Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83.250
Filtrar
1.
Gene ; 932: 148880, 2025 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-39181273

RESUMEN

It has been discovered that Trichorhinophalangeal Syndrome-1 (TRPS1), a novel member of the GATA transcription factor family, participates in both normal physiological processes and the development of numerous diseases. Recently, TRPS1 has been identified as a new biomarker to aid in cancer diagnosis and is very common in breast cancer (BC), especially in triple-negative breast cancer (TNBC). In this review, we discussed the structure and function of TRPS1 in various normal cells, focused on its role in tumorigenesis and tumor development, and summarize the research status of TRPS1 in the occurrence and development of BC. We also analyzed the potential use of TRPS1 in guiding clinically personalized precision treatment and the development of targeted drugs.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Mama , Proteínas de Unión al ADN , Proteínas Represoras , Factores de Transcripción , Humanos , Femenino , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Regulación Neoplásica de la Expresión Génica , Carcinogénesis/genética , Carcinogénesis/metabolismo , Animales
2.
Methods Mol Biol ; 2856: 11-22, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39283444

RESUMEN

The Structural Maintenance of Chromosomes (SMC) protein complexes are DNA-binding molecular machines required to shape chromosomes into functional units and to safeguard the genome through cell division. These ring-shaped multi-subunit protein complexes, which are present in all kingdoms of life, achieve this by organizing chromosomes in three-dimensional space. Mechanistically, the SMC complexes hydrolyze ATP to either stably entrap DNA molecules within their lumen, or rapidly reel DNA into large loops, which allow them to link two stretches of DNA in cis or trans. In this chapter, the canonical structure of the SMC complexes is first introduced, followed by a description of the composition and general functions of the main types of eukaryotic and prokaryotic SMC complexes. Thereafter, the current model for how SMC complexes perform in vitro DNA loop extrusion is presented. Lastly, chromosome loop formation by SMC complexes is introduced, and how the DNA loop extrusion mechanism contributes to chromosome looping by SMC complexes in cells is discussed.


Asunto(s)
Cromosomas , Cromosomas/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/química , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , ADN/química , ADN/metabolismo , ADN/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/química , Adenosina Trifosfato/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/química
3.
FASEB J ; 38(19): e70085, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39352691

RESUMEN

As renal progenitor cells, parietal epithelial cells (PECs) have demonstrated multilineage differentiation potential in response to kidney injury. However, the function of exosomes derived from PECs has not been extensively explored. Immunofluorescent staining of Claudin-1 was used to identify primary PECs isolated from mouse glomeruli. Transmission electron microscopy, nanoparticle tracking analysis, and western blotting were used to characterize the properties of PECs-derived exosomes (PEC-Exo). The therapeutic role of PEC-Exo in tubulointerstitial fibrosis was investigated in the unilateral ureteral obstruction (UUO) mouse model and TGF-ß1-stimulated HK-2 cells. High-throughput miRNA sequencing was employed to profile PEC-Exo miRNAs. One of the most enriched miRNAs in PEC-Exo was knocked down by transfecting miRNA inhibitor, and then we investigated whether this candidate miRNA was involved in PEC-Exo-mediated tubular repair. The primary PECs expressed Claudin-1, PEC-Exo was homing to obstructed kidney, and TGF-ß1 induced HK-2 cells. PEC-Exo significantly alleviated renal inflammation and ameliorated tubular fibrosis both in vivo and in vitro. Mechanistically, let-7b-5p, highly enriched in PEC-Exo, downregulated the protein levels of transforming growth factor beta receptor 1(TGFßR1) and AT-Rich Interaction Domain 3A(ARID3a) in tubular epithelial cells (TECs), leading to the inhibition of p21 and p27 to restoring cell cycle. Furthermore, administration of let-7b-5p agomir mitigated renal fibrosis in vivo. Our findings demonstrated that PEC-derived exosomes significantly repressed the expression of TGFßR1 and ARID3a by delivering let-7b-5p, thereby alleviating renal fibrosis. This study provides novel insights into the role of PEC-Exo in the repair of kidney injury and new ideas for renal fibrosis intervention.


