Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(16): 9596-9612, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39021334

RESUMEN

DNA damage severely impedes gene transcription by RNA polymerase II (Pol II), causing cellular dysfunction. Transcription-Coupled Nucleotide Excision Repair (TC-NER) specifically removes such transcription-blocking damage. TC-NER initiation relies on the CSB, CSA and UVSSA proteins; loss of any results in complete TC-NER deficiency. Strikingly, UVSSA deficiency results in UV-Sensitive Syndrome (UVSS), with mild cutaneous symptoms, while loss of CSA or CSB activity results in the severe Cockayne Syndrome (CS), characterized by neurodegeneration and premature aging. Thus far the underlying mechanism for these contrasting phenotypes remains unclear. Live-cell imaging approaches reveal that in TC-NER proficient cells, lesion-stalled Pol II is swiftly resolved, while in CSA and CSB knockout (KO) cells, elongating Pol II remains damage-bound, likely obstructing other DNA transacting processes and shielding the damage from alternative repair pathways. In contrast, in UVSSA KO cells, Pol II is cleared from the damage via VCP-mediated proteasomal degradation which is fully dependent on the CRL4CSA ubiquitin ligase activity. This Pol II degradation might provide access for alternative repair mechanisms, such as GG-NER, to remove the damage. Collectively, our data indicate that the inability to clear lesion-stalled Pol II from the chromatin, rather than TC-NER deficiency, causes the severe phenotypes observed in CS.


Asunto(s)
Síndrome de Cockayne , Daño del ADN , ADN Helicasas , Enzimas Reparadoras del ADN , Reparación del ADN , Proteínas de Unión a Poli-ADP-Ribosa , ARN Polimerasa II , Transcripción Genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Humanos , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ADN Helicasas/metabolismo , ADN Helicasas/genética , Síndrome de Cockayne/genética , Síndrome de Cockayne/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteína que Contiene Valosina/metabolismo , Proteína que Contiene Valosina/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/genética , Rayos Ultravioleta , Línea Celular , Reparación por Escisión , Proteínas Portadoras
2.
Nat Cell Biol ; 26(5): 770-783, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600236

RESUMEN

DNA-protein crosslinks (DPCs) arise from enzymatic intermediates, metabolism or chemicals like chemotherapeutics. DPCs are highly cytotoxic as they impede DNA-based processes such as replication, which is counteracted through proteolysis-mediated DPC removal by spartan (SPRTN) or the proteasome. However, whether DPCs affect transcription and how transcription-blocking DPCs are repaired remains largely unknown. Here we show that DPCs severely impede RNA polymerase II-mediated transcription and are preferentially repaired in active genes by transcription-coupled DPC (TC-DPC) repair. TC-DPC repair is initiated by recruiting the transcription-coupled nucleotide excision repair (TC-NER) factors CSB and CSA to DPC-stalled RNA polymerase II. CSA and CSB are indispensable for TC-DPC repair; however, the downstream TC-NER factors UVSSA and XPA are not, a result indicative of a non-canonical TC-NER mechanism. TC-DPC repair functions independently of SPRTN but is mediated by the ubiquitin ligase CRL4CSA and the proteasome. Thus, DPCs in genes are preferentially repaired in a transcription-coupled manner to facilitate unperturbed transcription.


Asunto(s)
ADN Helicasas , Enzimas Reparadoras del ADN , Reparación del ADN , Proteínas de Unión a Poli-ADP-Ribosa , Proteolisis , ARN Polimerasa II , Transcripción Genética , Humanos , Proteínas Portadoras , ADN/metabolismo , ADN/genética , Daño del ADN , ADN Helicasas/metabolismo , ADN Helicasas/genética , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Células HEK293 , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores de Interleucina-17 , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética
3.
Cell Rep Methods ; 4(1): 100674, 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38176411

RESUMEN

Transcription by RNA polymerase II (RNA Pol II) is crucial for cellular function, but DNA damage severely impedes this process. Thus far, transcription-blocking DNA lesions (TBLs) and their repair have been difficult to quantify in living cells. To overcome this, we generated, using CRISPR-Cas9-mediated gene editing, mScarletI-tagged Cockayne syndrome group B protein (CSB) and UV-stimulated scaffold protein A (UVSSA) knockin cells. These cells allowed us to study the binding dynamics of CSB and UVSSA to lesion-stalled RNA Pol II using fluorescence recovery after photobleaching (FRAP). We show that especially CSB mobility is a sensitive transcription stress marker at physiologically relevant DNA damage levels. Transcription-coupled nucleotide excision repair (TC-NER)-mediated repair can be assessed by studying CSB immobilization over time. Additionally, flow cytometry reveals the regulation of CSB protein levels by CRL4CSA-mediated ubiquitylation and deubiquitylation by USP7. This approach allows the sensitive detection of TBLs and their repair and the study of TC-NER complex assembly and stability in living cells.


Asunto(s)
Reparación del ADN , ARN Polimerasa II , ARN Polimerasa II/genética , Transcripción Genética , Daño del ADN , Proteínas/genética , ADN/genética
4.
Nucleic Acids Res ; 51(11): 5396-5413, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-36971114

RESUMEN

The deubiquitinating enzyme Ataxin-3 (ATXN3) contains a polyglutamine (PolyQ) region, the expansion of which causes spinocerebellar ataxia type-3 (SCA3). ATXN3 has multiple functions, such as regulating transcription or controlling genomic stability after DNA damage. Here we report the role of ATXN3 in chromatin organization during unperturbed conditions, in a catalytic-independent manner. The lack of ATXN3 leads to abnormalities in nuclear and nucleolar morphology, alters DNA replication timing and increases transcription. Additionally, indicators of more open chromatin, such as increased mobility of histone H1, changes in epigenetic marks and higher sensitivity to micrococcal nuclease digestion were detected in the absence of ATXN3. Interestingly, the effects observed in cells lacking ATXN3 are epistatic to the inhibition or lack of the histone deacetylase 3 (HDAC3), an interaction partner of ATXN3. The absence of ATXN3 decreases the recruitment of endogenous HDAC3 to the chromatin, as well as the HDAC3 nuclear/cytoplasm ratio after HDAC3 overexpression, suggesting that ATXN3 controls the subcellular localization of HDAC3. Importantly, the overexpression of a PolyQ-expanded version of ATXN3 behaves as a null mutant, altering DNA replication parameters, epigenetic marks and the subcellular distribution of HDAC3, giving new insights into the molecular basis of the disease.


Asunto(s)
Ataxina-3 , Cromatina , Replicación del ADN , Humanos , Ataxina-3/genética , Ataxina-3/metabolismo , Cromatina/genética , Daño del ADN , Enfermedad de Machado-Joseph/genética , Proteínas Represoras/metabolismo
5.
Nat Commun ; 13(1): 3624, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-35750669

RESUMEN

The precise regulation of RNA Polymerase II (Pol II) transcription after genotoxic stress is crucial for proper execution of the DNA damage-induced stress response. While stalling of Pol II on transcription-blocking lesions (TBLs) blocks transcript elongation and initiates DNA repair in cis, TBLs additionally elicit a response in trans that regulates transcription genome-wide. Here we uncover that, after an initial elongation block in cis, TBLs trigger the genome-wide VCP-mediated proteasomal degradation of promoter-bound, P-Ser5-modified Pol II in trans. This degradation is mechanistically distinct from processing of TBL-stalled Pol II, is signaled via GSK3, and contributes to the TBL-induced transcription block, even in transcription-coupled repair-deficient cells. Thus, our data reveal the targeted degradation of promoter-bound Pol II as a critical pathway that allows cells to cope with DNA damage-induced transcription stress and enables the genome-wide adaptation of transcription to genotoxic stress.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Transcripción Genética , Daño del ADN/genética , Reparación del ADN/genética , Glucógeno Sintasa Quinasa 3/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo
7.
Nat Cell Biol ; 23(6): 608-619, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34108662

RESUMEN

Correct transcription is crucial for life. However, DNA damage severely impedes elongating RNA polymerase II, causing transcription inhibition and transcription-replication conflicts. Cells are equipped with intricate mechanisms to counteract the severe consequence of these transcription-blocking lesions. However, the exact mechanism and factors involved remain largely unknown. Here, using a genome-wide CRISPR-Cas9 screen, we identified the elongation factor ELOF1 as an important factor in the transcription stress response following DNA damage. We show that ELOF1 has an evolutionarily conserved role in transcription-coupled nucleotide excision repair (TC-NER), where it promotes recruitment of the TC-NER factors UVSSA and TFIIH to efficiently repair transcription-blocking lesions and resume transcription. Additionally, ELOF1 modulates transcription to protect cells against transcription-mediated replication stress, thereby preserving genome stability. Thus, ELOF1 protects the transcription machinery from DNA damage via two distinct mechanisms.


Asunto(s)
Daño del ADN , Reparación del ADN , Inestabilidad Genómica , Factor 1 de Elongación Peptídica/metabolismo , Elongación de la Transcripción Genética , Sistemas CRISPR-Cas , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Evolución Molecular , Células HCT116 , Humanos , Factor 1 de Elongación Peptídica/genética , ARN Polimerasa II/metabolismo , Factor de Transcripción TFIIH/genética , Factor de Transcripción TFIIH/metabolismo , Ubiquitinación
8.
Nat Commun ; 10(1): 4887, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31653834

RESUMEN

Accumulation of DNA lesions causing transcription stress is associated with natural and accelerated aging and culminates with profound metabolic alterations. Our understanding of the mechanisms governing metabolic redesign upon genomic instability, however, is highly rudimentary. Using Ercc1-defective mice and Xpg knock-out mice, we demonstrate that combined defects in transcription-coupled DNA repair (TCR) and in nucleotide excision repair (NER) directly affect bioenergetics due to declined transcription, leading to increased ATP levels. This in turn inhibits glycolysis allosterically and favors glucose rerouting through the pentose phosphate shunt, eventually enhancing production of NADPH-reducing equivalents. In NER/TCR-defective mutants, augmented NADPH is not counterbalanced by increased production of pro-oxidants and thus pentose phosphate potentiation culminates in an over-reduced redox state. Skin fibroblasts from the TCR disease Cockayne syndrome confirm results in animal models. Overall, these findings unravel a mechanism connecting DNA damage and transcriptional stress to metabolic redesign and protective antioxidant defenses.


Asunto(s)
Adenosina Trifosfato/metabolismo , Antioxidantes/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Glucólisis/fisiología , NADP/metabolismo , Vía de Pentosa Fosfato/fisiología , Transcripción Genética/genética , Regulación Alostérica , Animales , Síndrome de Cockayne/metabolismo , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Fibroblastos/metabolismo , Inestabilidad Genómica , Metabolómica , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Oxidación-Reducción , Piel/citología , Factores de Transcripción/genética
9.
EMBO Rep ; 19(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30104204

RESUMEN

Histone acetylation influences protein interactions and chromatin accessibility and plays an important role in the regulation of transcription, replication, and DNA repair. Conversely, DNA damage affects these crucial cellular processes and induces changes in histone acetylation. However, a comprehensive overview of the effects of DNA damage on the histone acetylation landscape is currently lacking. To quantify changes in histone acetylation, we developed an unbiased quantitative mass spectrometry analysis on affinity-purified acetylated histone peptides, generated by differential parallel proteolysis. We identify a large number of histone acetylation sites and observe an overall reduction of acetylated histone residues in response to DNA damage, indicative of a histone-wide loss of acetyl modifications. This decrease is mainly caused by DNA damage-induced replication stress coupled to specific proteasome-dependent loss of acetylated histones. Strikingly, this degradation of acetylated histones is independent of ubiquitylation but requires the PA200-proteasome activator, a complex that specifically targets acetylated histones for degradation. The uncovered replication stress-induced degradation of acetylated histones represents an important chromatin-modifying response to cope with replication stress.


Asunto(s)
Cromatina/genética , Daño del ADN/genética , Proteínas Nucleares/genética , Complejo de la Endopetidasa Proteasomal/genética , Acetilación , Secuencia de Aminoácidos/genética , Reparación del ADN/genética , Replicación del ADN/genética , Histonas/genética , Humanos , Proteolisis , Ubiquitinación/genética
10.
Nucleic Acids Res ; 46(15): 7747-7756, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-29955842

RESUMEN

Transcription-coupled nucleotide excision repair factor Cockayne syndrome protein B (CSB) was suggested to function in the repair of oxidative DNA damage. However thus far, no clear role for CSB in base excision repair (BER), the dedicated pathway to remove abundant oxidative DNA damage, could be established. Using live cell imaging with a laser-assisted procedure to locally induce 8-oxo-7,8-dihydroguanine (8-oxoG) lesions, we previously showed that CSB is recruited to these lesions in a transcription-dependent but NER-independent fashion. Here we showed that recruitment of the preferred 8-oxoG-glycosylase 1 (OGG1) is independent of CSB or active transcription. In contrast, recruitment of the BER-scaffolding protein, X-ray repair cross-complementing protein 1 (XRCC1), to 8-oxoG lesions is stimulated by CSB and transcription. Remarkably, recruitment of XRCC1 to BER-unrelated single strand breaks (SSBs) does not require CSB or transcription. Together, our results suggest a specific transcription-dependent role for CSB in recruiting XRCC1 to BER-generated SSBs, whereas XRCC1 recruitment to SSBs generated independently of BER relies predominantly on PARP activation. Based on our results, we propose a model in which CSB plays a role in facilitating BER progression at transcribed genes, probably to allow XRCC1 recruitment to BER-intermediates masked by RNA polymerase II complexes stalled at these intermediates.


Asunto(s)
Daño del ADN , ADN Helicasas/genética , Enzimas Reparadoras del ADN/genética , Reparación del ADN , ADN/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Transcripción Genética , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/genética , Línea Celular , ADN/metabolismo , ADN Helicasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Células HEK293 , Humanos , Modelos Genéticos , Oxidación-Reducción , Estrés Oxidativo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo
11.
Proc Natl Acad Sci U S A ; 115(19): E4368-E4376, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29632207

RESUMEN

Initiation and promoter-proximal pausing are key regulatory steps of RNA Polymerase II (Pol II) transcription. To study the in vivo dynamics of endogenous Pol II during these steps, we generated fully functional GFP-RPB1 knockin cells. GFP-RPB1 photobleaching combined with computational modeling revealed four kinetically distinct Pol II fractions and showed that on average 7% of Pol II are freely diffusing, while 10% are chromatin-bound for 2.4 seconds during initiation, and 23% are promoter-paused for only 42 seconds. This unexpectedly high turnover of Pol II at promoters is most likely caused by premature termination of initiating and promoter-paused Pol II and is in sharp contrast to the 23 minutes that elongating Pol II resides on chromatin. Our live-cell-imaging approach provides insights into Pol II dynamics and suggests that the continuous release and reinitiation of promoter-bound Pol II is an important component of transcriptional regulation.


Asunto(s)
Regiones Promotoras Genéticas/fisiología , ARN Polimerasa II/metabolismo , Transcripción Genética/fisiología , Línea Celular Transformada , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , ARN Polimerasa II/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
12.
Nat Commun ; 6: 7499, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26151477

RESUMEN

XPC recognizes UV-induced DNA lesions and initiates their removal by nucleotide excision repair (NER). Damage recognition in NER is tightly controlled by ubiquitin and SUMO modifications. Recent studies have shown that the SUMO-targeted ubiquitin ligase RNF111 promotes K63-linked ubiquitylation of SUMOylated XPC after DNA damage. However, the exact regulatory function of these modifications in vivo remains elusive. Here we show that RNF111 is required for efficient repair of ultraviolet-induced DNA lesions. RNF111-mediated ubiquitylation promotes the release of XPC from damaged DNA after NER initiation, and is needed for stable incorporation of the NER endonucleases XPG and ERCC1/XPF. Our data suggest that RNF111, together with the CRL4(DDB2) ubiquitin ligase complex, is responsible for sequential XPC ubiquitylation, which regulates the recruitment and release of XPC and is crucial for efficient progression of the NER reaction, thereby providing an extra layer of quality control of NER.


Asunto(s)
Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas Nucleares/metabolismo , Proteína SUMO-1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Daño del ADN , Proteínas de Unión al ADN/genética , Endonucleasas/genética , Endonucleasas/metabolismo , Humanos , Proteínas Nucleares/genética , ARN Interferente Pequeño , Proteína SUMO-1/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética
13.
Breast Cancer Res Treat ; 148(1): 19-31, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25266129

RESUMEN

Breast cancer (BC) is a disease with intra- and inter-tumor heterogeneity, and models representing the complete variety of clinical BC phenotypes are not available. We explored the tumor growth potential and metastatic behavior of human BC cell lines and determined whether these cell lines can recapitulate subtype-related biological characteristics of tumors. Eighteen human BC cell lines were implanted under the mammary fat pad of nude mice. Subtype-specific differences in tumor growth, metastatic ability to distant sites, and tumor-related survival of mice were recorded. Eighty-nine percent of the cell lines gave rise to xenografts of which 56 % showed metastasis to distant sites. A clear difference was observed in growth of xenografts from cell lines of different molecular subtypes (P = 0.001; Kruskal-Wallis test). Mice bearing the basal-like and the normal-like xenografts showed poor tumor-related survival (HR: 10.50; P = 0.002 and HR: 9.89; P = 0.003, respectively) compared with those bearing the ERBB2-positive xenografts, which had the longest survival. Subtype-specific metastasis to distant sites between xenografts was not however observed. Comparable to clinical behavior in humans, we observed that the basal-like and the normal-like cell lines grew more aggressively in mice than the cell lines of other molecular subtypes. However, in contrast to clinical findings, we observed no relationships between intrinsic subtype and preferences for site of relapse. Importantly, we have established xenograft models from 16 phenotypically and molecularly diverse human BC cell lines, which can be exploited as useful tools to perform functional studies and screening of interfering drugs.


Asunto(s)
Neoplasias de la Mama/patología , Línea Celular Tumoral/patología , Modelos Animales de Enfermedad , Animales , Femenino , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Trasplante Heterólogo
14.
Proc Natl Acad Sci U S A ; 111(27): 9828-33, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958884

RESUMEN

Daily synchronous rhythms of cell division at the tissue or organism level are observed in many species and suggest that the circadian clock and cell cycle oscillators are coupled. For mammals, despite known mechanistic interactions, the effect of such coupling on clock and cell cycle progression, and hence its biological relevance, is not understood. In particular, we do not know how the temporal organization of cell division at the single-cell level produces this daily rhythm at the tissue level. Here we use multispectral imaging of single live cells, computational methods, and mathematical modeling to address this question in proliferating mouse fibroblasts. We show that in unsynchronized cells the cell cycle and circadian clock robustly phase lock each other in a 1:1 fashion so that in an expanding cell population the two oscillators oscillate in a synchronized way with a common frequency. Dexamethasone-induced synchronization reveals additional clock states. As well as the low-period phase-locked state there are distinct coexisting states with a significantly higher period clock. Cells transition to these states after dexamethasone synchronization. The temporal coordination of cell division by phase locking to the clock at a single-cell level has significant implications because disordered circadian function is increasingly being linked to the pathogenesis of many diseases, including cancer.


Asunto(s)
Proteínas CLOCK/metabolismo , Proteínas de Ciclo Celular/metabolismo , Animales , Ritmo Circadiano/efectos de los fármacos , Dexametasona/farmacología , Ratones , Células 3T3 NIH
15.
PLoS One ; 8(2): e56623, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23418588

RESUMEN

The transcription/translation feedback loop-based molecular oscillator underlying the generation of circadian gene expression is preserved in almost all organisms. Interestingly, the animal circadian clock proteins CRYPTOCHROME (CRY), PERIOD (PER) and TIMELESS (TIM) are strongly conserved at the amino acid level through evolution. Within this evolutionary frame, TIM represents a fascinating puzzle. While Drosophila contains two paralogs, dTIM and dTIM2, acting in clock/photoreception and chromosome integrity/photoreception respectively, mammals contain only one TIM homolog. Whereas TIM has been shown to regulate replication termination and cell cycle progression, its functional link to the circadian clock is under debate. Here we show that RNAi-mediated knockdown of TIM in NIH3T3 and U2OS cells shortens the period by 1 hour and diminishes DNA damage-dependent phase advancing. Furthermore, we reveal that the N-terminus of TIM is sufficient for interaction with CRY1 and CHK1 as well for homodimerization, and the C-terminus is necessary for nuclear localization. Interestingly, the long TIM isoform (l-TIM), but not the short (s-TIM), interacts with CRY1 and both proteins can reciprocally regulate their nuclear translocation in transiently transfected COS7 cells. Finally, we demonstrate that co-expression of PER2 abolishes the formation of the TIM/CRY1 complex through affinity binding competition to the C-terminal tail of CRY1. Notably, the presence of the latter protein region evolutionarily and structurally distinguishes mammalian from insect CRYs. We propose that the dynamic interaction between these three proteins could represent a post-translational aspect of the mammalian circadian clock that is important for its pace and adaption to external stimuli, such as DNA damage and/or light.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Relojes Circadianos/fisiología , Daño del ADN , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Western Blotting , Células COS , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Relojes Circadianos/genética , Criptocromos/genética , Criptocromos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Modelos Biológicos , Células 3T3 NIH , Señales de Localización Nuclear/genética , Unión Proteica , Interferencia de ARN , Factores de Tiempo
16.
Contrast Media Mol Imaging ; 8(2): 165-74, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23281289

RESUMEN

In cell therapy, noninvasive monitoring of in vivo cell fate is challenging. In this study we investigated possible differences in R1, R2 or R2* relaxation rate as a measure of overall cell viability for mesenchymal stem cells labeled with Gd-liposomes (Gd-MSCs) or iron oxide nanoparticles (SPIO-MSCs). Cells were also transduced with a luciferase vector, facilitating a correlation between MRI findings and cell viability using bioluminescence imaging (BLI). Viable Gd-MSCs were clearly distinguishable from nonviable Gd-MSCs under both in vitro and in vivo conditions, clearly differing quantitatively (ΔR1 and ΔR2) as well as by visual appearance (hypo- or hyperintense contrast). Immediately post-injection,viable Gd-MSCs caused a substantially larger ΔR2 and lower ΔR1 effect compared with nonviable MSCs. With time, the ΔR1 and ΔR2 relaxation rate showed a good negative correlation with increasing cell number following proliferation. Upon injection, no substantial quantitative or visual differences between viable and nonviable SPIO-MSCs were detected. Moreover, nonviable SPIO-MSCs caused a persisting signal void in vivo, compromising the specificity of this contrast agent. In vivo persistence of SPIO particles was confirmed by histological staining. A large difference was found between SPIO- and Gd-labeled cells in the accuracy of MR relaxometry in assessing the cell viability status. Gd-liposomes provide a more accurate and specific assessment of cell viability than SPIO particles. Viable Gd cells can be differentiated from nonviable Gd cells even by visual interpretation. These findings clearly indicate Gd to be the favourable contrast agent in qualitative and quantitative evaluation of labeled cell fate in future cell therapy experiments.


Asunto(s)
Gadolinio/efectos adversos , Hierro/efectos adversos , Mediciones Luminiscentes/métodos , Imagen por Resonancia Magnética/métodos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Medios de Contraste/efectos adversos , Ratas , Ratas Wistar , Coloración y Etiquetado
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA