Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(8)2023 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-37108510

RESUMO

Chronic inflammatory processes in the intestine result in serious conditions such as inflammatory bowel disease (IBD) and cancer. An increased detection of cytoplasmic DNA sensors has been reported in the IBD colon mucosa, suggesting their contribution in mucosal inflammation. Yet, the mechanisms altering DNA homeostasis and triggering the activation of DNA sensors remain poorly understood. In this study, we show that the epigenetic regulator HP1γ plays a role in preserving nuclear envelope and genomic integrity in enterocytic cells, thereby protecting against the presence of cytoplasmic DNA. Accordingly, HP1 loss of function led to the increased detection of cGAS/STING, a cytoplasmic DNA sensor that triggers inflammation. Thus, in addition to its role as a transcriptional silencer, HP1γ may also exert anti-inflammatory properties by preventing the activation of the endogenous cytoplasmic DNA response in the gut epithelium.


Assuntos
Adenocarcinoma , Neoplasias do Colo , Doenças Inflamatórias Intestinais , Humanos , Membrana Nuclear/metabolismo , Transdução de Sinais , Adenocarcinoma/genética , Neoplasias do Colo/genética , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Inflamação/patologia , DNA , Genômica
2.
Nat Commun ; 13(1): 6834, 2022 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-36400769

RESUMO

Defects in RNA splicing have been linked to human disorders, but remain poorly explored in inflammatory bowel disease (IBD). Here, we report that expression of the chromatin and alternative splicing regulator HP1γ is reduced in ulcerative colitis (UC). Accordingly, HP1γ gene inactivation in the mouse gut epithelium triggers IBD-like traits, including inflammation and dysbiosis. In parallel, we find that its loss of function broadly increases splicing noise, favoring the usage of cryptic splice sites at numerous genes with functions in gut biology. This results in the production of progerin, a toxic splice variant of prelamin A mRNA, responsible for the Hutchinson-Gilford Progeria Syndrome of premature aging. Splicing noise is also extensively detected in UC patients in association with inflammation, with progerin transcripts accumulating in the colon mucosa. We propose that monitoring HP1γ activity and RNA splicing precision can help in the management of IBD and, more generally, of accelerated aging.


Assuntos
Colite Ulcerativa , Progéria , Humanos , Camundongos , Animais , Homólogo 5 da Proteína Cromobox , Colite Ulcerativa/genética , Splicing de RNA/genética , Progéria/genética , Progéria/metabolismo , Inflamação
3.
Biomedicines ; 10(4)2022 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-35453483

RESUMO

Head and neck squamous cell carcinoma is the sixth leading cancer in the world. This cancer is difficult to treat and is characterized by recurrences that are often fatal. This cancer is generally removed surgically, but it often regrows from the edges of the lesion from where most recurrences reappear. In this study, we have investigated if the expression of GB3 in human cell lines, tissues from patient biopsies, and a murine animal model could be used as an early and determinant marker of HNC. We found that in all the investigated systems, this marker appears in neoplastic cells from the very early stages of their malignant transformation. Our conclusions support the hypothesis that GB3 is a reliable and independent target for HNC identification and selective delivery of treatments. Furthermore, we show that the level of expression of this marker correlates with the degree of malignancy of the tumor. These studies suggest that GB3 may provide the basis for the early identification and new targeted therapies for head and neck cancer.

4.
Bioact Mater ; 8: 153-164, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34541393

RESUMO

Nowadays, a number of promising strategies are being developed that aim at combining diagnostic and therapeutic capabilities into clinically effective formulations. Thus, the combination of a modified release provided by an organic encapsulation and the intrinsic physico-chemical properties from an inorganic counterpart opens new perspectives in biomedical applications. Herein, a biocompatible magnetic lipid nanocomposite vehicle was developed through an efficient, green and simple method to simultaneously incorporate magnetic nanoparticles and an anticancer drug (doxorubicin) into a natural nano-matrix. The theranostic performance of the final magnetic formulation was validated in vitro and in vivo, in melanoma tumors. The systemic administration of the proposed magnetic hybrid nanocomposite carrier enhanced anti-tumoral activity through a synergistic combination of magnetic hyperthermia effects and antimitotic therapy, together with MRI reporting capability. The application of an alternating magnetic field was found to play a dual role, (i) acting as an extra layer of control (remote, on-demand) over the chemotherapy release and (ii) inducing a local thermal ablation of tumor cells. This combination of chemotherapy with thermotherapy establishes a synergistic platform for the treatment of solid malignant tumors under lower drug dosing schemes, which may realize the dual goal of reduced systemic toxicity and enhanced anti-tumoral efficacy.

5.
Acta Neuropathol Commun ; 6(1): 68, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-30049290

RESUMO

Neurons are highly vulnerable to DNA damage induced by genotoxic agents such as topoisomerase activity, oxidative stress, ionizing radiation (IR) and chemotherapeutic drugs. To avert the detrimental effects of DNA lesions in genome stability, transcription and apoptosis, neurons activate robust DNA repair mechanisms. However, defective DNA repair with accumulation of unrepaired DNA are at the basis of brain ageing and several neurodegenerative diseases. Understanding the mechanisms by which neurons tolerate DNA damage accumulation as well as defining the genomic regions that are more vulnerable to DNA damage or refractory to DNA repair and therefore constitute potential targets in neurodegenerative diseases are essential issues in the field. In this work we investigated the nuclear topography and organization together with the genome-wide distribution of unrepaired DNA in rat cortical neurons 15 days upon IR. About 5% of non-irradiated and 55% of irradiated cells accumulate unrepaired DNA within persistent DNA damage foci (PDDF) of chromatin. These PDDF are featured by persistent activation of DNA damage/repair signaling, lack of transcription and localization in repressive nuclear microenvironments. Interestingly, the chromatin insulator CTCF is concentrated at the PDDF boundaries, likely contributing to isolate unrepaired DNA from intact transcriptionally active chromatin. By confining damaged DNA, PDDF would help preserving genomic integrity and preventing the production of aberrant proteins encoded by damaged genes.ChIP-seq analysis of genome-wide γH2AX distribution revealed a number of genomic regions enriched in γH2AX signal in IR-treated cortical neurons. Some of these regions are in close proximity to genes encoding essential proteins for neuronal functions and human neurodegenerative disorders such as epm2a (Lafora disease), serpini1 (familial encephalopathy with neuroserpin inclusion bodies) and il1rpl1 (mental retardation, X-linked 21). Persistent γH2AX signal close to those regions suggests that nearby genes could be either more vulnerable to DNA damage or more refractory to DNA repair.


Assuntos
Núcleo Celular/metabolismo , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Neurônios/efeitos da radiação , Raios X/efeitos adversos , Animais , Antineoplásicos/uso terapêutico , Proteínas de Ligação ao Cálcio/metabolismo , Núcleo Celular/efeitos da radiação , Córtex Cerebral/citologia , Imunoprecipitação da Cromatina , Reparo do DNA/efeitos dos fármacos , Ácido Fólico/análogos & derivados , Ácido Fólico/uso terapêutico , Regulação da Expressão Gênica/efeitos da radiação , Proteína Glial Fibrilar Ácida/metabolismo , Histonas/metabolismo , Masculino , Proteínas dos Microfilamentos/metabolismo , Microscopia Eletrônica , Neurônios/ultraestrutura , Quinazolinas/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
Acta Neuropathol Commun ; 4: 41, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-27102221

RESUMO

There is growing evidence that defective DNA repair in neurons with accumulation of DNA lesions and loss of genome integrity underlies aging and many neurodegenerative disorders. An important challenge is to understand how neurons can tolerate the accumulation of persistent DNA lesions without triggering the apoptotic pathway. Here we study the impact of the accumulation of unrepaired DNA on the chromatin architecture, kinetics of the DNA damage response and transcriptional activity in rat sensory ganglion neurons exposed to 1-to-3 doses of ionizing radiation (IR). In particular, we have characterized the structural, molecular and transcriptional compartmentalization of unrepaired DNA in persistent DNA damaged foci (PDDF). IR induced the formation of numerous transient foci, which repaired DNA within the 24 h post-IR, and a 1-to-3 PDDF. The latter concentrate DNA damage signaling and repair factors, including γH2AX, pATM, WRAP53 and 53BP1. The number and size of PDDF was dependent on the doses of IR administered. The proportion of neurons carrying PDDF decreased over time of post-IR, indicating that a slow DNA repair occurs in some foci. The fine structure of PDDF consisted of a loose network of unfolded 30 nm chromatin fiber intermediates, which may provide a structural scaffold accessible for DNA repair factors. Furthermore, the transcription assay demonstrated that PDDF are transcriptionally silent, although transcription occurred in flanking euchromatin. Therefore, the expression of γH2AX can be used as a reliable marker of gene silencing in DNA damaged neurons. Moreover, PDDF were located in repressive nuclear environments, preferentially in the perinucleolar domain where they were frequently associated with Cajal bodies or heterochromatin clumps forming a structural triad. We propose that the sequestration of unrepaired DNA in discrete PDDF and the transcriptional silencing can be essential to preserve genome stability and prevent the synthesis of aberrant mRNA and protein products encoded by damaged genes.


Assuntos
Cromatina/metabolismo , Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Regulação da Expressão Gênica/fisiologia , Neurônios/ultraestrutura , Fatores Etários , Animais , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Cromatina/efeitos da radiação , Cromatina/ultraestrutura , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta à Radiação , Gânglios Sensitivos/citologia , Regulação da Expressão Gênica/efeitos da radiação , Histonas/genética , Histonas/metabolismo , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/efeitos da radiação , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Lesões por Radiação , Ratos , Ratos Sprague-Dawley , Telomerase/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Raios X/efeitos adversos
7.
Adv Healthc Mater ; 5(9): 1080-7, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26866927

RESUMO

Understanding the molecular mechanisms underlying the biosynthetic interactions between particular nanomaterials with specific cells or proteins opens new alternatives in nanomedicine and nanotoxicology. Multiwalled carbon nanotubes (MWCNTs) have long been explored as drug delivery systems and nanomedicines against cancer. There are high expectations for their use in therapy and diagnosis. These filaments can translocate inside cultured cells and intermingle with the protein nanofilaments of the cytoskeleton, interfering with the biomechanics of cell division mimicking the effect of traditional microtubule-binding anti-cancer drugs such as paclitaxel. Here, it is shown how MWCNTs can trigger significant anti-tumoral effects in vivo, in solid malignant melanomas produced by allograft transplantation. Interestingly, the MWCNT anti-tumoral effects are maintained even in solid melanomas generated from paclitaxel-resistant cells. These findings provide great expectation in the development of groundbreaking adjuvant synthetic microtubule-stabilizing chemotherapies to overcome drug resistance in cancer.


Assuntos
Portadores de Fármacos , Melanoma/tratamento farmacológico , Nanotubos de Carbono/química , Neoplasias Experimentais/tratamento farmacológico , Paclitaxel , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Paclitaxel/química , Paclitaxel/farmacologia
8.
Mol Neurobiol ; 53(10): 6799-6808, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26660115

RESUMO

Neurons are highly vulnerable to genotoxic agents. To restore genome integrity upon DNA lesions, neurons trigger a DNA damage response (DDR) that requires chromatin modifications and transcriptional silencing at DNA damage sites. To study the reorganization of the active RNA polymerase II (Pol II), which transcribes all mRNA-encoding genes, and the participation of the ubiquitin-proteasome system (UPS) in the neuronal DDR, we have used rat sensory ganglion neurons exposed to X-rays (4 Gy) ionizing radiation (IR). In control neurons, Pol II appears concentrated in numerous chromatin microfoci identified as transcription factories by the incorporation of 5'-fluorouridine into nascent RNA. Upon IR treatment, numerous IR-induced foci (IRIF), which were immunoreactive for γH2AX and 53BP1, were observed as early as 30 min post-IR; their number progressively reduced at 3 h, 1 day, and 3 days post-IR. The formation of IRIF was associated with a decrease in Pol II levels by both immunofluorescence and Western blotting. Treatment with the proteasome inhibitor bortezomib strongly increased Pol II levels in both control and irradiated neurons, suggesting that proteasome plays a proteolytic role by clearing stalled Pol II complexes at DNA damage sites, as a prelude to DNA repair. Neuronal IRIF recruited ubiquitylated proteins, including ubiquitylated histone H2A (Ub-H2A), and the catalytic proteasome 20S. Ub-H2A has been associated with transcriptional silencing at DNA damage sites. On the other hand, the participation of UPS in neuronal DDR may be essential for the ubiquitylation of Pol II and other proteasome substrates of the DNA repair machinery and their subsequent proteasome-mediated degradation.


Assuntos
Dano ao DNA , Neurônios/patologia , Neurônios/efeitos da radiação , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Polimerase II/metabolismo , Radiação Ionizante , Ubiquitina/metabolismo , Animais , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Regulação para Baixo/efeitos da radiação , Eucromatina/metabolismo , Masculino , Neurônios/metabolismo , Ratos Sprague-Dawley , Transcrição Gênica/efeitos da radiação , Proteínas Ubiquitinadas/metabolismo
9.
Biochim Biophys Acta ; 1842(6): 848-59, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24269586