Asunto(s)
Células Epiteliales , Exosomas , Fibrosis , MicroARNs , Receptor Tipo I de Factor de Crecimiento Transformador beta , Animales , MicroARNs/genética , MicroARNs/metabolismo , Ratones , Fibrosis/metabolismo , Exosomas/metabolismo , Células Epiteliales/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Humanos , Masculino , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Ratones Endogámicos C57BL , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/genética , Factor de Crecimiento Transformador beta1/metabolismo , Línea Celular
4.
Cell Commun Signal ; 22(1): 467, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39350161

RESUMEN

Traumatic brain injury (TBI) is an acquired insult to the brain caused by an external mechanical force, potentially resulting in temporary or permanent impairment. Microglia, the resident immune cells of the central nervous system, are activated in response to TBI, participating in tissue repair process. However, the underlying epigenetic mechanisms in microglia during TBI remain poorly understood. ARID1A (AT-Rich Interaction Domain 1 A), a pivotal subunit of the multi-protein SWI/SNF chromatin remodeling complex, has received little attention in microglia, especially in the context of brain injury. In this study, we generated a Arid1a cKO mouse line to investigate the potential roles of ARID1A in microglia in response to TBI. We found that glial scar formation was exacerbated due to increased microglial migration and a heightened inflammatory response in Arid1a cKO mice following TBI. Mechanistically, loss of ARID1A led to an up-regulation of the chemokine CCL5 in microglia upon the injury, while the CCL5-neutralizing antibody reduced migration and inflammatory response of LPS-stimulated Arid1a cKO microglia. Importantly, administration of auraptene (AUR), an inhibitor of CCL5, repressed the microglial migration and inflammatory response, as well as the glial scar formation after TBI. These findings suggest that ARID1A is critical for microglial response to injury and that AUR has a therapeutic potential for the treatment of TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Quimiocina CCL5 , Proteínas de Unión al ADN , Ratones Noqueados , Microglía , Factores de Transcripción , Animales , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/genética , Microglía/metabolismo , Microglía/patología , Quimiocina CCL5/metabolismo , Quimiocina CCL5/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ratones , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Movimiento Celular , Cicatriz/patología , Cicatriz/metabolismo , Ratones Endogámicos C57BL , Masculino
5.
J Exp Clin Cancer Res ; 43(1): 272, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39350229

RESUMEN

BACKGROUND: Cholangiocarcinoma (CCA) is a highly malignant, rapidly progressing tumor of the bile duct. Owing to its chemoresistance, it always has an extremely poor prognosis. Therefore, detailed elucidation of the mechanisms of chemoresistance and identification of therapeutic targets are still needed. METHODS: We analyzed the expression of MBD2 (Methyl-CpG-binding domain 2) in CCA and normal bile duct tissues using the public database and immunohistochemistry (IHC). The roles of MBD2 in CCA cell proliferation, migration, and chemoresistance ability were validated through CCK-8, plate cloning assay, wound healing assays and xenograft mouse model. In addition, we constructed a primary CCA mouse model to further confirm the effect of MBD2. RNA-seq and co-IP-MS were used to identify the mechanisms by how MBD2 leads to chemoresistance. RESULTS: MBD2 was upregulated in CCA. It promoted the proliferation, migration and chemoresistance of CCA cells. Mechanistically, MBD2 directly interacted with WDR5, bound to the promoter of ABCB1, promoted the trimethylation of H3K4 in this region through KMT2A, and activated the expression of ABCB1. Knocking down WDR5 or KMT2A blocked the transcriptional activation of ABCB1 by MBD2. The molecular inhibitor MM-102 targeted the interaction of WDR5 with KMT2A. MM-102 inhibited the expression of ABCB1 in CCA cells and decreased the chemoresistance of CCA to cisplatin. CONCLUSION: MBD2 promotes the progression and chemoresistance of CCA through interactions with WDR5. MM-102 can effectively block this process and increase the sensitivity of CCA to cisplatin.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Proteínas de Unión al ADN , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Péptidos y Proteínas de Señalización Intracelular , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/patología , Humanos , Animales , Ratones , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Proliferación Celular , Línea Celular Tumoral , Masculino , Femenino , Regulación Neoplásica de la Expresión Génica , Ensayos Antitumor por Modelo de Xenoinjerto , Movimiento Celular , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética
6.
J Clin Invest ; 134(19)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39352379

