Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Laryngoscope Investig Otolaryngol ; 7(1): 161-169, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35155794

ABSTRACT

BACKGROUND: To build an automatic pathological diagnosis model to assess the lymph node metastasis status of head and neck squamous cell carcinoma (HNSCC) based on deep learning algorithms. STUDY DESIGN: A retrospective study. METHODS: A diagnostic model integrating two-step deep learning networks was trained to analyze the metastasis status in 85 images of HNSCC lymph nodes. The diagnostic model was tested in a test set of 21 images with metastasis and 29 images without metastasis. All images were scanned from HNSCC lymph node sections stained with hematoxylin-eosin (HE). RESULTS: In the test set, the overall accuracy, sensitivity, and specificity of the diagnostic model reached 86%, 100%, and 75.9%, respectively. CONCLUSIONS: Our two-step diagnostic model can be used to automatically assess the status of HNSCC lymph node metastasis with high sensitivity. LEVEL OF EVIDENCE: NA.

2.
Oncogene ; 39(11): 2358-2376, 2020 03.
Article in English | MEDLINE | ID: mdl-31911619

ABSTRACT

Dysregulated metabolism contributes to cancer initiation and progression, but the key drivers of these pathways are just being discovered. Here, we report a critical role for proline catabolism in non-small cell lung cancer (NSCLC). Proline dehydrogenase (PRODH) is activated to reduce proline levels by the chromatin remodeling factor lymphoid-specific helicase (LSH), an epigenetic driver of NSCLC. PRODH promotes NSCLC tumorigenesis by inducing epithelial to mesenchymal transition (EMT) and IKKα-dependent inflammatory genes, including CXCL1, LCN2, and IL17C. Consistently, proline addition promotes the expression of these inflammatory genes, as well as EMT, tumor cell proliferation, and migration in vitro and tumor growth in vivo, while the depletion or inhibition of PRODH blocks these phenotypes. In summary, we reveal an essential metabolic pathway amenable to targeting in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Proline/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Humans , Lung Neoplasms/pathology , Mice , Transfection
3.
Mol Biomed ; 1(1): 7, 2020 Sep 20.
Article in English | MEDLINE | ID: mdl-35006429

ABSTRACT

Cyclic GMP-AMP (cGAMP) synthase (cGAS) is a cytosolic DNA sensor and innate immune response initiator. Binding with exogenous or endogenous nucleic acids, cGAS activates its downstream adaptor, stimulator of interferon genes (STING). STING then triggers protective immune to enable the elimination of the pathogens and the clearance of cancerous cells. Apparently, aberrantly activated by self-DNA, cGAS/STING pathway is threatening to cause autoimmune and inflammatory diseases. The effects of cGAS/STING in defenses against infection and autoimmune diseases have been well studied, still it is worthwhile to discuss the roles of cGAS/STING pathway beyond the "classical" realm of innate immunity. Recent studies have revealed its involvement in non-canonical inflammasome formation, calcium hemostasis regulation, endoplasmic reticulum (ER) stress response, perception of leaking mitochondrial DNA (mtDNA), autophagy induction, cellular senescence and senescence-associated secretory phenotype (SASP) production, providing an exciting area for future exploration. Previous studies generally focused on the function of cGAS/STING pathway in cytoplasm and immune response. In this review, we summarize the latest research of this pathway on the regulation of other physiological process and STING independent reactions to DNA in micronuclei and nuclei. Together, these studies provide a new perspective of cGAS/STING pathway in human diseases.

4.
Article in English | MEDLINE | ID: mdl-31637019

ABSTRACT

The trillions of microorganisms in the gut microbiome have attracted much attention recently owing to their sophisticated and widespread impacts on numerous aspects of host pathophysiology. Remarkable progress in large-scale sequencing and mass spectrometry has increased our understanding of the influence of the microbiome and/or its metabolites on the onset and progression of extraintestinal cancers and the efficacy of cancer immunotherapy. Given the plasticity in microbial composition and function, microbial-based therapeutic interventions, including dietary modulation, prebiotics, and probiotics, as well as fecal microbial transplantation, potentially permit the development of novel strategies for cancer therapy to improve clinical outcomes. Herein, we summarize the latest evidence on the involvement of the gut microbiome in host immunity and metabolism, the effects of the microbiome on extraintestinal cancers and the immune response, and strategies to modulate the gut microbiome, and we discuss ongoing studies and future areas of research that deserve focused research efforts.

5.
Oncogene ; 38(46): 7133-7145, 2019 11.
Article in English | MEDLINE | ID: mdl-31417184

ABSTRACT

Elucidating mechanisms in tumor suppressors and epigenetic modifiers are needed to gain insights into the etiology and treatment of cancer, the interplay between long intergenic non-coding RNAs (lncRNAs) and chromatin remodeling remains unclear. Here, we showed that GIAT4RA, a poorly characterized lncRNA LOC102723729, was significantly decreased in lung cancer cells and tissues; while no association was observed with clinical risk factors, expression was linked with clinical stage and lymphatic metastasis. Higher expression of GIAT4RA was linked with overall survival in NSCLC. GIAT4RA inhibited many characteristics of tumorigenesis including cell growth, clonal formation, migration and invasion, epithelial-mesenchymal transition, tumor sphere and tumor growth in vivo. Mechanistically, GIAT4RA was essential for the degradation of chromatin modifier lymphoid-specific helicase (LSH) by counteracting the deubiquintination in proteasome pathway by binding to 227-589 AA of LSH. GIAT4RA interfered with ubiquitin hydrolase Uchl3-mediated interaction and stabilization of LSH. LSH knockdown rescued GIAT4RA-promoted features, and LSH overexpression prevented GIAT4RA-induced phenotypes. Taken together, lncRNA GIAT4RA plays a critical role in NSCLC adenocarcinoma as a ubiquitination regulator and tumor suppressor.


Subject(s)
Adenocarcinoma of Lung/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , DNA Helicases/metabolism , Gene Expression Regulation, Neoplastic/physiology , Lung Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Ubiquitin Thiolesterase/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Genes, Tumor Suppressor , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Ubiquitination
6.
Int J Biol Sci ; 15(6): 1187-1199, 2019.
Article in English | MEDLINE | ID: mdl-31223279

ABSTRACT

Traditional antitumor drugs inhibit the proliferation and metastasis of tumour cells by restraining the replication and expression of DNA. These drugs are usually highly cytotoxic. They kill tumour cells while also cause damage to normal cells at the same time, especially the hematopoietic cells that divide vigorously. Patients are exposed to other serious situations such as a severe infection caused by a decrease in the number of white blood cells. Energy metabolism is an essential process for the survival of all cells, but differs greatly between normal cells and tumour cells in metabolic pathways and metabolic intermediates. Whether this difference could be used as new therapeutic target while reducing damage to normal tissues is the topic of this paper. In this paper, we introduce five major metabolic intermediates in detail, including acetyl-CoA, SAM, FAD, NAD+ and THF. Their contents and functions in tumour cells and normal cells are significantly different. And the possible regulatory mechanisms that lead to these differences are proposed carefully. It is hoped that the key enzymes in these regulatory pathways could be used as new targets for tumour therapy.


Subject(s)
Antineoplastic Agents/adverse effects , Carcinogenesis/metabolism , Neoplasms/metabolism , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Acetyl Coenzyme A/physiology , Cell Proliferation/drug effects , Disease Progression , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Flavin-Adenine Dinucleotide/physiology , Humans , NAD/chemistry , NAD/metabolism , NAD/physiology , Neoplasm Invasiveness , Neoplasms/pathology , Neoplasms/therapy , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Tetrahydrofolates/chemistry , Tetrahydrofolates/metabolism , Tetrahydrofolates/physiology
7.
J Exp Clin Cancer Res ; 38(1): 280, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-31253190

ABSTRACT

BACKGROUND: Elucidating mechanisms in oncogenes and epigenetic modifiers are needed to gain insights into the etiology and treatment of cancer, regulation of oncogene by chromatin modifiers at post-transcriptional level is critical and remains unclear. We have investigated the role of GINS4 in NSCLC. METHODS: The expression of chromatin modifier lymphoid-specific helicase (LSH) and GINS4 was assessed in tumor and normal tissue from 79 patients with NSCLC with clinical characteristics. HBE, A549, H358, and H522, PC9, 95C and 95D were cultured after overexpression or silencing of GIAT4RA. Cell proliferation assay, cell migration and invasion assays, plate colony formation assay, immunofluorescence assay, Operetta® high-content screening and analysis, Western blot analysis and Co-Immunoprecipitation (Co-IP) assay, RNA immunoprecipitation assay and tumor growth assay was used to address the potential interplay of between GINS4 and LSH, and the functional of GINS4. RESULTS: GINS4 is highly expressed in lung cancer cells and tissues, and GINS4 expression is not association with clinical risk factors, but linked with clinical stage and lymphatic metastasis status. Higher expression of GINS4 poorly linked with overall survival in lung adenocarcinomas. Furthermore, GINS4 promoted many characteristics of tumorigenesis including cell growth, clonal formation, migration and invasion, epithelial-mesenchymal transition, tumor sphere and tumor growth in vivo. Interestingly, our results demonstrated that LSH increases GINS4 expression through binding to 3'UTR region of GINS4 and stabilizing its mRNA levels. Finally, LSH overexpression rescues GINS4 knockdown-induced features. CONCLUSIONS: GINS4 facilitates lung cancer progression by promoting key characteristics of tumor potential, and LSH epigenetically interacts with and stabilizes GINS4 transcripts.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/metabolism , Lung Neoplasms/genetics , RNA Stability , RNA, Messenger/metabolism , Animals , Carcinogenesis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/genetics , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Xenograft Model Antitumor Assays
8.
Am J Transl Res ; 11(12): 7604-7613, 2019.
Article in English | MEDLINE | ID: mdl-31934304

ABSTRACT

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) initiates a variety of signals that lead to the invasion and metastasis of gastric cancer. Though drugs targeting HER2 have been applied in clinical practice, drug resistance remains a big challenge. This study aimed to propose a new therapeutic target by exploring the regulating pathway of HER2. METHODS: Reverse transcription polymerase chain reaction (RT-PCR), western blot and immunohistochemistry staining were used respectively to detect the expression of HER2, Twist, E-cadherin and Fascin1 in both HER2 knockdown and overexpressed cell lines. Trans-well chamber assay and wound healing assay were used to detect the invasive ability of gastric cancer cells. The correlation between HER2 and Twist was analyzed based on specimens obtained from 118 patients with gastric cancer. RESULTS: HER2 silencing decreased the expression of Twist (P<0.05) and increased the expression of E-cadherin (P<0.05), while the expression of Fascin1 remained unchanged (P>0.05) and the migration and invasion abilities of cancer cells were weakened (P<0.01). On the contrary overexpression of HER2 increased the expression of Twist (P<0.05) and decreased the expression of E-cadherin (P<0.05), while the expression of Fascin1 still remained unchanged (P>0.05), and the migration and invasion abilities of cancer cells were enhanced (P<0.01). Our data indicated that the HER2 kinase domain was not involved in the regulation of Twist or E-cadherin. In addition, the expression of HER2 was positively correlated with the EMT-related transcription factor Twist in gastric cancer tissues. CONCLUSION: HER2 could promote the invasion and migration of gastric cancer cells by down-regulating E-cadherin and up-regulating Twist, which indicated that E-cadherin and Twist were both promising therapeutic targets.

9.
Oncotarget ; 9(1): 901-914, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29416665

ABSTRACT

Baicalin hydrate (BH), a natural compound, has been investigated for many years because of its traditional medicinal properties. However, the anti-tumor activities of BH and its epigenetic role in NPC have not been elucidated. In this study, we identified that BH inhibits NPC cell growth in vivo and in vitro by inducing apoptosis and cell cycle arrest. BH epigenetically regulated genome instability by up-regulating the expression of satellite 2 (Sat2), alpha satellite (α-Sat), and major satellite (Major-Sat). BH also increased the level of IKKα, Suv39H1, and H3K9me3 and decreased LSH expression. Interestingly, BH promoted the splicing of Suv39H1 via the enhancement of m6A RNA methylation, rather than DNA methylation. Taken together, our results demonstrated that BH has an anti-tumor role in NPC and revealed a unique role of BH in genome instability and splicing in response to DNA damage.

10.
Biochim Biophys Acta Rev Cancer ; 1868(2): 359-371, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28757126

ABSTRACT

Metabolism is essential to all living organisms that provide cells with energy, regulators, building blocks, enzyme cofactors and signaling molecules, and is in tune with nutritional conditions and the function of cells to make the appropriate developmental decisions or maintain homeostasis. As a fundamental biological process, metabolism state affects the production of multiple metabolites and the activation of various enzymes that participate in regulating gene expression, cell apoptosis, cancer progression and immunoreactions. Previous studies generally focus on the function played by the metabolic enzymes in the cytoplasm and mitochondrion. In this review, we conclude the role of them in the nucleus and their implications for cancer progression, immunity and metastasis.


Subject(s)
Cell Nucleus/metabolism , Immunity , Neoplasm Metastasis , Neoplasms/etiology , ATP Citrate (pro-S)-Lyase/physiology , Active Transport, Cell Nucleus , Animals , Carrier Proteins/physiology , Gene Expression Regulation , Humans , Membrane Proteins/physiology , Protein Transport , Pyruvate Dehydrogenase Complex/physiology , Thyroid Hormones/physiology , Thyroid Hormone-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...