RESUMO

The dysfunction of the ubiquitin proteasome system has been related to a broad array of neurodegenerative disorders in which the accumulation of misfolded protein aggregates causes proteotoxicity. The ability of proteasome inhibitors to induce cell cycle arrest and apoptosis has emerged as a powerful strategy for cancer therapy. Bortezomib is a proteasome inhibitor used as an antineoplastic drug, although its neurotoxicity frequently causes a severe sensory peripheral neuropathy. In this study we used a rat model of bortezomib treatment to study the nucleolar and Cajal body responses to the proteasome inhibition in sensory ganglion neurons that are major targets of bortezomib-induced neurotoxicity. Treatment with bortezomib induced dose-dependent dissociation of protein synthesis machinery (chromatolysis) and nuclear retention of poly(A) RNA granules resulting in neuronal dysfunction. However, as a compensatory response to the proteotoxic stress, both nucleoli and Cajal bodies exhibited reactive changes. These include an increase in the number and size of nucleoli, strong nucleolar incorporation of the RNA precursor 5'-fluorouridine, and increased expression of both 45S rRNA and genes encoding nucleolar proteins UBF, fibrillarin and B23. Taken together, these findings appear to reflect the activation of the nucleolar transcription in response to proteotoxic stress Furthermore, the number of Cajal bodies, a parameter related to transcriptional activity, increases upon proteasome inhibition. We propose that nucleoli and Cajal bodies are important targets in the signaling pathways that are activated by the proteotoxic stress response to proteasome inhibition. The coordinating activity of these two organelles in the production of snRNA, snoRNA and rRNA may contribute to neuronal survival after proteasome inhibition. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.


Assuntos
Corpos Enovelados/metabolismo , Gânglios Sensitivos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Ácidos Borônicos/administração & dosagem , Bortezomib , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Nucléolo Celular/metabolismo , Núcleo Celular , Citoplasma/metabolismo , Gânglios Sensitivos/crescimento & desenvolvimento , Humanos , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Pirazinas/administração & dosagem , Ratos , Transdução de Sinais/efeitos dos fármacos
10.
Cell Mol Life Sci ; 71(10): 1961-75, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24061536

RESUMO

Bortezomib is a reversible proteasome inhibitor used as an anticancer drug. However, its clinical use is limited since it causes peripheral neurotoxicity. We have used Sprague-Dawley rats as an animal model to investigate the cellular mechanisms affected by both short-term and chronic bortezomib treatments in sensory ganglia neurons. Proteasome inhibition induces dose-dependent alterations in the architecture, positioning, shape and polarity of the neuronal nucleus. It also produces DNA damage without affecting neuronal survival, and severe disruption of the protein synthesis machinery at the central cytoplasm accompanied by decreased expression of the brain-derived neurotrophic factor. As a compensatory or adaptive survival response against proteotoxic stress caused by bortezomib treatment, sensory neurons preserve basal levels of transcriptional activity, up-regulate the expression of proteasome subunit genes, and generate a new cytoplasmic perinuclear domain for protein synthesis. We propose that proteasome activity is crucial for controlling nuclear architecture, DNA repair and the organization of the protein synthesis machinery in sensory neurons. These neurons are primary targets of bortezomib neurotoxicity, for which reason their dysfunction may contribute to the pathogenesis of the bortezomib-induced peripheral neuropathy in treated patients.


Assuntos
Núcleo Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Gânglios Sensitivos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Animais , Antineoplásicos/farmacologia , Ácidos Borônicos/farmacologia , Bortezomib , Núcleo Celular/fisiologia , Reparo do DNA/efeitos dos fármacos , Gânglios Sensitivos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Masculino , Microscopia Confocal , Corpos de Nissl/efeitos dos fármacos , Corpos de Nissl/fisiologia , Complexo de Endopeptidases do Proteassoma/química , Complexo de Endopeptidases do Proteassoma/genética , Pirazinas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
11.
Gene ; 532(2): 165-72, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24096177

RESUMO

We reported previously that the expression of Wnt-related genes is lower in osteoporotic hip fractures than in osteoarthritis. We aimed to confirm those results by analyzing ß-catenin levels and explored potential genetic and epigenetic mechanisms involved. ß-Catenin gene expression and nuclear levels were analyzed by real time PCR and confocal immunofluorescence. Increased nuclear ß-catenin was found in osteoblasts isolated from patients with osteoarthritis (99 ± 4 units vs. 76 ± 12, p=0.01, n=10), without differences in gene transcription, which is consistent with a post-translational down-regulation of ß-catenin and decreased Wnt pathway activity. Twenty four single nucleotide polymorphisms (SNPs) of genes showing differential expression between fractures and osteoarthritis (WNT4, WNT10A, WNT16 and SFRP1) were analyzed in DNA isolated from blood of 853 patients. The genotypic frequencies were similar in both groups of patients, with no significant differences. Methylation of Wnt pathway genes was analyzed in bone tissue samples (15 with fractures and 15 with osteoarthritis) by interrogating a CpG-based methylation array. Six genes showed significant methylation differences between both groups of patients: FZD10, TBL1X, CSNK1E, WNT8A, CSNK1A1L and SFRP4. The DNA demethylating agent 5-deoxycytidine up-regulated 8 genes, including FZD10, in an osteoblast-like cell line, whereas it down-regulated other 16 genes. In conclusion, Wnt activity is reduced in patients with hip fractures, in comparison with those with osteoarthritis. It does not appear to be related to differences in the allele frequencies of the Wnt genes studied. On the other hand, methylation differences between both groups could contribute to explain the differences in Wnt activity.


Assuntos
Epigênese Genética , Fraturas do Quadril/genética , Osteoartrite do Quadril/genética , Osteoporose/genética , Via de Sinalização Wnt/genética , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Metilação de DNA , Feminino , Expressão Gênica , Frequência do Gene , Fraturas do Quadril/etiologia , Fraturas do Quadril/metabolismo , Humanos , Masculino , Osteoartrite do Quadril/metabolismo , Osteoporose/complicações , Osteoporose/metabolismo , Polimorfismo de Nucleotídeo Único , beta Catenina/genética , beta Catenina/metabolismo
12.
Neurobiol Dis ; 46(1): 118-29, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22249111

RESUMO

Nuclear speckles are essential nuclear compartments involved in the assembly, delivery and recycling of pre-mRNA processing factors, and in the post-transcriptional processing of pre-mRNAs. Oculopharyngeal muscular dystrophy (OPMD) is caused by a small expansion of the polyalanine tract in the poly(A)-binding protein nuclear 1 (PABPN1). Aggregation of expanded PABPN1 into intranuclear inclusions (INIs) in skeletal muscle fibers is the pathological hallmark of OPMD. In this study what we have analyzed in muscle fibers of OPMD patients and in primary cultures of human myoblasts are the relationships between nuclear speckles and INIs, and the contribution of the former to the biogenesis of the latter. While nuclear speckles concentrate snRNP splicing factors and PABPN1 in control muscle fibers, they are depleted of PABPN1 and appear closely associated with INIs in muscle fibers of OPMD patients. The induction of INI formation in human myoblasts expressing either wild type GFP-PABPN1 or expanded GFP-PABPN1-17ala demonstrates that the initial aggregation of PABPN1 proteins and their subsequent growth in INIs occurs at the edges of the nuclear speckles. Moreover, the growing of INIs gradually depletes PABPN1 proteins and poly(A) RNA from nuclear speckles, although the existence of these nuclear compartments is preserved. Time-lapse experiments in cultured myoblasts confirm nuclear speckles as biogenesis sites of PABPN1 inclusions. Given the functional importance of nuclear speckles in the post-transcriptional processing of pre-mRNAs, the INI-dependent molecular reorganization of these nuclear compartments in muscle fibers may cause a severe dysfunction in nuclear trafficking and processing of polyadenylated mRNAs, thereby contributing to the molecular pathophysiology of OPMD. Our results emphasize the potential importance of nuclear speckles as nuclear targets of neuromuscular disorders.


Assuntos
Fibras Musculares Esqueléticas/patologia , Fibras Musculares Esqueléticas/fisiologia , Distrofia Muscular Oculofaríngea/patologia , Distrofia Muscular Oculofaríngea/fisiopatologia , Proteína I de Ligação a Poli(A)/fisiologia , Idoso de 80 Anos ou mais , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Oculofaríngea/genética , Proteína I de Ligação a Poli(A)/genética
13.
Acta Neuropathol ; 122(4): 481-93, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21915754

RESUMO

Neurons are very sensitive to DNA damage induced by endogenous and exogenous genotoxic agents, as defective DNA repair can lead to neurodevelopmental disorders, brain tumors and neurodegenerative diseases with severe clinical manifestations. Understanding the impact of DNA damage/repair mechanisms on the nuclear organization, particularly on the regulation of transcription and cell cycle, is essential to know the pathophysiology of defective DNA repair syndromes. In this work, we study the nuclear architecture and spatiotemporal organization of chromatin compartments involved in the DNA damage response (DDR) in rat sensory ganglion neurons exposed to X-ray irradiation (IR). We demonstrate that the neuronal DDR involves the formation of two categories of DNA-damage processing chromatin compartments: transient, disappearing within the 1 day post-IR, and persistent, where unrepaired DNA is accumulated. Both compartments concentrate components of the DDR pathway, including γH2AX, pATM and 53BP1. Furthermore, DNA damage does not induce neuronal apoptosis but triggers the G0-G1 cell cycle phase transition, which is mediated by the activation of the ATM-p53 pathway and increased protein levels of p21 and cyclin D1. Moreover, the run on transcription assay reveals a severe inhibition of transcription at 0.5 h post-IR, followed by its rapid recovery over the 1 day post-IR in parallel with the progression of DNA repair. Therefore, the response of healthy neurons to DNA damage involves a transcription- and cell cycle-dependent but apoptosis-independent process. Furthermore, we propose that the segregation of unrepaired DNA in a few persistent chromatin compartments preserves genomic stability of undamaged DNA and the global transcription rate in neurons.


Assuntos
Compartimento Celular/efeitos da radiação , Ciclo Celular/efeitos da radiação , Núcleo Celular/efeitos da radiação , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Gânglios Sensitivos/efeitos da radiação , Células Receptoras Sensoriais/efeitos da radiação , Animais , Compartimento Celular/genética , Ciclo Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Dano ao DNA/fisiologia , Reparo do DNA/genética , Gânglios Sensitivos/citologia , Gânglios Sensitivos/metabolismo , Masculino , Lesões por Radiação/genética , Lesões por Radiação/metabolismo , Lesões por Radiação/patologia , Radiação Ionizante , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo
14.
J Biol Chem ; 286(32): 28287-302, 2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21700704

RESUMO

DNA repair protects neurons against spontaneous or disease-associated DNA damage. Dysfunctions of this mechanism underlie a growing list of neurodegenerative disorders. The Purkinje cell (PC) degeneration mutation causes the loss of nna1 expression and is associated with the postnatal degeneration of PCs. This PC degeneration dramatically affects nuclear architecture and provides an excellent model to elucidate the nuclear mechanisms involved in a whole array of neurodegenerative disorders. We used immunocytochemistry for histone variants and components of the DNA damage response, an in situ transcription assay, and in situ hybridization for telomeres to analyze changes in chromatin architecture and function. We demonstrate that the phosphorylation of H2AX, a DNA damage signal, and the trimethylation of the histone H4K20, a repressive mark, in extensive domains of genome are epigenetic hallmarks of chromatin in degenerating PCs. These histone modifications are associated with a large scale reorganization of chromatin, telomere clustering, and heterochromatin-induced gene silencing, all of them key factors in PC degeneration. Furthermore, ataxia telangiectasia mutated and 53BP1, two components of the DNA repair pathway, fail to be concentrated in the damaged chromatin compartments, even though the expression levels of their coding genes were slightly up-regulated. Although the mechanism by which Nna1 loss of function leads to PC neurodegeneration is undefined, the progressive accumulation of DNA damage in chromosome territories irreversibly compromises global gene transcription and seems to trigger PC degeneration and death.


Assuntos
Montagem e Desmontagem da Cromatina , Reparo do DNA , Inativação Gênica , Doenças Neurodegenerativas/metabolismo , Células de Purkinje/metabolismo , Transdução de Sinais , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Histonas/genética , Histonas/metabolismo , Masculino , Camundongos , Camundongos Mutantes , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Células de Purkinje/patologia , D-Ala-D-Ala Carboxipeptidase Tipo Serina/genética , D-Ala-D-Ala Carboxipeptidase Tipo Serina/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
15.
Biochim Biophys Acta ; 1813(5): 1050-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21070824

RESUMO

DREAM is a Ca(2+)-binding protein with specific functions in different cell compartments. In the nucleus, DREAM acts as a transcriptional repressor, although the mechanism that controls its nuclear localization is unknown. Yeast two-hybrid assay revealed the interaction between DREAM and the SUMO-conjugating enzyme Ubc9 and bioinformatic analysis identified four sumoylation-susceptible sites in the DREAM sequence. Single K-to-R mutations at positions K26 and K90 prevented in vitro sumoylation of recombinant DREAM. DREAM sumoylation mutants retained the ability to bind to the DRE sequence but showed reduced nuclear localization and failed to regulate DRE-dependent transcription. In PC12 cells, sumoylated DREAM is present exclusively in the nucleus and neuronal differentiation induced nuclear accumulation of sumoylated DREAM. In fully differentiated trigeminal neurons, DREAM and SUMO-1 colocalized in nuclear domains associated with transcription. Our results show that sumoylation regulates the nuclear localization of DREAM in differentiated neurons. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.


Assuntos
Núcleo Celular/metabolismo , Proteínas Interatuantes com Canais de Kv/metabolismo , Proteínas Repressoras/metabolismo , Sumoilação , Sequência de Aminoácidos , Animais , Diferenciação Celular , Análise Mutacional de DNA , Células HEK293 , Células HeLa , Humanos , Proteínas Interatuantes com Canais de Kv/química , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células PC12 , Ligação Proteica , Transporte Proteico , Ratos , Proteínas Repressoras/química , Proteína SUMO-1/metabolismo , Alinhamento de Sequência , Nervo Trigêmeo/metabolismo , Nervo Trigêmeo/ultraestrutura , Enzimas de Conjugação de Ubiquitina/metabolismo
16.
Brain Pathol ; 21(4): 374-88, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21054627

RESUMO

The Purkinje cell (PC) degeneration (pcd) phenotype results from mutation in nna1 gene and is associated with the degeneration and death of PCs during the postnatal life. Although the pcd mutation is a model of the ataxic mouse, it shares clinical and pathological characteristics of inherited human spinocerebellar ataxias. PC degeneration in pcd mice provides a useful neuronal system to study nuclear mechanisms involved in DNA damage-dependent neurodegeneration, particularly the contribution of nucleoli and Cajal bodies (CBs). Both nuclear structures are engaged in housekeeping functions for neuronal survival, the biogenesis of ribosomes and the maturation of snRNPs and snoRNPs required for pre-mRNA and pre-rRNA processing, respectively. In this study, we use ultrastructural analysis, in situ transcription assay and molecular markers for DNA damage, nucleoli and CB components to demonstrate that PC degeneration involves the progressive accumulation of nuclear DNA damage associated with disruption of nucleoli and CBs, disassembly of polyribosomes into monoribosomes, ribophagy and shut down of nucleolar and extranucleolar transcription. Microarray analysis reveals that four genes encoding repressors of nucleolar rRNA synthesis (p53, Rb, PTEN and SNF2) are upregulated in the cerebellum of pcd mice. Collectively, these data support that nucleolar and CB alterations are hallmarks of DNA damage-induced neurodegeneration.


Assuntos
Nucléolo Celular/patologia , Corpos Enovelados/patologia , Dano ao DNA , Degeneração Neural/patologia , Células de Purkinje/patologia , Animais , Nucléolo Celular/genética , Nucléolo Celular/metabolismo , Corpos Enovelados/genética , Corpos Enovelados/metabolismo , Feminino , Imunofluorescência , Immunoblotting , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Mutantes , Microscopia Eletrônica de Transmissão , Degeneração Neural/genética , Degeneração Neural/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Células de Purkinje/metabolismo
17.
Neurotox Res ; 17(2): 167-78, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19609631

RESUMO

The ubiquitin-dependent proteasome system (UPS) is the major pathway responsible for selective nuclear and cytoplasmic protein degradation. Bortezomib, a boronic acid dipeptide, is a reversible 20S proteasome inhibitor used as novel anticancer drug, particularly in the treatment of multiple myeloma and certain lymphomas. Bortezomib-induced peripheral neuropathy (BIPN) is a widely recognized dose-limiting neurotoxicity of this proteasome inhibitor, which causes a significant negative impact on the quality of life. The pathogenic mechanisms underlying bortezomib neurotoxicity are little known. In this study a rat was used as our animal model to investigate the bortezomib-induced nuclear changes in dorsal root ganglia (DRG) neurons. Our results indicate that this neuronal population is an important target of bortezomib neurotoxicity. Nuclear changes include accumulation of ubiquitin-protein conjugates, reduction of transcriptional activity, and nuclear retention of poly(A) RNAs in numerous spherical or ring-shaped dense granules. They also contained the RNA-binding proteins PABPN1 (poly(A) binding protein nuclear 1) and Sam68, but lacked the mRNA nuclear export factors REF and Y14. At the cytoplasmic level, most neurons exhibited chromatolysis, supporting the inhibition of mRNA translation. Our results indicate that bortezomib interferes with transcription, nuclear processing and transport, and cytoplasmic translation of mRNAs in DRG neurons. They also support that this neuronal dysfunction is an essential pathogenic mechanism in the BIPN, which is characterized by sensory impairment including sensory ataxia.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ácidos Borônicos/farmacologia , Proteína II de Ligação a Poli(A)/metabolismo , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , RNA Mensageiro/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Bortezomib , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Gânglios Espinais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Microscopia Eletrônica de Transmissão/métodos , Proteína II de Ligação a Poli(A)/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/ultraestrutura , Fatores de Tempo , Ubiquitina/metabolismo , Uridina/análogos & derivados , Uridina/metabolismo
18.
J Struct Biol ; 167(3): 235-41, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19539030

RESUMO

TDP-43 is a RNA/DNA-binding protein structurally related to nuclear hnRNP proteins. Previous biochemical studies have shown that this nuclear protein plays a role in the regulation of gene transcription, alternative splicing and mRNA stability. Despite the ubiquitous distribution of TDP-43, the growing list of TDP-43 proteinopathies is primarily associated with neurodegenerative disorders. Particularly, TDP-43 redistributes to the cytoplasm and forms pathological inclusions in amyotrophic lateral sclerosis and several forms of sporadic and familiar frontotemporal lobar degeneration. Here, we have studied the nuclear compartmentalization of TDP-43 in normal rat neurons by using light and electron microscopy immunocytochemistry with molecular markers for nuclear compartments, a transcription assay with 5'-fluorouridine, and in situ hybridization for telomeric DNA. TDP-43 is concentrated in euchromatin domains, specifically in perichromatin fibrils, nuclear sites of transcription and cotranscriptional splicing. In these structures, TDP-43 colocalizes with 5'-fluorouridine incorporation sites into nascent pre-mRNA. TDP-43 is absent in transcriptionally silent centromeric and telomeric heterochromatin, as well as in the Cajal body, a transcription free nuclear compartment. Furthermore, a weak TDP-43 immunolabeling is found in nuclear speckles of splicing factors. The specific localization of TDP-43 in active sites of transcription and cotranscriptional splicing is consistent with biochemical data indicating a role of TDP-43 in the regulation of transcription and alternative splicing.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Transcrição Gênica , Animais , Núcleo Celular/metabolismo , Cromatina/metabolismo , Imuno-Histoquímica , Masculino , Processamento Pós-Transcricional do RNA , Ratos , Ratos Sprague-Dawley , Telômero/metabolismo , Uridina/análogos & derivados , Uridina/química
19.
Chromosoma ; 118(4): 437-43, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19404660

RESUMO

In 1906, the Spanish neurobiologist Santiago Ramón y Cajal was awarded the Nobel Prize in Physiology or Medicine in recognition of his work on the structure of neurons and their connections. Cajal is commonly regarded as the father of modern neuroscience. What is less well known is that Cajal also had a great interest in intracellular neuronal structures and developed the reduced silver nitrate method for the study of neurofibrils (neurofilaments) and nuclear subcompartments. It was in 1903 that Cajal discovered the "accessory body" ("Cajal body") and seven years later, published an article on the organization of the cell nucleus in mammalian neurons that represents a masterpiece of nuclear structure at the light microscopy level. In addition to the accessory body, it includes the analysis of several nuclear components currently recognized as fibrillar centers of the nucleolus, nuclear speckles of splicing factors, transcription foci, nuclear matrix, and the double nuclear membrane. The aim of this article is to revisit Cajal's contributions to the knowledge of the neuronal nucleus in light of our current understanding of nuclear structure and function.


Assuntos
Núcleo Celular/metabolismo , Corpos Enovelados/metabolismo , Neurônios/metabolismo , Neurociências/história , História do Século XIX , História do Século XX , Neurônios/citologia , Prêmio Nobel , Espanha
20.
J Struct Biol ; 159(3): 451-61, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17587596

RESUMO

This paper studies the molecular organization, neuronal distribution and cellular differentiation dynamics of the giant fibrillar centers (GFCs) of nucleoli in rat sensory ganglia neurons. The GFC appeared as a round nucleolar domain (1-2 microm in diameter) partially surrounded by the dense fibrillar component and accompanied by numerous small FCs. By immunocytochemistry, the GFC concentrated the upstream binding factor, which may serve as a marker of this structure, and also contain RNA polymerase I, DNA topoisomerase I, SUMO-1 and Ubc9. However, they lack ubiquitin-proteasome conjugates and 20S proteasome. Transcription assay with 5'-fluorouridine incorporation revealed the presence of nascent RNA on the dense fibrillar component of the neuronal nucleolus, but not within the low electron-density area of the GFC. The formation of GFCs is neuronal size dependent: they were found in 58%, 30% and 0% of the large, medium and small neurons, respectively. GFCs first appeared during the postnatal period, concomitantly with a stage of neuronal growth, myelination and bioelectrical maturation. GFCs were not observed in segregated nucleoli induced by severe inhibition of RNA synthesis. We suggest that the formation of GFCs is associated with a high rate of ribosome biogenesis of the transcriptionally more active large-size neurons.


Assuntos
Nucléolo Celular/ultraestrutura , Gânglios Sensitivos/crescimento & desenvolvimento , Gânglios Sensitivos/ultraestrutura , Neurônios Aferentes/ultraestrutura , Proteínas Pol1 do Complexo de Iniciação de Transcrição/análise , Animais , Diferenciação Celular , Nucléolo Celular/química , Gânglios Sensitivos/metabolismo , Masculino , Microscopia Imunoeletrônica , Neurônios Aferentes/metabolismo , Complexo de Endopeptidases do Proteassoma/análise , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Ribossômico/análise , RNA Ribossômico/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína SUMO-1/análise , Proteína SUMO-1/metabolismo , Transcrição Gênica , Ubiquitina/análise , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...