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) has emerged as a previously unrecognized, potent, age-related, and common risk factor for atherosclerosis. Somatic mutations in certain known leukemia driver genes give rise to clones of mutant cells in peripheral blood. The increased risk of developing hematologic malignancy does not, on its own, explain excess mortality in individuals with CHIP. Cardiovascular disease accounts for much of this gap. Experimental evidence supports the causality of certain CHIP mutations in accelerated atherosclerosis. CHIP due to mutations in different driver genes varies in their promotion of atherosclerotic events and in the region of augmented atherosclerotic involvement. For example, CHIP due to mutations in DNMT3a appears less atherogenic than CHIP that arises from TET2 or JAK2, forms of CHIP that incite inflammation. The recognition of certain CHIP mutations as promoters of atherosclerotic risk has opened new insights into understanding of the pathophysiology of this disease. The accentuated cardiovascular risk and involvement of distinct pathways of various forms of CHIP also inform novel approaches to allocation of targeted therapies, affording a step toward personalized medicine.


Asunto(s)
Aterosclerosis , Hematopoyesis Clonal , ADN Metiltransferasa 3A , Dioxigenasas , Mutación , Humanos , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Hematopoyesis Clonal/genética , ADN Metiltransferasa 3A/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Animales , Janus Quinasa 2/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Riesgo
7.
Rinsho Ketsueki ; 65(9): 954-960, 2024.
Artículo en Japonés | MEDLINE | ID: mdl-39358295

RESUMEN

EVI1 is a zinc finger transcription factor encoded by the MECOM locus and is essential for the development and maintenance of hematopoietic stem cells. However, overexpression of EVI1 in various myeloid malignancies is associated with aggressive clinical behavior and poor outcome. The locus encodes multiple isoforms that are differentially acting and independently regulated. EVI1 interacts with a variety of transcription and epigenetic factors via different domains. It also regulates cell survival, differentiation, and proliferation through a variety of mechanisms, including transcriptional activation and repression, regulation of other transcription factors' activity, and chromatin remodeling. While the mechanism by which 3q26 translocation leads to high EVI1 expression through enhancer hijacking of genes active in myeloid development is now better understood, regulation of EVI1 expression in the absence of chromosomal translocations and in normal hematopoiesis remains unclear. Recent studies have provided insight into the regulatory mechanisms of EVI1 expression and action, which may lead to development of targeted therapies in the near future.


Asunto(s)
Neoplasias Hematológicas , Hematopoyesis , Proteína del Locus del Complejo MDS1 y EV11 , Proteína del Locus del Complejo MDS1 y EV11/genética , Proteína del Locus del Complejo MDS1 y EV11/metabolismo , Humanos , Hematopoyesis/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Animales , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proto-Oncogenes/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica
8.
Elife ; 122024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39356734

RESUMEN

To function effectively as an integrated system, the transcriptional and post-transcriptional machineries must communicate through mechanisms that are still poorly understood. Here, we focus on the zinc-finger Sfp1, known to regulate transcription of proliferation-related genes. We show that Sfp1 can regulate transcription either by binding to promoters, like most known transcription activators, or by binding to the transcribed regions (gene bodies), probably via RNA polymerase II (Pol II). We further studied the first mode of Sfp1 activity and found that, following promoter binding, Sfp1 binds to gene bodies and affects Pol II configuration, manifested by dissociation or conformational change of its Rpb4 subunit and increased backtracking. Surprisingly, Sfp1 binds to a subset of mRNAs co-transcriptionally and stabilizes them. The interaction between Sfp1 and its client mRNAs is controlled by their respective promoters and coincides with Sfp1's dissociation from chromatin. Intriguingly, Sfp1 dissociation from the chromatin correlates with the extent of the backtracked Pol II. We propose that, following promoter recruitment, Sfp1 accompanies Pol II and regulates backtracking. The backtracked Pol II is more compatible with Sfp1's relocation to the nascent transcripts, whereupon Sfp1 accompanies these mRNAs to the cytoplasm and regulates their stability. Thus, Sfp1's co-transcriptional binding imprints the mRNA fate, serving as a paradigm for the cross-talk between the synthesis and decay of specific mRNAs, and a paradigm for the dual-role of some zinc-finger proteins. The interplay between Sfp1's two modes of transcription regulation remains to be examined.


