Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Langenbecks Arch Surg ; 408(1): 388, 2023 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-37796313

RESUMO

BACKGROUND: Postoperative adhesions are frequent and significant complications that typically arise following abdominal surgery. Currently, the existing evidence for predicting the risk of adhesive small bowel obstruction (ASBO) after emergency gastrointestinal surgery (EGS) remains inadequate. A reliable perioperative model that quantifies the risk of ASBO after EGS serves as a practical tool for guiding individually tailored surveillance. METHODS: A consecutive series of 1296 patients who underwent EGS for radiologically confirmed bowel/visceral inflammation or perforation between 2012 and 2022 at a tertiary academic medical center were included in this study to establish a best-fit nomogram. The nomogram was externally validated by assessing discrimination and calibration using an independent cohort from a separate medical center. RESULTS: A total of 116 patients (8.9%) developed at least one episode of ASBO after EGS during a median follow-up duration of 26 months. The results of multivariable logistic analysis indicated that male sex (P = 0.043), preoperative albumin level (P = 0.002), history of pelvic radiotherapy (P = 0.038), laparotomy (P = 0.044), and intensive care unit stay ≥ 72 h (P = 0.047) were identified as independent risk factors for developing ASBO. By incorporating these predictors, the developed nomogram exhibited good accuracy in risk estimation, as evidenced by a guide-corrected C-index score of 0.852 (95% CI 0.667-0.920) in the external validation cohort. Decision curve analysis and clinical impact curve demonstrated a clinically effective predictive model. CONCLUSION: By incorporating the nomogram as a supplemental tool in perioperative management, it becomes possible to accurately assess the individual's likelihood of developing ASBOs. This quantification enables surgeons to implement appropriate preventive measures, ultimately leading to improved outcomes.


Assuntos
Procedimentos Cirúrgicos do Sistema Digestório , Obstrução Intestinal , Humanos , Masculino , Procedimentos Cirúrgicos do Sistema Digestório/efeitos adversos , Nomogramas , Estudos Retrospectivos , Obstrução Intestinal/diagnóstico , Obstrução Intestinal/etiologia , Obstrução Intestinal/cirurgia , Aderências Teciduais/etiologia
2.
Clin Med Insights Oncol ; 16: 11795549221137399, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36468151

RESUMO

Background: Colorectal cancer (CRC) has a high prevalence and poor prognosis. This study aimed to identify biomarkers related to the clinical stage (I-IV) of CRC. Methods: The LinkedOmics database was used as the discovery cohort, and two Gene Expression Omnibus (GEO) databases (GSE41258 and GSE422848) served as validation cohorts. The trend test of genes related to clinical stage (I-IV) of CRC patients was identified by the Jonckheere-Terpstra test. The cBioPortal database, Gene Expression Profiling Interactive Analysis (GEPIA) and PrognoScan databases were used to explore the expression change and prognostic value of clinical stage-related genes in CRC patients. CRC cells overexpressed AGPAT5 were constructed and used for cell counting kit-8 (CCK-8), flow cytometric, and wound healing assays in vitro. Results: We identified four clinical stage-related genes, GSR, AGPAT5, CRLF1, and NPR3, in CRC. The CNA frequencies of GSR, CRLF1, AGPAT5, and NPR3 occurred in 11%, 2.4%, 13%, and 3% of patients, respectively. The expression of GSR and AGPAT5 tended to decrease with CRC stage (I-IV) progression, and the expression of CRLF1 and NPR3 tended to increase with CRC stage (I-IV) progression. Compared with the normal group, AGPAT5 expression was markedly decreased in stage IV CRC. Higher GSR and AGPAT5 expression levels were associated with better overall survival (OS) and disease-free survival (DFS) in CRC patients. Lower CRLF1 and NPR3 expression levels were associated with better OS and DFS in CRC. GSR, CRLF1, AGPAT5, and NPR3 expression were related to CRC progression, microsatellite instability, and tumour purity in CRC. Furthermore, AGPAT5 was downregulated in CRC cell lines, and overexpression of AGPAT5 inhibited cell proliferation and migration and promoted cell apoptosis in CRC cells. Conclusion: Low AGPAT5 expression may serve as a poor prognostic factor and clinical stage biomarker in CRC. In addition, AGPAT5 acts as a tumour suppressor in CRC progression.

3.
Bioinformatics ; 37(21): 3774-3780, 2021 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-34196686

RESUMO

MOTIVATION: The increasing amount of time-series single-cell RNA sequencing (scRNA-seq) data raises the key issue of connecting cell states (i.e. cell clusters or cell types) to obtain the continuous temporal dynamics of transcription, which can highlight the unified biological mechanisms involved in cell state transitions. However, most existing trajectory methods are specifically designed for individual cells, so they can hardly meet the needs of accurately inferring the trajectory topology of the cell state, which usually contains cells assigned to different branches. RESULTS: Here, we present CStreet, a computed Cell State trajectory inference method for time-series scRNA-seq data. It uses time-series information to construct the k-nearest neighbor connections between cells within each time point and between adjacent time points. Then, CStreet estimates the connection probabilities of the cell states and visualizes the trajectory, which may include multiple starting points and paths, using a force-directed graph. By comparing the performance of CStreet with that of six commonly used cell state trajectory reconstruction methods on simulated data and real data, we demonstrate the high accuracy and high tolerance of CStreet. AVAILABILITY AND IMPLEMENTATION: CStreet is written in Python and freely available on the web at https://github.com/TongjiZhanglab/CStreet and https://doi.org/10.5281/zenodo.4483205. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Perfilação da Expressão Gênica , Software , Análise de Sequência de RNA/métodos , Perfilação da Expressão Gênica/métodos , Análise de Célula Única/métodos , Probabilidade
4.
J Mol Cell Biol ; 11(6): 463-473, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30299501

RESUMO

Trophoblast stem cells (TSCs), which can be derived from the trophoectoderm of a blastocyst, have the ability to sustain self-renewal and differentiate into various placental trophoblast cell types. Meanwhile, essential insights into the molecular mechanisms controlling the placental development can be gained by using TSCs as the cell model. Esrrb is a transcription factor that has been shown to play pivotal roles in both embryonic stem cell (ESC) and TSC, but the precise mechanism whereby Esrrb regulates TSC-specific transcriptome during differentiation and reprogramming is still largely unknown. In the present study, we elucidate the function of Esrrb in self-renewal and differentiation of TSCs, as well as during the induced TSC (iTSC) reprogramming. We demonstrate that the precise level of Esrrb is critical for stem state maintenance and further trophoblast differentiation of TSCs, as ectopically expressed Esrrb can partially block the rapid differentiation of TSCs in the absence of fibroblast growth factor 4. However, Esrrb depletion results in downregulation of certain key TSC-specific transcription factors, consequently causing a rapid differentiation of TSCs and these Esrrb-deficient TSCs lose the ability of hemorrhagic lesion formation in vivo. This function of Esrrb is exerted by directly binding and activating a core set of TSC-specific target genes including Cdx2, Eomes, Sox2, Fgfr4, and Bmp4. Furthermore, we show that Esrrb overexpression can facilitate the MEF-to-iTSC conversion. Moreover, Esrrb can substitute for Eomes to generate GEsTM-iTSCs. Thus, our findings provide a better understanding of the molecular mechanism of Esrrb in maintaining TSC self-renewal and during iTSC reprogramming.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Receptores de Estrogênio/metabolismo , Trofoblastos/metabolismo , Animais , Técnicas de Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Camundongos Transgênicos , Receptores de Estrogênio/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Trofoblastos/citologia
5.
Cell Stem Cell ; 23(3): 426-435.e5, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30146410

RESUMO

Somatic cell nuclear transfer (SCNT) enables cloning of differentiated cells by reprogramming their nuclei to a totipotent state. However, successful full-term development of SCNT embryos is a low-efficiency process and arrested embryos frequently exhibit epigenetic abnormalities. Here, we generated genome-wide DNA methylation maps from mouse pre-implantation SCNT embryos. We identified widespread regions that were aberrantly re-methylated, leading to mis-expression of genes and retrotransposons important for zygotic genome activation. Inhibition of DNA methyltransferases (Dnmts) specifically rescued these re-methylation defects and improved the developmental capacity of cloned embryos. Moreover, combining inhibition of Dnmts with overexpression of histone demethylases led to stronger reductions in inappropriate DNA methylation and synergistic enhancement of full-term SCNT embryo development. These findings show that excessive DNA re-methylation is a potent barrier that limits full-term development of SCNT embryos and that removing multiple epigenetic barriers is a promising approach to achieve higher cloning efficiency.


Assuntos
Metilação de DNA , DNA/metabolismo , Desenvolvimento Embrionário , Técnicas de Transferência Nuclear , Animais , Células Cultivadas , DNA/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos ICR
6.
Biomaterials ; 119: 53-67, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28006658

RESUMO

The generation of functional neural progenitor cells (NPCs) holds great promise for both research and clinical applications in neurodegenerative diseases. Traditionally, NPCs are derived from embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), or NPCs can be directly converted from somatic cells by sets of transcription factors or by a combination of chemical cocktails and/or hypoxia. However, the ethical issues of ESCs, the risk of tumorigenesis from iPSCs and transgenic integration from exogenous genes as well as complicated manipulation and time-consuming of chemical induced NPCs (ciNPCs) limit the applications of these strategies. Here, we describe a novel method for generating growth factor-induced neural progenitor cells (giNPCs) from mouse embryonic and adult fibroblasts by using inductive and/or permissive signaling culture conditions. These giNPCs closely resemble brain-derived NPCs in terms of transcription networks and neural lineage differentiation potentials. Moreover, this somatic cell to NPC induction is a gradual process that includes initiation, intermediate, maturation and stabilization stages. Importantly, gene expression and histone modification analyses further indicate a partially reprogrammed state during the generation process of induced NPCs, in which lineage specific genes and pluripotency associated genes are transiently activated. Our study therefore describes the potential safety problems that also exist in the transgene-free direct induction strategy and highlights the importance of excluding the possibility of residual partially reprogrammed and/or teratoma-like cells from the generated NPCs for future clinical trials.


Assuntos
Técnicas de Reprogramação Celular/métodos , Reprogramação Celular/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Animais , Técnicas de Cultura Celular por Lotes/métodos , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Engenharia Tecidual/métodos
7.
Cell Discov ; 2: 16010, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27462457

RESUMO

Differentiated somatic cells can be reprogrammed into totipotent embryos through somatic cell nuclear transfer. However, most cloned embryos arrest at early stages and the underlying molecular mechanism remains largely unexplored. Here, we first developed a somatic cell nuclear transfer embryo biopsy system at two- or four-cell stage, which allows us to trace the developmental fate of the biopsied embryos precisely. Then, through single-cell transcriptome sequencing of somatic cell nuclear transfer embryos with different developmental fates, we identified that inactivation of Kdm4b, a histone H3 lysine 9 trimethylation demethylase, functions as a barrier for two-cell arrest of cloned embryos. Moreover, we discovered that inactivation of another histone demethylase Kdm5b accounts for the arrest of cloned embryos at the four-cell stage through single-cell analysis. Co-injection of Kdm4b and Kdm5b can restore transcriptional profiles of somatic cell nuclear transfer embryos and greatly improve the blastocyst development (over 95%) as well as the production of cloned mice. Our study therefore provides an effective approach to identify key factors responsible for the developmental arrest of somatic cell cloned embryos.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA