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











Base de dados
Intervalo de ano de publicação
1.
Mol Cell ; 83(20): 3596-3607, 2023 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-37716351

RESUMO

Mitotic DNA synthesis (MiDAS) is an unusual form of DNA replication that occurs during mitosis. Initially, MiDAS was characterized as a process associated with intrinsically unstable loci known as common fragile sites that occurs after cells experience DNA replication stress (RS). However, it is now believed to be a more widespread "salvage" mechanism that is called upon to complete the duplication of any under-replicated genomic region. Emerging data suggest that MiDAS is a DNA repair process potentially involving two or more pathways working in parallel or sequentially. In this review, we introduce the causes of RS, regions of the human genome known to be especially vulnerable to RS, and the strategies used to complete DNA replication outside of S phase. Additionally, because MiDAS is a prominent feature of aneuploid cancer cells, we will discuss how targeting MiDAS might potentially lead to improvements in cancer therapy.


Assuntos
Reparo do DNA , Replicação do DNA , Humanos , Fase S/genética , Mitose/genética , Replicação Viral
2.
Nat Commun ; 14(1): 706, 2023 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-36759509

RESUMO

Oncogene activation creates DNA replication stress (RS) in cancer cells, which can generate under-replicated DNA regions (UDRs) that persist until cells enter mitosis. UDRs also have the potential to generate DNA bridges in anaphase cells or micronuclei in the daughter cells, which could promote genomic instability. To suppress such damaging changes to the genome, human cells have developed a strategy to conduct 'unscheduled' DNA synthesis in mitosis (termed MiDAS) that serves to rescue under-replicated loci. Previous studies have shown that MiDAS proceeds via a POLD3-dependent pathway that shows some features of break-induced replication. Here, we define how human cells utilize both DNA gap filling (REV1 and Pol ζ) and replicative (Pol δ) DNA polymerases to complete genome duplication following a perturbed S-phase. We present evidence for the existence of a polymerase-switch during MiDAS that is required for new DNA synthesis at UDRs. Moreover, we reveal that, upon oncogene activation, cancer cell survival is significantly compromised when REV1 is depleted, suggesting that REV1 inhibition might be a feasible approach for the treatment of some human cancers.


Assuntos
Replicação do DNA , DNA Polimerase Dirigida por DNA , Humanos , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Reparo do DNA , DNA/genética , DNA/metabolismo , Dano ao DNA
3.
Mol Cell ; 82(18): 3366-3381.e9, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36002000

RESUMO

Oncogene activation during tumorigenesis promotes DNA replication stress (RS), which subsequently drives the formation of cancer-associated chromosomal rearrangements. Many episodes of physiological RS likely arise due to conflicts between the DNA replication and transcription machineries operating simultaneously at the same loci. One role of the RAD51 recombinase in human cells is to protect replication forks undergoing RS. Here, we have identified a key role for RAD51 in preventing transcription-replication conflicts (TRCs) from triggering replication fork breakage. The genomic regions most affected by RAD51 deficiency are characterized by being replicated and transcribed in early S-phase and show significant overlap with loci prone to cancer-associated amplification. Consistent with a role for RAD51 in protecting against transcription-replication conflicts, many of the adverse effects of RAD51 depletion are ameliorated by inhibiting early S-phase transcription. We propose a model whereby RAD51 suppresses fork breakage and subsequent inadvertent amplification of genomic loci prone to experiencing TRCs.


Assuntos
Replicação do DNA , Rad51 Recombinase , Cromossomos/metabolismo , Humanos , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Fase S/genética , Transcrição Gênica
4.
Int J Mol Sci ; 23(9)2022 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-35563522

RESUMO

Chromosomal instability (CIN) can be a driver of tumorigenesis but is also a promising therapeutic target for cancer associated with poor prognosis such as triple negative breast cancer (TNBC). The treatment of TNBC cells with defects in DNA repair genes with poly(ADP-ribose) polymerase inhibitor (PARPi) massively increases CIN, resulting in apoptosis. Here, we identified a previously unknown role of microRNA-449a in CIN. The transfection of TNBC cell lines HCC38, HCC1937 and HCC1395 with microRNA-449a mimics led to induced apoptosis, reduced cell proliferation, and reduced expression of genes in homology directed repair (HDR) in microarray analyses. EME1 was identified as a new target gene by immunoprecipitation and luciferase assays. The reduced expression of EME1 led to an increased frequency of ultrafine bridges, 53BP1 foci, and micronuclei. The induced expression of microRNA-449a elevated CIN beyond tolerable levels and induced apoptosis in TNBC cell lines by two different mechanisms: (I) promoting chromatid mis-segregation by targeting endonuclease EME1 and (II) inhibiting HDR by downregulating key players of the HDR network such as E2F3, BIRC5, BRCA2 and RAD51. The ectopic expression of microRNA-449a enhanced the toxic effect of PARPi in cells with pathogenic germline BRCA1 variants. The newly identified role makes microRNA-449a an interesting therapeutic target for TNBC.


Assuntos
Antineoplásicos , MicroRNAs , Neoplasias de Mama Triplo Negativas , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cromátides/metabolismo , Reparo do DNA/genética , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Neoplasias de Mama Triplo Negativas/patologia
6.
Cell Res ; 30(11): 997-1008, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32561860

RESUMO

DNA replication stress, a feature of human cancers, often leads to instability at specific genomic loci, such as the common fragile sites (CFSs). Cells experiencing DNA replication stress may also exhibit mitotic DNA synthesis (MiDAS). To understand the physiological function of MiDAS and its relationship to CFSs, we mapped, at high resolution, the genomic sites of MiDAS in cells treated with the DNA polymerase inhibitor aphidicolin. Sites of MiDAS were evident as well-defined peaks that were largely conserved between cell lines and encompassed all known CFSs. The MiDAS peaks mapped within large, transcribed, origin-poor genomic regions. In cells that had been treated with aphidicolin, these regions remained unreplicated even in late S phase; MiDAS then served to complete their replication after the cells entered mitosis. Interestingly, leading and lagging strand synthesis were uncoupled in MiDAS, consistent with MiDAS being a form of break-induced replication, a repair mechanism for collapsed DNA replication forks. Our results provide a better understanding of the mechanisms leading to genomic instability at CFSs and in cancer cells.


Assuntos
Sítios Frágeis do Cromossomo/genética , DNA/biossíntese , Genoma Humano , Mitose/genética , Análise de Sequência de DNA , Linhagem Celular Tumoral , Quebra Cromossômica , Período de Replicação do DNA/genética , Instabilidade Genômica , Humanos , Anotação de Sequência Molecular , Neoplasias/genética , Origem de Replicação/genética
7.
Nat Struct Mol Biol ; 27(5): 424-437, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32398827

RESUMO

Oncogene activation during tumorigenesis generates DNA replication stress, a known driver of genome rearrangements. In response to replication stress, certain loci, such as common fragile sites and telomeres, remain under-replicated during interphase and subsequently complete locus duplication in mitosis in a process known as 'MiDAS'. Here, we demonstrate that RTEL1 (regulator of telomere elongation helicase 1) has a genome-wide role in MiDAS at loci prone to form G-quadruplex-associated R-loops, in a process that is dependent on its helicase function. We reveal that SLX4 is required for the timely recruitment of RTEL1 to the affected loci, which in turn facilitates recruitment of other proteins required for MiDAS, including RAD52 and POLD3. Our findings demonstrate that RTEL1 is required for MiDAS and suggest that RTEL1 maintains genome stability by resolving conflicts that can arise between the replication and transcription machineries.