The ability to fine-tune the production of proteins in a cell is essential for organisms to exist. An imbalance in protein levels can be the cause of various diseases. Messenger RNA molecules (mRNA) link the genetic information encoded in DNA and the produced proteins. Exactly how much protein is made mostly depends on the amount of mRNA in the cell's cytoplasm. This is controlled by two processes: the synthesis of mRNA (also known as transcription) and mRNA being actively degraded. Although much is known about mechanisms regulating transcription and degradation, how cells detect if they need to degrade mRNA based on the levels of its synthesis and vice versa is poorly understood. In 2013, researchers found that proteins known as 'RNA decay factors' responsible for mRNA degradation are actively moved from the cell's cytoplasm into its nucleus to instruct the transcription machinery to produce more mRNA. Kelbert, Jordán-Pla, de-Miguel-Jiménez et al. ­ including some of the researchers involved in the 2013 work ­ investigated how mRNA synthesis and degradation are coordinated to ensure a proper mRNA level. The researchers used advanced genome engineering methods to carefully manipulate and measure mRNA production and degradation in yeast cells. The experiments revealed that the protein Sfp1 ­ a well-characterized transcription factor for stimulating the synthesis of a specific class of mRNAs inside the nucleus ­ can also prevent the degradation of these mRNAs outside the nucleus. During transcription, Sfp1 bound directly to mRNA. The investigators could manipulate the co-transcriptional binding of Sfp1 to a certain mRNA, thereby changing the mRNA stability in the cytoplasm. This suggests that the ability of Sfp1 to regulate both the production and decay of mRNA is dependent on one another and that transcription can influence the fate of its transcripts. This combined activity can rapidly change mRNA levels in response to changes in the cell's environment. RNA plays a key role in ensuring correct levels of proteins. It can also function as an RNA molecule, independently of its coding capacity. Many cancers and developmental disorders are known to be caused by faulty interactions between transcription factors and nucleic acids. The finding that some transcription factors can directly regulate both mRNA synthesis and its destruction introduces new angles for studying and understanding these diseases.


Asunto(s)
ARN Polimerasa II , ARN Mensajero , Factores de Transcripción , ARN Mensajero/metabolismo , ARN Mensajero/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Estabilidad del ARN , Regiones Promotoras Genéticas , Unión Proteica , Dedos de Zinc , Transcripción Genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Citoplasma/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae
9.
FASEB J ; 38(19): e70081, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39360635

RESUMEN

Rho guanine nucleotide exchange factor (RGNEF) is a guanine nucleotide exchange factor (GEF) mainly involved in regulating the activity of Rho-family GTPases. It is a bi-functional protein, acting both as a guanine exchange factor and as an RNA-binding protein. RGNEF is known to act as a destabilizing factor of neurofilament light chain RNA (NEFL) and it could potentially contribute to their sequestration in nuclear cytoplasmic inclusions. Most importantly, RGNEF inclusions in the spinal motor neurons of ALS patients have been shown to co-localize with inclusions of TDP-43, the major well-known RNA-binding protein aggregating in the brain and spinal cord of human patients. Therefore, it can be hypothesized that loss-of-function of both proteins following aggregation may contribute to motor neuron death/survival in ALS patients. To further characterize their relationship, we have compared the transcriptomic profiles of neuronal cells depleted of TDP-43 and RGNEF and show that these two factors predominantly act in an antagonistic manner when regulating the expression of axon guidance genes. From a mechanistic point of view, our experiments show that the effect of these genes on the processivity of long introns can explain their mode of action. Taken together, our results show that loss-of-function of factors co-aggregating with TDP-43 can potentially affect the expression of commonly regulated neuronal genes in a very significant manner, potentially acting as disease modifiers. This finding further highlights that neurodegenerative processes at the RNA level are the result of combinatorial interactions between different RNA-binding factors that can be co-aggregated in neuronal cells. A deeper understanding of these complex scenarios may lead to a better understanding of pathogenic mechanisms occurring in patients, where more than one specific protein may be aggregating in their neurons.


Asunto(s)
Proteínas de Unión al ADN , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Humanos , Intrones , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Animales , Orientación del Axón/genética , Neuronas Motoras/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Regulación de la Expresión Génica
10.
Proc Natl Acad Sci U S A ; 121(41): e2407647121, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39361642

RESUMEN

Memory effect refers to the phenomenon where past events influence a system's current and future states or behaviors. In biology, memory effects often arise from intra- or intermolecular interactions, leading to temporally correlated behaviors. Single-molecule studies have shown that enzymes and DNA-binding proteins can exhibit time-correlated behaviors of their activity. While memory effects are well documented and studied in vitro, no such examples exist in cells to our knowledge. Combining single-molecule tracking (SMT) and single-cell protein quantitation, we find in living Escherichia coli cells distinct temporal correlations in the binding/unbinding events on DNA by MerR- and Fur-family metalloregulators, manifesting as memory effects with timescales of ~1 s. These memory effects persist irrespective of the type of the metalloregulators or their metallation states. Moreover, these temporal correlations of metalloregulator-DNA interactions are associated with spatial confinements of the metalloregulators near their DNA binding sites, suggesting microdomains of ~100 nm in size that possibly result from the spatial organizations of the bacterial chromosome without the involvement of membranes. These microdomains likely facilitate repeated binding events, enhancing regulator-DNA contact frequency and potentially gene regulation efficiency. These findings provide unique insights into the spatiotemporal dynamics of protein-DNA interactions in bacterial cells, introducing the concept of microdomains as a crucial player in memory effect-driven gene regulation.


Asunto(s)
ADN Bacteriano , Proteínas de Escherichia coli , Escherichia coli , Escherichia coli/metabolismo , Escherichia coli/genética , ADN Bacteriano/metabolismo , ADN Bacteriano/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Regulación Bacteriana de la Expresión Génica , Unión Proteica , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Sitios de Unión
11.
Mol Biol (Mosk) ; 58(2): 305-313, 2024.
Artículo en Ruso | MEDLINE | ID: mdl-39355887

RESUMEN

An RNA interference-based method was proposed to achieve an inducible knockdown of genes essential for cell viability. In the method, a genetic cassette in which a copper ion-dependent inducible metallothionein promoter controls expression of a siRNA precursor is inserted into a genomic pre-integrated transgene by CRIPSR/Cas9 technology. The endogenous siRNA source allows the gene knockdown in cell cultures that are refractory to conventional transfection with exogenous siRNA. The efficiency of the method was demonstrated in Drosophila ovarian somatic cell culture (OSC) for two genes that are essential for oogenesis: Cul3, encoding a component of the multiprotein ubiquitin-ligase complex with versatile functions in proteostasis, and cut, encoding a transcription factor regulating differentiation of ovarian follicular cells.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Técnicas de Silenciamiento del Gen , Animales , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ovario/metabolismo , Ovario/citología , Oogénesis/genética , Interferencia de ARN , Genes Esenciales , Sistemas CRISPR-Cas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo
12.
Acta Neuropathol Commun ; 12(1): 156, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39363348

RESUMEN

Abnormal cytoplasmic localization and accumulation of pathological transactive response DNA binding protein of 43 kDa (TDP-43) underlies several devastating diseases such as amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration with TDP-43 pathology (FTLD-TDP). A key element is the correlation between disease progression and spatio-temporal propagation of TDP-43-mediated pathology in the central nervous system. Several lines of evidence support the concept of templated aggregation and cell to cell spreading of pathological TDP-43. To further investigate this mechanism in vivo, we explored the efficacy of capturing and masking the seeding-competent region of extracellular TDP-43 species. For this, we generated a novel monoclonal antibody (mAb), ACI-6677, that targets the pathogenic protease-resistant amyloid core of TDP-43. ACI-6677 has a picomolar binding affinity for TDP-43 and is capable of binding to all C-terminal TDP-43 fragments. In vitro, ACI-6677 inhibited TDP-43 aggregation and boosted removal of pathological TDP-43 aggregates by phagocytosis. When injecting FTLD-TDP brain extracts unilaterally in the CamKIIa-hTDP-43NLSm mouse model, ACI-6677 significantly limited the induction of phosphorylated TDP-43 (pTDP-43) inclusions. Strikingly, on the contralateral side, the mAb significantly prevented pTDP-43 inclusion appearance exemplifying blocking of the spreading process. Taken together, these data demonstrate for the first time that an immunotherapy targeting the protease-resistant amyloid core of TDP-43 has the potential to restrict spreading, substantially slowing or stopping progression of disease.


Asunto(s)
Esclerosis Amiotrófica Lateral , Anticuerpos Monoclonales , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Demencia Frontotemporal , Animales , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Unión al ADN/metabolismo , Ratones , Demencia Frontotemporal/patología , Demencia Frontotemporal/metabolismo , Anticuerpos Monoclonales/farmacología , Humanos , Ratones Transgénicos
13.
J Cell Mol Med ; 28(19): e70130, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39365284

RESUMEN

Pancreatic ß-cell damage mediated by apoptosis is believed to be a main trigger of type 1 diabetes mellitus (T1DM), which is proposed as an organ-specific autoimmune disease mediated by T cells. Nonetheless, the fundamental origins of T1DM remain uncertain. Here, we illustrate that an increase in PLAGL1 expression induces ß-cell apoptosis, as evidenced by mitochondrial membrane impairment and nucleolar degradation. The gene expression levels from cDNA samples were determined using qRT-PCR method. Western blot and Co-immunoprecipitation were applied for protein expression and interactions, respectively. Flow cytometry and TUNEL assay were used to detect pancreatic ß cell apoptosis. Female NOD/LtJ mice with recent-onset T1DM has been used in in vivo studies. Glucose-stimulated insulin secretion (GSIS) and glucose tolerance test (GTT) method is used for islet function assessment. Haematoxylin and Eosin (H&E) and Immunohistochemistry (IHC) were performed to evalute histological improvement of islet beta. Subsequent cytoplasmic DNA accumulation triggers DNA senser, the cyclic guanosine monophosphate-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. STING activation further stimulates downstream IRF3 and NF-kB pathways, thus boost type-I interferon signalling and NF-kB mediated inflammation. These findings elucidate a molecular mechanism linking PLAGL1 induced cell apoptosis to type-I interferon signalling and suggest a potential benefit for targeting cGAS/STING in T1DM treatment.


Asunto(s)
Apoptosis , Células Secretoras de Insulina , Proteínas de la Membrana , Nucleotidiltransferasas , Animales , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Femenino , Transducción de Señal , Citoplasma/metabolismo , ADN/metabolismo , ADN/genética , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Ratones Endogámicos NOD , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , FN-kappa B/metabolismo
14.
Nat Commun ; 15(1): 8511, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39353945

RESUMEN

Nucleotide excision repair (NER) is vital for genome integrity. Yet, our understanding of the complex NER protein machinery remains incomplete. Combining cryo-EM and XL-MS data with AlphaFold2 predictions, we build an integrative model of the NER pre-incision complex(PInC). Here TFIIH serves as a molecular ruler, defining the DNA bubble size and precisely positioning the XPG and XPF nucleases for incision. Using simulations and graph theoretical analyses, we unveil PInC's assembly, global motions, and partitioning into dynamic communities. Remarkably, XPG caps XPD's DNA-binding groove and bridges both junctions of the DNA bubble, suggesting a novel coordination mechanism of PInC's dual incision. XPA rigging interlaces XPF/ERCC1 with RPA, XPD, XPB, and 5' ssDNA, exposing XPA's crucial role in licensing the XPF/ERCC1 incision. Mapping disease mutations onto our models reveals clustering into distinct mechanistic classes, elucidating xeroderma pigmentosum and Cockayne syndrome disease etiology.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN , Endonucleasas , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/química , Humanos , Endonucleasas/metabolismo , Endonucleasas/genética , Factor de Transcripción TFIIH/metabolismo , Factor de Transcripción TFIIH/química , Factor de Transcripción TFIIH/genética , Proteína de la Xerodermia Pigmentosa del Grupo D/metabolismo , Proteína de la Xerodermia Pigmentosa del Grupo D/genética , Proteína de la Xerodermia Pigmentosa del Grupo D/química , Microscopía por Crioelectrón , Proteína de la Xerodermia Pigmentosa del Grupo A/metabolismo , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Unión Proteica , ADN/metabolismo , ADN/química , ADN/genética , Proteína de Replicación A/metabolismo , Proteína de Replicación A/genética , Modelos Moleculares , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/genética , Reparación por Escisión , Proteínas Nucleares
15.
Sci Rep ; 14(1): 22910, 2024 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-39358423

RESUMEN

Studies of proteins, found in one of the most stress-resistant animals tardigrade Ramazzottius varieornatus, aim to reveal molecular principles of extreme tolerance to various types of stress and developing applications based on them for medicine, biotechnology, pharmacy, and space research. Tardigrade DNA/RNA-binding damage suppressor protein (Dsup) reduces DNA damage caused by reactive oxygen spices (ROS) produced upon irradiation and oxidative stresses in Dsup-expressing transgenic organisms. This work is focused on the determination of structural features of Dsup protein and Dsup-DNA complex, which refines details of protective mechanism. For the first time, intrinsically disordered nature of Dsup protein with highly flexible structure was experimentally proven and characterized by the combination of small angle X-ray scattering (SAXS) technique, circular dichroism spectroscopy, and computational methods. Low resolution models of Dsup protein and an ensemble of conformations were presented. In addition, we have shown that Dsup forms fuzzy complex with DNA.


Asunto(s)
ADN , Proteínas Intrínsecamente Desordenadas , Tardigrada , Animales , Tardigrada/metabolismo , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas Intrínsecamente Desordenadas/genética , ADN/metabolismo , ADN/química , Daño del ADN , Dispersión del Ángulo Pequeño , Dicroismo Circular , Modelos Moleculares , Unión Proteica , Difracción de Rayos X , Conformación Proteica , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética
16.
Sci Rep ; 14(1): 23363, 2024 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-39375425

RESUMEN

This study aimed to investigate the activation of error-prone DNA repair pathway in response to Helicobacter pylori infection. Relative changes in the expression levels of genes involved in the non-homologous end-joining pathway (NHEJ) in H. pylori-infected (Cases) and non-infected patients (Controls) with chronic gastritis were measured. A significant increase in the relative expression level of TP53, and significant decrease in the relative transcription of lncRNA LINP1 and XRCC5 were detected in the case group. The transcription of Lig4 and XRCC6 was increased in the case group, which was not statistically significant. The Spearman's Correlation Coefficient analysis showed a significant positive-correlation between the transcriptional levels of LINP1 and XRCC4/XRCC5/Lig4, and between XRCC5 and TP53/Lig4 both in the case and control groups. Moreover, a significant positive correlation between LinP1 and XRCC6 in the case, and a significant positive correlation between XRCC4 and Lig4, and a negative correlation between TP53 and LinP1/XRCC4/XRCC5 in the control group was detected. Although a relative difference was detected in transcriptional levels of the NHEJ gene mediators, downregulation of LinP1 in H. pylori-infected patients proposed the activation of a negative feedback loop, which may interfere with the NHEJ activity at the early stages of gastritis.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Gastritis , Infecciones por Helicobacter , Helicobacter pylori , Proteína p53 Supresora de Tumor , Humanos , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/metabolismo , Gastritis/microbiología , Gastritis/genética , Gastritis/metabolismo , Masculino , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Femenino , Persona de Mediana Edad , Adulto , ADN Ligasa (ATP)/metabolismo , ADN Ligasa (ATP)/genética , Autoantígeno Ku/metabolismo , Autoantígeno Ku/genética , Retroalimentación Fisiológica , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética
17.
Commun Biol ; 7(1): 1280, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39379486

RESUMEN

D-site binding protein, DBP, is a clock-controlled transcription factor and drives daily rhythms of physiological processes through the regulation of an array of genes harboring a DNA binding motif, D-box. DBP protein levels show a circadian oscillation with an extremely robust peak/trough ratio, but it is elusive how the temporal pattern is regulated by post-translational regulation. In this study, we show that DBP protein levels are down-regulated by the ubiquitin-proteasome pathway. Analysis using 19 dominant-negative forms of E2 enzymes have revealed that UBE2G1 and UBE2T mediate the degradation of DBP. A proteomic analysis of DBP-interacting proteins and database screening have identified Tumor necrosis factor Receptor-Associated Factor 7 (TRAF7), a RING-type E3 ligase, that forms a complex with UBE2G1 and/or UBE2T. Ubiquitination analysis have revealed that TRAF7 enhances K48-linked polyubiquitination of DBP in cultured cells. Overexpression of TRAF7 down-regulates DBP protein level, while knockdown of TRAF7 up-regulates DBP in cultured cells. Knockout of TRAF7 in NIH3T3 cells have revealed that TRAF7 mediates the time-of-the-day-dependent regulation of DBP levels. Furthermore, TRAF7 has a period-shortening effect on the cellular clock. Together, TRAF7 plays an important role in circadian clock oscillation through destabilization of DBP.


Asunto(s)
Ritmo Circadiano , Ubiquitinación , Animales , Ratones , Células 3T3 NIH , Ritmo Circadiano/genética , Humanos , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Proteolisis , Relojes Circadianos/genética
18.
Immunity ; 57(10): 2263-2265, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39383842

RESUMEN

It's crucial for skin to establish efficient defense strategies. Liu et al. reveal that the transcription factor ZNF750 recruits the histone demethylase KDM1A to silence pattern recognition receptors in the outer epidermis, making their expression limited to deeper, undifferentiated keratinocytes to address threats penetrating the skin.


Asunto(s)
Queratinocitos , Piel , Factores de Transcripción , Humanos , Queratinocitos/metabolismo , Queratinocitos/inmunología , Piel/inmunología , Piel/metabolismo , Animales , Factores de Transcripción/metabolismo , Histona Demetilasas/metabolismo , Epidermis/metabolismo , Epidermis/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Proteínas de Unión al ADN/metabolismo
19.
Oncotarget ; 15: 750-763, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39392394

RESUMEN

This study presents an observational, cross-sectional analysis of 64 patients diagnosed with small cell lung cancer (SCLC) at a reference laboratory for thoracic pathology between 2022 and 2024. The primary objective was to evaluate the expression of Delta-like ligand 3 (DLL3) and other neuroendocrine markers such as Chromogranin, and Synaptophysin, utilizing both traditional immunohistochemistry and digital pathology tools. Patients were primarily older adults, with a median age of over 71, and most biopsies were obtained from lung parenchyma. Immunohistochemistry (IHC) was performed using specific monoclonal antibodies, with DLL3 showing variable expression across the samples. Notably, DLL3 was expressed in 72.3% of the cases, with varied intensities and a semi-quantitative H-score applied for more nuanced analysis. ASCL1 was expressed in 97% of cases, with the majority considered low-expressors. Only 11% had high expression. TTF-1, traditionally not a conventional marker for the diagnosis of SCLC, was positive in half of the cases, suggesting its potential as a biomarker. The study underscores the significant variability in the expression of neuroendocrine markers in SCLC, with implications for both diagnosis and potential therapeutic targeting. DLL3, particularly, shows promise as a therapeutic target due to its high expression rate in the cohort. The use of digital pathology software QuPath enhanced the accuracy and depth of analysis, allowing for detailed morphometric analysis and potentially informing more personalized treatment approaches. The findings emphasize the need for further research into the role of these markers in the management and treatment of SCLC, considering the poor prognosis and high mortality rate observed in the cohort.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Biomarcadores de Tumor , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Pulmonares , Proteínas de la Membrana , Medicina de Precisión , Carcinoma Pulmonar de Células Pequeñas , Factores de Transcripción , Humanos , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/mortalidad , Masculino , Femenino , Anciano , Persona de Mediana Edad , Biomarcadores de Tumor/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Medicina de Precisión/métodos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Anciano de 80 o más Años , Factores de Transcripción/metabolismo , Estudios Transversales , Inmunohistoquímica , Proteínas de Unión al ADN/metabolismo , Adulto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA