Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 52(7): 3794-3809, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38340339

RESUMO

Meiotic recombination is initiated by programmed double-strand breaks (DSBs). Studies in Saccharomyces cerevisiae have shown that, following rapid resection to generate 3' single-stranded DNA (ssDNA) tails, one DSB end engages a homolog partner chromatid and is extended by DNA synthesis, whereas the other end remains associated with its sister. Then, after regulated differentiation into crossover- and noncrossover-fated types, the second DSB end participates in the reaction by strand annealing with the extended first end, along both pathways. This second-end capture is dependent on Rad52, presumably via its known capacity to anneal two ssDNAs. Here, using physical analysis of DNA recombination, we demonstrate that this process is dependent on direct interaction of Rad52 with the ssDNA binding protein, replication protein A (RPA). Furthermore, the absence of this Rad52-RPA joint activity results in a cytologically-prominent RPA spike, which emerges from the homolog axes at sites of crossovers during the pachytene stage of the meiotic prophase. Our findings suggest that this spike represents the DSB end of a broken chromatid caused by either the displaced leading DSB end or the second DSB end, which has been unable to engage with the partner homolog-associated ssDNA. These and other results imply a close correspondence between Rad52-RPA roles in meiotic recombination and mitotic DSB repair.


Assuntos
Troca Genética , Quebras de DNA de Cadeia Dupla , Meiose , Proteína Rad52 de Recombinação e Reparo de DNA , Proteína de Replicação A , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Proteína de Replicação A/metabolismo , Proteína de Replicação A/genética , Meiose/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Recombinação Genética , DNA de Cadeia Simples/metabolismo , DNA de Cadeia Simples/genética , Recombinação Homóloga/genética
2.
Exp Mol Med ; 55(7): 1348-1356, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37394581

RESUMO

Thioredoxin-interacting protein (TXNIP), which is also known as thioredoxin-binding protein 2 (TBP2), directly interacts with the major antioxidant protein thioredoxin (TRX) and inhibits its antioxidant function and expression. However, recent studies have demonstrated that TXNIP is a multifunctional protein with functions beyond increasing intracellular oxidative stress. TXNIP activates endoplasmic reticulum (ER) stress-mediated nucleotide-binding oligomerization domain (NOD)-like receptor protein-3 (NLRP3) inflammasome complex formation, triggers mitochondrial stress-induced apoptosis, and stimulates inflammatory cell death (pyroptosis). These newly discovered functions of TXNIP highlight its role in disease development, especially in response to several cellular stress factors. In this review, we provide an overview of the multiple functions of TXNIP in pathological conditions and summarize its involvement in various diseases, such as diabetes, chronic kidney disease, and neurodegenerative diseases. We also discuss the potential of TXNIP as a therapeutic target and TXNIP inhibitors as novel therapeutic drugs for treating these diseases.


Assuntos
Antioxidantes , Proteína 3 que Contém Domínio de Pirina da Família NLR , Antioxidantes/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Estresse Oxidativo , Tiorredoxinas/genética , Inflamassomos/metabolismo
3.
BMB Rep ; 56(2): 102-107, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36513383

RESUMO

Genome editing using CRISPR-associated technology is widely used to modify the genomes rapidly and efficiently on specific DNA double-strand breaks (DSBs) induced by Cas9 endonuclease. However, despite swift advance in Cas9 engineering, structural basis of Cas9-recognition and cleavage complex remains unclear. Proper assembly of this complex correlates to effective Cas9 activity, leading to high efficacy of genome editing events. Here, we develop a CRISPR/Cas9-RAD51 plasmid constitutively expressing RAD51, which can bind to singlestranded DNA for DSB repair. We show that the efficiency of CRISPR-mediated genome editing can be significantly improved by expressing RAD51, responsible for DSB repair via homologous recombination (HR), in both gene knock-out and knock-in processes. In cells with CRISPR/Cas9-RAD51 plasmid, expression of the target genes (cohesin SMC3 and GAPDH) was reduced by more than 1.9-fold compared to the CRISPR/Cas9 plasmid for knock-out of genes. Furthermore, CRISPR/Cas9-RAD51 enhanced the knock-in efficiency of DsRed donor DNA. Thus, the CRISPR/Cas9-RAD51 system is useful for applications requiring precise and efficient genome edits not accessible to HR-deficient cell genome editing and for developing CRISPR/Cas9-mediated knockout technology. [BMB Reports 2023; 56(2): 102-107].


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Proteína 9 Associada à CRISPR/genética , Quebras de DNA de Cadeia Dupla , Genoma
4.
BMB Rep ; 56(2): 108-113, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36571142

RESUMO

Cohesin is a ring-shaped protein complex that comprises the SMC1, SMC3, and α-kleisin proteins, STAG1/2/3 subunits, and auxiliary factors. Cohesin participates in chromatin remodeling, chromosome segregation, DNA replication, and gene expression regulation during the cell cycle. Mitosis-specific α-kleisin factor RAD21 and meiosis-specific α-kleisin factor REC8 are expressed in embryonic stem cells (ESCs) to maintain pluripotency. Here, we demonstrated that RAD21 and REC8 were involved in maintaining genomic stability and modulating chromatin modification in murine ESCs. When the kleisin subunits were depleted, DNA repair genes were downregulated, thereby reducing cell viability and causing replication protein A (RPA) accumulation. This finding suggested that the repair of exposed single-stranded DNA was inefficient. Furthermore, the depletion of kleisin subunits induced DNA hypermethylation by upregulating DNA methylation proteins. Thus, we proposed that the cohesin complex plays two distinct roles in chromatin remodeling and genomic integrity to ensure the maintenance of pluripotency in ESCs. [BMB Reports 2023; 56(2): 108-113].


Assuntos
Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona , Animais , Camundongos , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Cromossomos/metabolismo , Meiose , Mitose , Coesinas
5.
Mol Cells ; 45(11): 820-832, 2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36172976

RESUMO

As a potential candidate to generate an everlasting cell source to treat various diseases, embryonic stem cells are regarded as a promising therapeutic tool in the regenerative medicine field. Cohesin, a multi-functional complex that controls various cellular activities, plays roles not only in organizing chromosome dynamics but also in controlling transcriptional activities related to self-renewal and differentiation of stem cells. Here, we report a novel role of the α-kleisin subunits of cohesin (RAD21 and REC8) in the maintenance of the balance between these two stem-cell processes. By knocking down REC8, RAD21, or the non-kleisin cohesin subunit SMC3 in mouse embryonic stem cells, we show that reduction in cohesin level impairs their self-renewal. Interestingly, the transcriptomic analysis revealed that knocking down each cohesin subunit enables the differentiation of embryonic stem cells into specific lineages. Specifically, embryonic stem cells in which cohesin subunit RAD21 were knocked down differentiated into cells expressing neural alongside germline lineage markers. Thus, we conclude that cohesin appears to control the fate determination of embryonic stem cells.


Assuntos
Meiose , Proteínas Nucleares , Animais , Camundongos , Proteínas Nucleares/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ciclo Celular/genética , Células-Tronco Embrionárias , Coesinas
6.
Genome Biol ; 23(1): 70, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35241136

RESUMO

BACKGROUND: Cohesin is a chromosome-associated SMC-kleisin complex that mediates sister chromatid cohesion, recombination, and most chromosomal processes during mitosis and meiosis. However, it remains unclear whether meiosis-specific cohesin complexes are functionally active in mitotic chromosomes. RESULTS: Through high-resolution 3D-structured illumination microscopy (3D-SIM) and functional analyses, we report multiple biological processes associated with the meiosis-specific cohesin components, α-kleisin REC8 and STAG3, and the distinct loss of function of meiotic cohesin during the cell cycle of embryonic stem cells (ESCs). First, we show that STAG3 is required for the efficient localization of REC8 to the nucleus by interacting with REC8. REC8-STAG3-containing cohesin regulates topological properties of chromosomes and maintains sister chromatid cohesion. Second, REC8-cohesin has additional sister chromatid cohesion roles in concert with mitotic RAD21-cohesin on ESC chromosomes. SIM imaging of REC8 and RAD21 co-staining revealed that the two types of α-kleisin subunits exhibited distinct loading patterns along ESC chromosomes. Third, knockdown of REC8 or RAD21-cohesin not only leads to higher rates of premature sister chromatid separation and delayed replication fork progression, which can cause proliferation and developmental defects, but also enhances chromosome compaction by hyperloading of retinoblastoma protein-condensin complexes from the prophase onward. CONCLUSIONS: Our findings indicate that the delicate balance between mitotic and meiotic cohesins may regulate ESC-specific chromosomal organization and the mitotic program.


Assuntos
Proteínas de Ciclo Celular , Proteínas Nucleares , Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona , Cromossomos , Células-Tronco Embrionárias/metabolismo , Meiose , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Coesinas
7.
Exp Mol Med ; 52(8): 1220-1229, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32770082

RESUMO

Embryonic stem cells (ESCs) possess specific gene expression patterns that confer the ability to proliferate indefinitely and enable pluripotency, which allows ESCs to differentiate into diverse cell types in response to developmental signals. Compared to differentiated cells, ESCs harbor an elevated level of homologous recombination (HR)-related proteins and exhibit exceptional cell cycle control, characterized by a high proliferation rate and a prolonged S phase. HR is involved in several aspects of chromosome maintenance. For instance, HR repairs impaired chromosomes and prevents the collapse of DNA replication forks during cell proliferation. Thus, HR is essential for the maintenance of genomic integrity and prevents cellular dysregulation and lethal events. In addition, abundant HR proteins in the prolonged S phase can efficiently protect ESCs from external damages and protect against genomic instability caused by DNA breaks, facilitating rapid and accurate DNA break repair following chromosome duplication. The maintenance of genome integrity is key to preserving the functions of ESCs and reducing the risks of cancer development, cell cycle arrest, and abnormal replication. Here, we review the fundamental links between the stem cell-specific HR process and DNA damage response as well as the different strategies employed by ESCs to maintain genomic integrity.


Assuntos
Células-Tronco Embrionárias/metabolismo , Instabilidade Genômica , Recombinação Homóloga/genética , Animais , Ciclo Celular/genética , Replicação do DNA/genética , Células-Tronco Embrionárias/citologia , Humanos , Modelos Biológicos
8.
Nat Commun ; 11(1): 2010, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332759

RESUMO

The balance between major DNA double-strand break (DSB) repair pathways is influenced by binding of the Ku complex, a XRCC5/6 heterodimer, to DSB ends, initiating non-homologous end joining (NHEJ) but preventing additional DSB end resection and homologous recombination (HR). However, the key molecular cue for Ku recruitment to DSB sites is unknown. Here, we report that FOXL2, a forkhead family transcriptional factor, directs DSB repair pathway choice by acetylation-dependent binding to Ku. Upon DSB induction, SIRT1 translocates to the nucleus and deacetylates FOXL2 at lysine 124, leading to liberation of XRCC5 and XRCC6 from FOXL2 and formation of the Ku complex. FOXL2 ablation enhances Ku recruitment to DSB sites, imbalances DSB repair kinetics by accelerating NHEJ and inhibiting HR, and thus leads to catastrophic genomic events. Our study unveils the SIRT1-(de)acetylated FOXL2-Ku axis that governs the balance of DSB repair pathways to maintain genome integrity.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Proteína Forkhead Box L2/metabolismo , Autoantígeno Ku/metabolismo , Acetilação , Linhagem Celular Tumoral , Proteína Forkhead Box L2/genética , Células HEK293 , Recombinação Homóloga , Humanos , Autoantígeno Ku/genética , Mutação , Ligação Proteica/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sirtuína 1/metabolismo
9.
Exp Mol Med ; 51(9): 1-12, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31534120

RESUMO

The human genome is constantly exposed to both endogenous and exogenous stresses, which can lead to errors in DNA replication and the accumulation of DNA mutations, thereby increasing the risk of cancer development. The transcription factor E2F1 is a key regulator of DNA repair. E2F1 also has defined roles in the replication of many cell cycle-related genes and is highly expressed in cancer cells, and its abundance is strongly associated with poor prognosis in cancers. Studies on colon cancer have demonstrated that the depletion of E2F1 leads to reduced levels of homologous recombination (HR), resulting in interrupted DNA replication and the subsequent accumulation of DNA lesions. Our results demonstrate that the depletion of E2F1 also causes reduced RAD51-mediated DNA repair and diminished cell viability resulting from DNA damage. Furthermore, the extent of RAD51 and RPA colocalization is reduced in response to DNA damage; however, RPA single-stranded DNA (ssDNA) nucleofilament formation is not affected following the depletion of E2F1, implying that ssDNA gaps accumulate when RAD51-mediated DNA gap filling or repair is diminished. Surprisingly, we also demonstrate that E2F1 forms foci with RAD51 or RPA at DNA break sites on damaged DNA. These findings provide evidence of a molecular mechanism underlying the E2F1-mediated regulation of HR activity and predict a fundamental shift in the function of E2F1 from regulating cell division to accelerating tumor development.


Assuntos
Fator de Transcrição E2F1/genética , Recombinação Homóloga/genética , Rad51 Recombinase/genética , Proteína de Replicação A/genética , Carcinogênese/genética , Divisão Celular/genética , Neoplasias do Colo/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/genética , Reparo do DNA/genética , Replicação do DNA/genética , DNA de Cadeia Simples/genética , Instabilidade Genômica/efeitos dos fármacos , Células HCT116 , Recombinação Homóloga/efeitos dos fármacos , Humanos , Metanossulfonato de Metila/farmacologia
10.
Exp Mol Med ; 50(8): 1-12, 2018 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-30154456

RESUMO

An important event enabling meiotic prophase I to proceed is the close juxtaposition of conjoined chromosome axes of homologs and their assembly via an array of transverse filaments and meiosis-specific axial elements into the synaptonemal complex (SC). During meiosis, recombination requires the establishment of a platform for recombinational interactions between the chromosome axes and their subsequent stabilization. This is essential for ensuring crossover recombination and proper segregation of homologous chromosomes. Thus, well-established SCs are essential for supporting these processes. The regulation of recombination intermediates on the chromosome axis/SC and dynamic positioning of double-strand breaks are not well understood. Here, using super-resolution microscopy (structured illumination microscopy), we determined the localization of the replication protein A (RPA) complex on the chromosome axes in the early phase of leptonema/zygonema and within the CEs of SC in the pachynema during meiotic prophase in mouse spermatocytes. RPA, which marks the intermediate steps of pairing and recombination, appears in large numbers and is positioned on the chromosome axes at the zygonema. In the pachynema, RPA foci are reduced but do not completely disappear; instead, they are placed between lateral elements. Our results reveal the precise structure of SC and localization dynamics of recombination intermediates on meiocyte chromosomes undergoing homolog pairing and meiotic recombination.


Assuntos
Cromossomos de Mamíferos/genética , Imageamento Tridimensional , Mamíferos/metabolismo , Meiose , Microscopia/métodos , Proteína de Replicação A/metabolismo , Animais , Arabidopsis/ultraestrutura , Pareamento Cromossômico , Cromossomos de Plantas/ultraestrutura , Reparo do DNA , Histonas/metabolismo , Camundongos Endogâmicos C57BL , Polimerização , Complexo Sinaptonêmico
11.
Mol Ther ; 26(4): 1154-1165, 2018 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-29503196

RESUMO

Homologous recombination (HR), which ensures accurate DNA replication and strand-break repair, is necessary to preserve embryonic stem cell (ESC) self-renewal. However, little is known about how HR factors modulate ESC differentiation and replication stress-associated DNA breaks caused by unique cell-cycle progression. Here, we report that ESCs utilize Rad51-dependent HR to enhance viability and induce rapid proliferation through a replication-coupled pathway. In addition, ESC differentiation was shown to be enhanced by ectopic expression of a subset of recombinases. Abundant expression of HR proteins throughout the ESC cycle, but not during differentiation, facilitated immediate HR-mediated repair of single-stranded DNA (ssDNA) gaps incurred during S-phase, via a mechanism that does not perturb cellular progression. Intriguingly, combined ectopic expression of two recombinases, Rad51 and Rad52, resulted in efficient ESC differentiation and diminished cell death, indicating that HR factors promote cellular differentiation by repairing global DNA breaks induced by chromatin remodeling signals. Collectively, these findings provide insight into the role of key HR factors in rapid DNA break repair following chromosome duplication during self-renewal and differentiation of ESCs.


Assuntos
Diferenciação Celular/genética , Expressão Ectópica do Gene , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Recombinação Homóloga , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Animais , Biomarcadores , Ciclo Celular/genética , Linhagem Celular , Autorrenovação Celular , Cromatina/genética , Cromatina/metabolismo , Quebras de DNA de Cadeia Dupla , Camundongos
12.
Sci Rep ; 7(1): 11610, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28912486

RESUMO

Embryonic stem (ES) cells require homologous recombination (HR) to cope with genomic instability caused during self-renewal. Here, we report expression dynamics and localization of endogenous HR factors in DNA break repair of ES cells. In addition, we analyzed gene expression patterns of HR-related factors at the transcript level with RNA-sequencing experiments. We showed that ES cells constitutively expressed diverse HR proteins throughout the cell cycle and that HR protein expression was not significantly changed even in the DNA damaging conditions. We further analyzed that depleting Rad51 resulted in the accumulation of larger single-stranded DNA (ssDNA) gaps, but did not perturb DNA replication, indicating that ES cells were able to enter the G2-phase in the presence of unrepaired DNA gaps, consistent with the possibility that post-replication repair helps avoid stalling at the G2/M checkpoint. Interestingly, caffeine treatment inhibited the formation of Rad51 or Rad54 foci, but not the formation of γH2AX and Exo1 foci, which led to incomplete HR in ssDNA, thus increasing DNA damage sensitivity. Our results suggested that ES cells possess conserved HR-promoting machinery to ensure effective recruitment of the HR proteins to DNA breaks, thereby driving proper chromosome duplication and cell cycle progression in ES cells.


Assuntos
Reparo do DNA , Replicação do DNA , Recombinação Homóloga , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Biomarcadores , Ciclo Celular/genética , Dano ao DNA , DNA Helicases/metabolismo , Camundongos , Rad51 Recombinase/metabolismo
13.
Mol Cells ; 40(2): 143-150, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28190324

RESUMO

Homologous recombination (HR) is necessary for maintenance of genomic integrity and prevention of various mutations in tumor suppressor genes and proto-oncogenes. Rad51 and Rad54 are key HR factors that cope with replication stress and DNA breaks in eukaryotes. Rad51 binds to single-stranded DNA (ssDNA) to form the presynaptic filament that promotes a homology search and DNA strand exchange, and Rad54 stimulates the strand-pairing function of Rad51. Here, we studied the molecular dynamics of Rad51 and Rad54 during the cell cycle of HeLa cells. These cells constitutively express Rad51 and Rad54 throughout the entire cell cycle, and the formation of foci immediately increased in response to various types of DNA damage and replication stress, except for caffeine, which suppressed the Rad51-dependent HR pathway. Depletion of Rad51 caused severe defects in response to postreplicative stress. Accordingly, HeLa cells were arrested at the G2-M transition although a small amount of Rad51 was steadily maintained in HeLa cells. Our results suggest that cell cycle progression and proliferation of HeLa cells can be tightly controlled by the abundance of HR proteins, which are essential for the rapid response to postreplicative stress and DNA damage stress.


Assuntos
Dano ao DNA , DNA Helicases/genética , Proteínas Nucleares/genética , Rad51 Recombinase/genética , Ciclo Celular/genética , Divisão Celular/genética , DNA Helicases/biossíntese , Proteínas de Ligação a DNA , Fase G2/genética , Células HeLa , Recombinação Homóloga , Humanos , Proteínas Nucleares/biossíntese , Rad51 Recombinase/biossíntese , Estresse Fisiológico/genética
14.
J Microbiol Biotechnol ; 25(7): 1026-35, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25975613

RESUMO

Condensin is not only responsible for chromosome condensation, but is also involved in double-strand break (DSB) processing in the cell cycle. During meiosis, the condensin complex serves as a component of the meiotic chromosome axis, and mediates both proper assembly of the synaptonemal complex and DSB repair, in order to ensure proper homologous chromosome segregation. Here, we used the budding yeast Saccharomyces cerevisiae to show that condensin participates in a variety of chromosome organization processes and exhibits crucial molecular functions that contribute to meiotic recombination during meiotic prophase I. We demonstrate that Ycs4 is required for efficient DSB formation and establishing homolog bias at the early stage of meiotic prophase I, which allows efficient formation of interhomolog recombination products. In the Ycs4 meiosis-specific allele (ycs4S), interhomolog products were formed at substantial levels, but with the same reduction in crossovers and noncrossovers. We further show that, in prophase chromosomal events, ycs4S relieved the defects in the progression of recombination interactions induced as a result of the absence of Rec8. These results suggest that condensin is a crucial coordinator of the recombination process and chromosome organization during meiosis.


Assuntos
Adenosina Trifosfatases/metabolismo , Cromossomos Fúngicos/metabolismo , Quebras de DNA de Cadeia Dupla , DNA Fúngico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Recombinação Homóloga , Meiose , Complexos Multiproteicos/metabolismo , Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
15.
Angle Orthod ; 72(1): 36-42, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11843271

RESUMO

The purpose of this study was to identify the potential projection errors of posteroanterior cephalometric radiographs due to head rotation in the vertical Z-axis. For this investigation, 20 human dry skull samples with permanent dentition were collected from the Department of Anatomy in the College of Medicine, Chosun University, Korea. They had no gross asymmetries and were well preserved. Each dry skull was rotated from 0 degrees to +/- 10 degrees at 1 degrees intervals. A vertical axis, the Z-axis, was used as a rotational axis for the exposure of 420 posteroanterior cephalometric radiographs. Most of the abscissa values of each landmark showed statistically significant differences in the head rotation from each rotational angle (P < .05), whereas the ordinate values were almost the same in all rotational angles regardless of the head rotation. The abscissa values of each landmark anterior to the vertical rotational axis displaced in the same direction as the head rotation, whereas those of other landmarks posterior to the vertical rotational axis displaced in the opposite direction. The mean differences of the abscissa values, per 1 degrees of head rotation, were larger as the landmark was located further anteroposteriorly from the vertical rotational axis and smaller as the landmark was located nearer the vertical rotational axis. In view of the projection error, a posteroanterior cephalometric radiograph is a more valuable diagnostic tool when it is exposed with no head rotation about the vertical Z-axis.


Assuntos
Cefalometria , Erros de Diagnóstico , Assimetria Facial/diagnóstico por imagem , Cabeça , Postura , Humanos , Radiografia , Valores de Referência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...