (HEK) 293 cells. Surprisingly, this effect is achievable and even enhanced in DNA (cytosine-5)-methyltransferase 3A and 3B (Dnmt3A-/-, Dnmt3b-/-) depleted cells. Our results suggest that dCas9-KRABd-MeCP2 fusions are useful for long-term epigenetic gene silencing with utility in cell biology and potentially in therapeutical settings.


Asunto(s)
Sistemas CRISPR-Cas , Metilación de ADN , Animales , Sistemas CRISPR-Cas/genética , Metilación de ADN/genética , Epigénesis Genética/genética , Edición Génica/métodos , Células HEK293 , Humanos , Ratones , ARN Guía de Kinetoplastida/genética
9.
FASEB J ; 35(2): e21193, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33205517

RESUMEN

The miRNA biogenesis is tightly regulated to avoid dysfunction and consequent disease development. Here, we describe modulation of miRNA processing as a novel noncanonical function of the 5-lipoxygenase (5-LO) enzyme in monocytic cells. In differentiated Mono Mac 6 (MM6) cells, we found an in situ interaction of 5-LO with Dicer, a key enzyme in miRNA biogenesis. RNA sequencing of small noncoding RNAs revealed a functional impact, knockout of 5-LO altered the expression profile of several miRNAs. Effects of 5-LO could be observed at two levels. qPCR analyses thus indicated that (a) 5-LO promotes the transcription of the evolutionarily conserved miR-99b/let-7e/miR-125a cluster and (b) the 5-LO-Dicer interaction downregulates the processing of pre-let-7e, resulting in an increase in miR-125a and miR-99b levels by 5-LO without concomitant changes in let-7e levels in differentiated MM6 cells. Our observations suggest that 5-LO regulates the miRNA profile by modulating the Dicer-mediated processing of distinct pre-miRNAs. 5-LO inhibits the formation of let-7e which is a well-known inducer of cell differentiation, but promotes the generation of miR-99b and miR-125a known to induce cell proliferation and the maintenance of leukemic stem cell functions.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , MicroARNs/metabolismo , Procesamiento Postranscripcional del ARN , Araquidonato 5-Lipooxigenasa/genética , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , MicroARNs/genética , Ribonucleasa III/metabolismo , Transcriptoma
10.
Genes (Basel) ; 11(5)2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32384610

RESUMEN

In contrast to CRISPR/Cas9 nucleases, CRISPR base editors (BE) and prime editors (PE) enable predefined nucleotide exchanges in genomic sequences without generating DNA double strand breaks. Here, we employed BE and PE mRNAs in conjunction with chemically synthesized sgRNAs and pegRNAs for efficient editing of human induced pluripotent stem cells (iPSC). Whereas we were unable to correct a disease-causing mutation in patient derived iPSCs using a CRISPR/Cas9 nuclease approach, we corrected the mutation back to wild type with high efficiency utilizing an adenine BE. We also used adenine and cytosine BEs to introduce nine different cancer associated TP53 mutations into human iPSCs with up to 90% efficiency, generating a panel of cell lines to investigate the biology of these mutations in an isogenic background. Finally, we pioneered the use of prime editing in human iPSCs, opening this important cell type for the precise modification of nucleotides not addressable by BEs and to multiple nucleotide exchanges. These approaches eliminate the necessity of deriving disease specific iPSCs from human donors and allows the comparison of different disease-causing mutations in isogenic genetic backgrounds.


Asunto(s)
Adenina/química , Sistemas CRISPR-Cas , Citosina/química , Edición Génica/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Mutagénesis , Mutación , ARN Mensajero/genética , Aminohidrolasas , Enfermedades Autoinmunes del Sistema Nervioso/genética , Secuencia de Bases , Proteína 9 Asociada a CRISPR , Línea Celular , Técnicas de Reprogramación Celular , Cuerpos Embrioides , Genes p53 , Células HEK293 , Humanos , Imidazoles/farmacología , Malformaciones del Sistema Nervioso/genética , Piperazinas/farmacología , ARN Guía de Kinetoplastida/genética , ARN Mensajero/metabolismo , Proteína 1 que Contiene Dominios SAM y HD/genética , Transfección
11.
Blood ; 133(2): 168-179, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30498063

RESUMEN

Autophagy maintains hematopoietic stem cell integrity and prevents malignant transformation. In addition to bulk degradation, selective autophagy serves as an intracellular quality control mechanism and requires autophagy receptors, such as p62 (SQSTM1), to specifically bridge the ubiquitinated cargos into autophagosomes. Here, we investigated the function of p62 in acute myeloid leukemia (AML) in vitro and in murine in vivo models of AML. Loss of p62 impaired expansion and colony-forming ability of leukemia cells and prolonged latency of leukemia development in mice. High p62 expression was associated with poor prognosis in human AML. Using quantitative mass spectrometry, we identified enrichment of mitochondrial proteins upon immunoprecipitation of p62. Loss of p62 significantly delayed removal of dysfunctional mitochondria, increased mitochondrial superoxide levels, and impaired mitochondrial respiration. Moreover, we demonstrated that the autophagy-dependent function of p62 is essential for cell growth and effective mitochondrial degradation by mitophagy. Our results highlight the prominent role of selective autophagy in leukemia progression, and specifically, the importance of mitophagy to maintain mitochondrial integrity.


Asunto(s)
Autofagia , Leucemia Experimental/patología , Leucemia Mieloide Aguda/patología , Mitofagia , Proteína Sequestosoma-1/metabolismo , Proteína Sequestosoma-1/fisiología , Animales , Estudios de Seguimiento , Humanos , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Noqueados , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
12.
FASEB J ; 33(2): 1711-1726, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30188757

RESUMEN

Sphingosine-1-phosphate (S1P) is involved in the regulation of important cellular processes, including immune-cell trafficking and proliferation. Altered S1P signaling is strongly associated with inflammation, cancer progression, and atherosclerosis; however, the mechanisms underlying its pathophysiologic effects are only partially understood. This study evaluated the effects of S1P in vitro and in vivo on the biosynthesis of leukotrienes (LTs), which form a class of lipid mediators involved in the pathogenesis of inflammatory diseases. Here, we report for the first time that S1P potently suppresses LT biosynthesis in Ca2+-ionophore-stimulated intact human neutrophils. S1P treatment resulted in intracellular Ca2+ mobilization, perinuclear translocation, and finally irreversible suicide inactivation of the LT biosynthesis key enzyme 5-lipoxygenase (5-LO). Agonist studies and S1P receptor mRNA expression analysis provided evidence for a S1P receptor 4-mediated effect, which was confirmed by a functional knockout of S1P4 in HL60 cells. Systemic administration of S1P in wild-type mice decreased both macrophage and neutrophil migration in the lungs in response to LPS and significantly attenuated 5-LO product formation, whereas these effects were abrogated in 5-LO or S1P4 knockout mice. In summary, targeting the 5-LO pathway is an important mechanism to explain S1P-mediated pathophysiologic effects. Furthermore, agonism at S1P4 represents a novel effective strategy in pharmacotherapy of inflammation.-Fettel, J., Kühn, B., Guillen, N. A., Sürün, D., Peters, M., Bauer, R., Angioni, C., Geisslinger, G., Schnütgen, F., Meyer zu Heringdorf, D., Werz, O., Meybohm, P., Zacharowski, K., Steinhilber, D., Roos, J., Maier, T. J. Sphingosine-1-phosphate (S1P) induces potent anti-inflammatory effects in vitro and in vivo by S1P receptor 4-mediated suppression of 5-lipoxygenase activity.


Asunto(s)
Antiinflamatorios/farmacología , Araquidonato 5-Lipooxigenasa/efectos de los fármacos , Lisofosfolípidos/farmacología , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/análogos & derivados , Animales , Araquidonato 5-Lipooxigenasa/biosíntesis , Araquidonato 5-Lipooxigenasa/metabolismo , Ácido Araquidónico/metabolismo , Calcio/metabolismo , Línea Celular , Femenino , Humanos , Lisofosfolípidos/metabolismo , Ratones , Ratones Endogámicos C57BL , Neutrófilos/enzimología , Neutrófilos/metabolismo , Neumonía/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/metabolismo , Transducción de Señal , Esfingosina/metabolismo , Esfingosina/farmacología , Especificidad por Sustrato
13.
Drug Discov Today Technol ; 28: 33-39, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30205879

RESUMEN

The development of genome editing tools capable of modifying specific genomic sequences with unprecedented accuracy has opened up a wide range of new possibilities in targeted gene manipulation. In particular, the CRISPR/Cas9 system, a repurposed prokaryotic adaptive immune system, has been widely adopted because of its unmatched simplicity and flexibility. In this review we discuss achievements and current limitations of CRISPR/Cas9 genome editing in hematopoietic cells with special emphasis on its potential use in ex vivo gene therapy of monogenic blood disorders, HIV and cancer.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Terapia Genética/métodos , Síndrome de Inmunodeficiencia Adquirida/genética , Síndrome de Inmunodeficiencia Adquirida/terapia , Animales , Marcación de Gen/métodos , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/terapia , Humanos , Neoplasias/genética , Neoplasias/terapia
14.
Cell Death Dis ; 9(7): 756, 2018 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-29988033

RESUMEN

Metastasis formation requires active energy production and is regulated at multiple levels by mitochondrial metabolism. The hyperactive metabolism of cancer cells supports their extreme adaptability and plasticity and facilitates resistance to common anticancer therapies. In spite the potential relevance of a metastasis metabolic control therapy, so far, limited experience is available in this direction. Here, we evaluated the effect of the recently described α-ketoglutarate dehydrogenase (KGDH) inhibitor, (S)-2-[(2,6-dichlorobenzoyl) amino] succinic acid (AA6), in an orthotopic mouse model of breast cancer 4T1 and in other human breast cancer cell lines. In all conditions, AA6 altered Krebs cycle causing intracellular α-ketoglutarate (α-KG) accumulation. Consequently, the activity of the α-KG-dependent epigenetic enzymes, including the DNA demethylation ten-eleven translocation translocation hydroxylases (TETs), was increased. In mice, AA6 injection reduced metastasis formation and increased 5hmC levels in primary tumours. Moreover, in vitro and in vivo treatment with AA6 determined an α-KG accumulation paralleled by an enhanced production of nitric oxide (NO). This epigenetically remodelled metabolic environment efficiently counteracted the initiating steps of tumour invasion inhibiting the epithelial-to-mesenchymal transition (EMT). Mechanistically, AA6 treatment could be linked to upregulation of the NO-sensitive anti-metastatic miRNA 200 family and down-modulation of EMT-associated transcription factor Zeb1 and its CtBP1 cofactor. This scenario led to a decrease of the matrix metalloproteinase 3 (MMP3) and to an impairment of 4T1 aggressiveness. Overall, our data suggest that AA6 determines an α-KG-dependent epigenetic regulation of the TET-miR200-Zeb1/CtBP1-MMP3 axis providing an anti-metastatic effect in a mouse model of breast cancer-associated metastasis.


Asunto(s)
Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Ácido Succínico/uso terapéutico , Animales , Neoplasias de la Mama/tratamiento farmacológico , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Inhibidores Enzimáticos/química , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Ácido Succínico/química
15.
Nat Commun ; 9(1): 1281, 2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29599503

RESUMEN

Nitric oxide (NO) synthesis is a late event during differentiation of mouse embryonic stem cells (mESC) and occurs after release from serum and leukemia inhibitory factor (LIF). Here we show that after release from pluripotency, a subpopulation of mESC, kept in the naive state by 2i/LIF, expresses endothelial nitric oxide synthase (eNOS) and endogenously synthesizes NO. This eNOS/NO-positive subpopulation (ESNO+) expresses mesendodermal markers and is more efficient in the generation of cardiovascular precursors than eNOS/NO-negative cells. Mechanistically, production of endogenous NO triggers rapid Hdac2 S-nitrosylation, which reduces association of Hdac2 with the transcriptional repression factor Zeb1, allowing mesendodermal gene expression. In conclusion, our results suggest that the interaction between Zeb1, Hdac2, and eNOS is required for early mesendodermal differentiation of naive mESC.


Asunto(s)
Histona Desacetilasa 2/metabolismo , Células Madre Embrionarias de Ratones/citología , Miocardio/citología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/biosíntesis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular Tumoral , Células HeLa , Humanos , Factor Inhibidor de Leucemia/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Miocardio/metabolismo
16.
Mol Ther Nucleic Acids ; 10: 1-8, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29499925

RESUMEN

The CRISPR/Cas9 prokaryotic adaptive immune system and its swift repurposing for genome editing enables modification of any prespecified genomic sequence with unprecedented accuracy and efficiency, including targeted gene repair. We used the CRISPR/Cas9 system for targeted repair of patient-specific point mutations in the Cytochrome b-245 heavy chain gene (CYBB), whose inactivation causes chronic granulomatous disease (XCGD)-a life-threatening immunodeficiency disorder characterized by the inability of neutrophils and macrophages to produce microbicidal reactive oxygen species (ROS). We show that frameshift mutations can be effectively repaired in hematopoietic cells by non-integrating lentiviral vectors carrying RNA-guided Cas9 endonucleases (RGNs). Because about 25% of most inherited blood disorders are caused by frameshift mutations, our results suggest that up to a quarter of all patients suffering from monogenic blood disorders could benefit from gene therapy employing personalized, donor template-free RGNs.

17.
Circ Res ; 122(1): 31-46, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29158345

RESUMEN

RATIONALE: Human cardiac mesenchymal cells (CMSCs) are a therapeutically relevant primary cell population. Diabetes mellitus compromises CMSC function as consequence of metabolic alterations and incorporation of stable epigenetic changes. OBJECTIVE: To investigate the role of α-ketoglutarate (αKG) in the epimetabolic control of DNA demethylation in CMSCs. METHODS AND RESULTS: Quantitative global analysis, methylated and hydroxymethylated DNA sequencing, and gene-specific GC methylation detection revealed an accumulation of 5-methylcytosine, 5-hydroxymethylcytosine, and 5-formylcytosine in the genomic DNA of human CMSCs isolated from diabetic donors. Whole heart genomic DNA analysis revealed iterative oxidative cytosine modification accumulation in mice exposed to high-fat diet (HFD), injected with streptozotocin, or both in combination (streptozotocin/HFD). In this context, untargeted and targeted metabolomics indicated an intracellular reduction of αKG synthesis in diabetic CMSCs and in the whole heart of HFD mice. This observation was paralleled by a compromised TDG (thymine DNA glycosylase) and TET1 (ten-eleven translocation protein 1) association and function with TET1 relocating out of the nucleus. Molecular dynamics and mutational analyses showed that αKG binds TDG on Arg275 providing an enzymatic allosteric activation. As a consequence, the enzyme significantly increased its capacity to remove G/T nucleotide mismatches or 5-formylcytosine. Accordingly, an exogenous source of αKG restored the DNA demethylation cycle by promoting TDG function, TET1 nuclear localization, and TET/TDG association. TDG inactivation by CRISPR/Cas9 knockout or TET/TDG siRNA knockdown induced 5-formylcytosine accumulation, thus partially mimicking the diabetic epigenetic landscape in cells of nondiabetic origin. The novel compound (S)-2-[(2,6-dichlorobenzoyl)amino]succinic acid (AA6), identified as an inhibitor of αKG dehydrogenase, increased the αKG level in diabetic CMSCs and in the heart of HFD and streptozotocin mice eliciting, in HFD, DNA demethylation, glucose uptake, and insulin response. CONCLUSIONS: Restoring the epimetabolic control of DNA demethylation cycle promises beneficial effects on cells compromised by environmental metabolic changes.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Ácidos Cetoglutáricos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Timina ADN Glicosilasa/metabolismo , Animales , Células Cultivadas , Citosina/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Inhibidores Enzimáticos/farmacología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ácidos Cetoglutáricos/antagonistas & inhibidores , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos
18.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(5): 561-571, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28257804

RESUMEN

Human 5-lipoxygenase (5-LO-WT) initiates the leukotriene (LT) biosynthesis. LTs play an important role in diseases like asthma, atherosclerosis and in many types of cancer. In this study, we investigated the 5-LO isoforms 5-LO∆13, 5-LO∆4 and 5-LOp12, lacking the exons 13, 4 or a part of exon 12, respectively. We were able to detect the mRNA of the isoforms 5-LO∆13 and 5-LOp12 in B and T cell lines as well as in primary B and T cells and monocytes. Furthermore, we found that expression of 5-LO and particularly of the 5-LO∆13 and 5-LOp12 isoforms is increased in monocytes from patients with rheumatoid arthritis and sepsis. Confocal microscopy of HEK293T cells stably transfected with tagged 5-LO-WT and/or the isoforms revealed that 5-LO-WT is localized in the nucleus whereas all isoforms are located in the cytosol. Additionally, all isoforms are catalytically inactive and do not seem to influence the specific activity of 5-LO-WT. S271A mutation in 5-LO-WT and treatment of the cells with sorbitol or KN-93/SB203580 changes the localization of the WT enzyme to the cytosol. Despite colocalization with the S271A mutant, the isoforms did not affect LT biosynthesis. Analysis of the phosphorylation pattern of 5-LO-WT and all the isoforms revealed that 5-LOp12 and 5-LO∆13 are highly phosphorylated at Ser271 and 5-LOp12 at Ser523. Furthermore, coexpression of the isoforms inhibited or stimulated 5-LO-WT expression in transiently and stably transfected HEK293T cells suggesting that the isoforms have other functions than canonical LT biosynthesis.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Núcleo Celular/ultraestructura , Citosol/ultraestructura , Isoformas de Proteínas/metabolismo , Araquidonato 5-Lipooxigenasa/química , Araquidonato 5-Lipooxigenasa/aislamiento & purificación , Núcleo Celular/metabolismo , Citosol/metabolismo , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Humanos , Leucotrienos/biosíntesis , Neutrófilos/metabolismo , Neutrófilos/ultraestructura , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/aislamiento & purificación
19.
Matrix Biol ; 59: 109-120, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27645114

RESUMEN

Latent transforming growth factor beta binding protein 4 (LTBP4) belongs to the fibrillin/LTBP family of proteins and plays an important role as a structural component of extracellular matrix (ECM) and local regulator of TGFß signaling. We have previously reported that Ltbp4S knock out mice (Ltbp4S-/-) develop centrilobular emphysema reminiscent of late stage COPD, which could be partially rescued by inactivating the antioxidant protein Sestrin 2 (Sesn2). More recent studies showed that Sesn2 knock out mice upregulate Pdgfrß-controlled alveolar maintenance programs that protect against cigarette smoke induced pulmonary emphysema. Based on this, we hypothesized that the emphysema of Ltbp4S-/- mice is primarily caused by defective Pdgfrß signaling. Here we show that LTBP4 induces Pdgfrß signaling by inhibiting the antioxidant Nrf2/Keap1 pathway in a TGFß-dependent manner. Overall, our data identified Ltbp4 as a major player in lung remodeling and injury repair.


Asunto(s)
Matriz Extracelular/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Factor 2 Relacionado con NF-E2/genética , Enfisema Pulmonar/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Transformador beta/genética , Animales , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Matriz Extracelular/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Proteínas de Unión a TGF-beta Latente/deficiencia , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Visón , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Peroxidasas , Plásmidos/química , Plásmidos/metabolismo , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Tropoelastina/deficiencia , Tropoelastina/genética
20.
Artículo en Inglés | MEDLINE | ID: mdl-26210919

RESUMEN

The key enzyme in leukotriene (LT) biosynthesis is 5-lipoxygenase (5-LO), which is expressed in myeloid cells and in B lymphocytes. There are three phosphorylation sites on 5-LO (Ser271, Ser523 and Ser663). Protein kinase A (PKA) phosphorylates 5-LO on Ser523. In this report, we demonstrate by immunoblotting that native 5-LO in mantle B cell lymphoma (MCL) cells (Granta519, JEKO1, and Rec1) and in primary chronic B lymphocytic leukemia cells (B-CLL) is phosphorylated on Ser523. In contrast, we could not detect phosphorylation of 5-LO on Ser523 in human granulocytes or monocytes. Phosphorylated 5-LO was purified from Rec1 cells, using an ATP-agarose column, and the partially purified enzyme could be dephosphorylated with alkaline phosphatase. Incubation of Rec1 cells with 8-Br-cAMP or prostaglandin E2 stimulated phosphorylation at Ser523. Furthermore, FLAG-5LO was expressed in Rec1 cells, and the cells were cultivated in the presence of 8-Br-cAMP. The 5-LO protein from these cells was immunoprecipitated, first with anti-FLAG, followed by anti-pSer523-5-LO. The presence of 5-LO protein in the final precipitate further supported the finding that the protein recognized by the pSer523 antibody was 5-LO. Taken together, this study shows that 5-LO in B cells is phosphorylated on Ser523 and demonstrates for the first time a chemical difference between 5-LO in myeloid cells and B cells.


Asunto(s)
Araquidonato 5-Lipooxigenasa/metabolismo , Linfocitos B/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Linfoma de Células del Manto/metabolismo , Fosforilación , Serina/metabolismo , Línea Celular Tumoral , AMP Cíclico/metabolismo , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Linfoma de Células del Manto/patología , Células Mieloides/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...