Assuntos
DNA Helicases/genética , DNA Helicases/metabolismo , Quadruplex G , Genoma Humano/genética , Mitose , Animais , Linhagem Celular , DNA Helicases/química , DNA Polimerase III/genética , DNA Polimerase III/metabolismo , Instabilidade Genômica , Humanos , Imunoprecipitação , Camundongos , Enzimas Multifuncionais/genética , Enzimas Multifuncionais/metabolismo , Conformação de Ácido Nucleico , RNA Helicases/genética , RNA Helicases/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Recombinases/genética , Recombinases/metabolismo , Ribonuclease H/genética , Ribonuclease H/metabolismo
8.
Curr Biol ; 29(7): 1232-1242.e5, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30905608

RESUMO

Abscission is the final step of cell division when the cytokinetic furrow ingresses completely, leading to midbody formation and plasma membrane fission [1]. In human cells, the Aurora-B-driven abscission checkpoint delays cytokinesis until any residual chromatin spanning the midbody is removed [2-5]. If this does not occur efficiently, uneven segregation of daughter genomes can occur. The mechanism by which the abscission checkpoint becomes satisfied to permit cytokinesis is poorly defined. Here, we identify RIF1 and its binding partner, protein phosphatase 1 (PP1), as being critical for regulation of abscission timing in human cells. We show that RIF1 promotes cytokinesis through recruitment of PP1 to the midbody, which then counteracts Aurora B kinase activity, leading to dephosphorylation of a regulator of abscission timing, CHMP4C [6-10]. Although RIF1 binds to unresolved DNA bridges that persist into telophase [11], we show that this cytokinetic function of the RIF1-PP1 axis is not limited to instances where cell division is perturbed by the presence of bridges. Nevertheless, we show that altering the balance of the opposing Aurora B kinase and PP1 phosphatase activities makes cells unresponsive to DNA bridges and sensitizes cells to agents that induce bridge formation. Our data define a new mechanism for regulation of abscission timing and emphasize how antagonism between kinases and phosphatases is a widespread mechanism for determining the timing of mitotic transactions. Because cancer cells experiencing oncogene-induced replication stress generate excessive mitotic DNA bridging [12], targeting this new regulatory pathway could be a promising therapeutic strategy.


Assuntos
Mitose/fisiologia , Receptores de Neuropeptídeo Y/genética , Proteínas de Ligação a Telômeros/genética , Citocinese/fisiologia , Humanos , Mitose/genética , Receptores de Neuropeptídeo Y/metabolismo , Proteínas de Ligação a Telômeros/metabolismo
9.
Int J Biol Macromol ; 123: 228-238, 2019 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-30412755

RESUMO

A highly competent material, based on poly lactic acid (PLLA) grafted hydroxypropyl guar gum (HPG-g-PLLA) and polypyrrole/carboxylated multiwalled carbon nanotube (PPy/C-MWCNT) composite of various binary composition and copolymer of one of these nanocomposites have been synthesized successfully by in-situ polymerization. The environmentally affable nanocomposites have been characterized by spectroscopy, microscopy and thermogravimetry. Cytotoxicity of bio-nanocomposite has been inquired by cell viability study, which reveals its eco-friendly nature. The electrochemical properties of the biomaterials have been appraised by cyclic voltammetric studies. The PPy/C-MWCNT composite having 1 wt% C-MWCNT appears as the optimum composition from electrochemical studies. The hydrogel nanocomposite (HPG-g-PLLA5/0.5) copolymer behaves as a super ordinate material than pure PPy and PPy/C-MWCNT in every aspect of electrochemical properties like current density, stability, processibility and reversibility. Moreover the hydrogel nanocomposite, making electrode fabrication more simple and binder-free, nullifies all the interfacial complications arising from binders as well.


Assuntos
Biopolímeros/química , Nanotubos de Carbono/química , Poliésteres/síntese química , Polissacarídeos/síntese química , Galactanos/síntese química , Galactanos/química , Hidrogel de Polietilenoglicol-Dimetacrilato/síntese química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Mananas/síntese química , Mananas/química , Nanocompostos/química , Gomas Vegetais/síntese química , Gomas Vegetais/química , Poliésteres/química , Polimerização , Polímeros/química , Polissacarídeos/química , Pirróis/química
10.
J Cancer Res Ther ; 14(Supplement): S818-S819, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30249914

RESUMO

Thyroid abnormalities are found nearly 70% cases receiving sunitinib therapy. Mostly, patients suffer transient hypothyroidism rarely presents with overt acute symptoms requiring levothyroxine replacement. Onset is variable in published literature. We report a case of metastatic gastrointestinal stromal tumor receiving sunitinib with normal baseline thyroid function. The patient developed symptoms of acute severe hypothyroidism with high thyroid stimulating hormone level on the 4th week of therapy. The patient responded with oral levothyroxine. clinical and biochemical parameter resolved rapidly. Patient receiving sunitinib warns baseline and subsequent surveillance of thyroid function (both clinical and biochemical). This rare dreadful condition reverts promptly with thyroxine replacement.


Assuntos
Tumores do Estroma Gastrointestinal/tratamento farmacológico , Hipotireoidismo/patologia , Indóis/efeitos adversos , Pirróis/efeitos adversos , Idoso , Tumores do Estroma Gastrointestinal/sangue , Tumores do Estroma Gastrointestinal/patologia , Humanos , Hipotireoidismo/sangue , Hipotireoidismo/induzido quimicamente , Masculino , Sunitinibe , Tireotropina/sangue
11.
Oncotarget ; 9(22): 15836-15846, 2018 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-29662610

RESUMO

Telomeres resemble common fragile sites (CFSs) in that they are difficult-to-replicate and exhibit fragility in mitosis in response to DNA replication stress. At CFSs, this fragility is associated with a delay in the completion of DNA replication until early mitosis, whereupon cells are proposed to switch to a RAD52-dependent form of break-induced replication. Here, we show that this mitotic DNA synthesis (MiDAS) is also a feature of human telomeres. Telomeric MiDAS is not restricted to those telomeres displaying overt fragility, and is a feature of a wide range of cell lines irrespective of whether their telomeres are maintained by telomerase or by the alternative lengthening of telomeres (ALT) mechanism. MiDAS at telomeres requires RAD52, and is mechanistically similar to CFS-associated MiDAS, with the notable exception that telomeric MiDAS does not require the MUS81-EME1 endonuclease. We propose a model whereby replication stress initiates a RAD52-dependent form of break-induced replication that bypasses a requirement for MUS81-EME1 to complete DNA synthesis in mitosis.

12.
Methods Enzymol ; 601: 45-58, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29523241

RESUMO

Our conventional understanding of the process of DNA replication is that it occurs in the S-phase of the cell division cycle. However, during investigations into the mechanism by which common fragile sites (CFSs) drive genome instability, we observed that some DNA synthesis was still occurring in early mitosis at these loci. This curious phenomenon of mitotic DNA synthesis (which we now term "MiDAS") appears to be a form of break-induced DNA replication (BIR), a DNA repair process based on homologous recombination that has been characterized in detail only in lower eukaryotes. During MiDAS, it is proposed that parts of the human genome that are not fully replicated when cells enter mitotic prophase complete their replicative cycle at that point. To date, the loci that most depend upon this process are those whose replication can be affected by oncogene-induced DNA replication stress (RS), most notably, CFSs. From our studies, it is clear that the successful completion of MiDAS at CFSs can minimize chromosome missegregation and nondisjunction. Nevertheless, it is still not clear which loci that can undergo MiDAS, whether MiDAS is associated with mutations or genome rearrangements, or whether MiDAS really is a form of BIR. In this review, we describe methods for detecting MiDAS both in prometaphase cells and directly on isolated metaphase chromosomes. In addition, we have included methods for combining MiDAS detection either with immunofluorescence (IF) detection of proteins that are recruited to the MiDAS loci, or with fluorescence in situ hybridization using probes that target specific genomic loci.


Assuntos
Técnicas Genéticas , Mitose , Reparo de DNA por Recombinação , Linhagem Celular , DNA/metabolismo , Humanos , Metáfase , Prometáfase
13.
F1000Res ; 6: 666, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28620461

RESUMO

An unusual feature of many eukaryotic genomes is the presence of regions that appear intrinsically difficult to copy during the process of DNA replication. Curiously, the location of these difficult-to-replicate regions is often conserved between species, implying a valuable role in some aspect of genome organization or maintenance. The most prominent class of these regions in mammalian cells is defined as chromosome fragile sites, which acquired their name because of a propensity to form visible gaps/breaks on otherwise-condensed chromosomes in mitosis. This fragility is particularly apparent following perturbation of DNA replication-a phenomenon often referred to as "replication stress". Here, we review recent data on the molecular basis for chromosome fragility and the role of fragile sites in the etiology of cancer. In particular, we highlight how studies on fragile sites have provided unexpected insights into how the DNA repair machinery assists in the completion of DNA replication.

14.
Genes Dev ; 31(8): 816-829, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28487407

RESUMO

DNA replication fork progression can be disrupted at difficult to replicate loci in the human genome, which has the potential to challenge chromosome integrity. This replication fork disruption can lead to the dissociation of the replisome and the formation of DNA damage. To model the events stemming from replisome dissociation during DNA replication perturbation, we used a degron-based system for inducible proteolysis of a subunit of the replicative helicase. We show that MCM2-depleted cells activate a DNA damage response pathway and generate replication-associated DNA double-strand breaks (DSBs). Remarkably, these cells maintain some DNA synthesis in the absence of MCM2, and this requires the MCM8-9 complex, a paralog of the MCM2-7 replicative helicase. We show that MCM8-9 functions in a homologous recombination-based pathway downstream from RAD51, which is promoted by DSB induction. This RAD51/MCM8-9 axis is distinct from the recently described RAD52-dependent DNA synthesis pathway that operates in early mitosis at common fragile sites. We propose that stalled replication forks can be restarted in S phase via homologous recombination using MCM8-9 as an alternative replicative helicase.


Assuntos
Replicação do DNA/genética , DNA/biossíntese , Proteínas de Manutenção de Minicromossomo/metabolismo , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Ativação Enzimática/genética , Células HCT116 , Recombinação Homóloga/genética , Humanos , Componente 2 do Complexo de Manutenção de Minicromossomo/genética , Componente 2 do Complexo de Manutenção de Minicromossomo/metabolismo , Proteínas de Manutenção de Minicromossomo/genética , Mutação , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Fase S/genética
15.
Mol Cell ; 64(6): 1117-1126, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27984745

RESUMO

Homologous recombination (HR) is necessary to counteract DNA replication stress. Common fragile site (CFS) loci are particularly sensitive to replication stress and undergo pathological rearrangements in tumors. At these loci, replication stress frequently activates DNA repair synthesis in mitosis. This mitotic DNA synthesis, termed MiDAS, requires the MUS81-EME1 endonuclease and a non-catalytic subunit of the Pol-delta complex, POLD3. Here, we examine the contribution of HR factors in promoting MiDAS in human cells. We report that RAD51 and BRCA2 are dispensable for MiDAS but are required to counteract replication stress at CFS loci during S-phase. In contrast, MiDAS is RAD52 dependent, and RAD52 is required for the timely recruitment of MUS81 and POLD3 to CFSs in early mitosis. Our results provide further mechanistic insight into MiDAS and define a specific function for human RAD52. Furthermore, selective inhibition of MiDAS may comprise a potential therapeutic strategy to sensitize cancer cells undergoing replicative stress.


Assuntos
DNA Polimerase III/genética , Replicação do DNA , Proteínas de Ligação a DNA/genética , DNA/genética , Endonucleases/genética , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Reparo de DNA por Recombinação , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Linhagem Celular Tumoral , Sítios Frágeis do Cromossomo , DNA/metabolismo , Dano ao DNA , DNA Polimerase III/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/metabolismo , Endonucleases/metabolismo , Células HeLa , Humanos , Mitose , Osteoblastos/citologia , Osteoblastos/metabolismo , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Estresse Fisiológico
16.
Analyst ; 141(1): 225-35, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26584051

RESUMO

A new type of easily synthesized rhodamine-based chemosensor L(3), with potential NO2 donor atoms, selectively and rapidly recognizes Hg(2+) ions in the presence of all biologically relevant metal ions and toxic heavy metals. A very low detection limit (78 nM) along with cytoplasmic cell imaging applications with no or negligible cytotoxicity indicate good potential for in vitro/in vivo cell imaging studies. SEM and TEM studies reveal strongly agglomerated aggregations in the presence of 5 mM SDS which turn into isolated core shell microstructures in the presence of 9 mM SDS. The presence of SDS causes an enhanced quantum yield (φ) and stability constant (Kf) compared to those in the absence of SDS. Again, the FI of the [L(3)-Hg](2+) complex in an aqueous SDS (9 mM) medium is unprecedentedly enhanced (∼143 fold) compared to that in the absence of SDS. All of these observations clearly manifest in the enhanced rigidity of the [L(3)-Hg](2+) species in the micro-heterogeneous environment significantly restricting its dynamic movements. This phenomenon may be ascribed as an aggregation induced emission enhancement (AIEE). The fluorescence anisotropy assumes a maximum at 5 mM SDS due to strong trapping (sandwiching) of the doubly positively charged [L(3)-Hg](2+) complex between two co-facial laminar microstructures of SDS under pre-miceller conditions where there is a strong electrostatic interaction that causes an improved inhibition to dynamic movement of the probe-mercury complex. On increasing the SDS concentration there is a phase transition in the SDS microstructures and micellization starts to prevail at SDS ≥ 7.0 mM. The doubly positively charged [L(3)-Hg](2+) complex is trapped inside the hydrophobic inner core of the micelle which is apparent from the failure to quench the fluorescence of the complex on adding 10 equivalents of H2EDTA(2-) solution but in the absence of SDS it is quenched effectively.


Assuntos
Técnicas de Química Analítica/instrumentação , Corantes Fluorescentes/química , Rodaminas/química , Tensoativos/química , Polarização de Fluorescência , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Limite de Detecção , Mercúrio/análise , Mercúrio/química , Dodecilsulfato de Sódio/química
17.
Comput Biol Chem ; 51: 51-6, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24929545

RESUMO

Rotavirus, the major cause of infantile nonbacterial diarrhea, was found to be associated with development of diabetes-associated auto-antibodies. In our study we tried to find out further potential autoimmune threats of this virus using bioinformatics approach. We took rotaviral proteins to study similarity with Homo sapiens proteome and found most conserved structural protein VP6 matches at two regions with ryanodine receptor, an autoimmune target associated with myasthenia gravis. Myasthenia gravis, a chronic neurodegenerative autoimmune disorder with no typical known reason, is characterized by fluctuating muscle weakness which is typically enhanced during muscular effort. Affected patients generate auto antibodies against mainly acetyl choline receptor and sarcoplasmic reticulum calcium-release channel protein ryanodine receptor. Further, we observed that two regions which matched with ryanodine receptor remain conserved in all circulating rotaviral strains and showed significant antigenecity with respect to myasthenia gravis associated HLA haplotypes. Overall, our study detected rotaviral VP6 as a potential threat for myasthenia gravis and enlighten an area of virus associated autoimmune research.


Assuntos
Antígenos Virais/química , Proteínas do Capsídeo/química , Epitopos/química , Antígenos HLA/química , Rotavirus/química , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Sequência de Aminoácidos , Antígenos Virais/imunologia , Autoimunidade , Sítios de Ligação , Capsídeo/química , Capsídeo/imunologia , Proteínas do Capsídeo/imunologia , Biologia Computacional , Epitopos/imunologia , Antígenos HLA/imunologia , Haplótipos , Humanos , Modelos Moleculares , Mimetismo Molecular , Dados de Sequência Molecular , Miastenia Gravis/complicações , Miastenia Gravis/imunologia , Miastenia Gravis/virologia , Ligação Proteica , Rotavirus/imunologia , Infecções por Rotavirus/complicações , Infecções por Rotavirus/imunologia , Infecções por Rotavirus/virologia , Canal de Liberação de Cálcio do Receptor de Rianodina/imunologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
18.
Virology ; 454-455: 270-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24725954

RESUMO

Viruses, obligate cellular parasites rely on host cellular functions and target the host cell cycle for their own benefit. In this study, effect of rotavirus infection on cell cycle machinery was explored. We found that rotavirus (RV) infection in MA104 cells induces the expression of cyclins and cyclin dependent kinases and down-regulates expression of CDK inhibitors, resulting in G1 to S phase transition. The rotavirus induced S phase accumulation was found to be concurrent with induction in expression of calmodulin and activation of CaMKI which is reported as inducer of G1-S phase transition. This cell cycle manipulation was found to be Ca(+2)/Calmodulin pathway dependent. The physiological relevance of G1 to S phase transition was established when viral gene expressions as well as viral titers were found to be increased in S phase synchronized cells and decreased in G0/G1 phase synchronized cells compared to unsynchronized cells during rotavirus infection.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Ciclo Celular , Interações Hospedeiro-Patógeno , Rotavirus/fisiologia , Animais , Linhagem Celular , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Células Epiteliais/fisiologia , Células Epiteliais/virologia , Macaca mulatta , Inibidores de Proteínas Quinases/metabolismo
19.
PLoS One ; 9(3): e92126, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24643253

RESUMO

Rotavirus is the single, most important agent of infantile gastroenteritis in many animal species, including humans. In developing countries, rotavirus infection attributes approximately 500,000 deaths annually. Like other viruses it establishes an intimate and complex interaction with the host cell to counteract the antiviral responses elicited by the cell. Among various pattern recognition receptors (PAMPs) of the host, the cytosolic RNA helicases interact with viral RNA to activate the Mitochondrial Antiviral Signaling protein (MAVS), which regulates cellular interferon response. With an aim to identify the role of different PAMPs in rotavirus infected cell, MAVS was found to degrade in a time dependent and strain independent manner. Rotavirus non-structural protein 1 (NSP1) which is a known IFN antagonist, interacted with MAVS and degraded it in a strain independent manner, resulting in a complete loss of RNA sensing machinery in the infected cell. To best of our knowledge, this is the first report on NSP1 functionality where a signaling protein is targeted unanimously in all strains. In addition NSP1 inhibited the formation of detergent resistant MAVS aggregates, thereby averting the antiviral signaling cascade. The present study highlights the multifunctional role of rotavirus NSP1 and reinforces the fact that the virus orchestrates the cellular antiviral response to its own benefit by various back up strategies.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Interações Hospedeiro-Patógeno , Rotavirus/genética , Proteínas não Estruturais Virais/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Células HT29 , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/antagonistas & inibidores , Interferon beta/genética , Interferon beta/metabolismo , Leupeptinas/farmacologia , Luciferases/genética , Luciferases/metabolismo , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Rotavirus/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/metabolismo
20.
J Biol Chem ; 288(20): 14554-14568, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23548901

RESUMO

During infection, viral proteins target cellular pathways that regulate cellular innate immune responses and cell death. We demonstrate that influenza A virus matrix 1 protein (M1), an established proapoptotic protein, activates nuclear factor-κB member RelB-mediated survival genes (cIAP1, cIAP2, and cFLIP), a function that is linked with its nuclear translocation during early infection. Death domain-associated protein 6 (Daxx) is a transcription co-repressor of the RelB-responsive gene promoters. During influenza virus infection M1 binds to and stabilizes Daxx protein by preventing its ubiquitination and proteasomal degradation. Binding of M1 with Daxx through its Daxx binding motif prevents binding of RelB and Daxx, resulting in up-regulation of survival genes. This interaction also prevents promoter recruitment of DNA methyltransferases (Dnmt1 and Dnmt3a) and lowers CpG methylation of the survival gene promoters, leading to the activation of these genes. Thus, M1 prevents repressional function of Daxx during infection, thereby exerting a survival role. In addition to its nuclear localization signal, translocation of M1 to the nucleus depends on cellular kinase-mediated phosphorylation as the protein kinase C inhibitor calphostin C effectively down-regulates virus replication. The study reconciles the ambiguity of dual antagonistic function of viral protein and potentiates a possible target to limit virus infection.


Assuntos
Apoptose , Regulação Viral da Expressão Gênica , Fosforilação/fisiologia , Proteínas da Matriz Viral/metabolismo , Transporte Ativo do Núcleo Celular , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Correpressoras , Ilhas de CpG , Cães , Células HEK293 , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Células Madin Darby de Rim Canino , Modelos Biológicos , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Frações Subcelulares , Fator de Transcrição RelB/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA