Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
J Exp Med ; 221(3)2024 Mar 04.
Article En | MEDLINE | ID: mdl-38284990

Human lung adenosquamous cell carcinoma (LUAS), containing both adenomatous and squamous pathologies, exhibits strong cancer plasticity. We find that ALK rearrangement is detectable in 5.1-7.5% of human LUAS, and transgenic expression of EML4-ALK drives lung adenocarcinoma (LUAD) formation initially and squamous transition at late stage. We identify club cells as the main cell-of-origin for squamous transition. Through recapitulating lineage transition in organoid system, we identify JAK-STAT signaling, activated by EML4-ALK phase separation, significantly promotes squamous transition. Integrative study with scRNA-seq and immunostaining identify a plastic cell subpopulation in ALK-rearranged human LUAD showing squamous biomarker expression. Moreover, those relapsed ALK-rearranged LUAD show notable upregulation of squamous biomarkers. Consistently, mouse squamous tumors or LUAD with squamous signature display certain resistance to ALK inhibitor, which can be overcome by combined JAK1/2 inhibitor treatment. This study uncovers strong plasticity of ALK-rearranged tumors in orchestrating phenotypic transition and drug resistance and proposes a potentially effective therapeutic strategy.


Adenocarcinoma of Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Humans , Animals , Mice , Lung Neoplasms/genetics , Lung , Receptor Protein-Tyrosine Kinases , Oncogene Proteins, Fusion/genetics
2.
Zool Res ; 43(5): 805-812, 2022 Sep 18.
Article En | MEDLINE | ID: mdl-35993132

The divergence and continuous evolution of plants and animals contribute to ecological diversity. Promoters and transcription factors (TFs) are key determinants of gene regulation and transcription throughout life. However, the evolutionary trajectories and relationships of promoters and TFs are still poorly understood. Here, we conducted extensive analysis of large-scale multi-omics sequences in 420 animal species and 223 plant species spanning nearly a billion years of evolutionary history. Results showed that promoter GC-content and TF isoelectric points, as features/signatures that accompany long biological evolution, exhibited increasing growth in animal cells but a decreasing trend in plant cells. Furthermore, the evolutionary trajectories of promoter and TF signatures in the animal kingdom provided further evidence that Mammalia as well as Aves evolved directly from the ancestor Reptilia. The strong correlation between promoter and TF signatures indicates that promoters and TFs formed antagonistic coevolution in the animal kingdom, but mutualistic coevolution in the plant kingdom. The distinct coevolutionary patterns potentially drive the plant-animal divergence,divergent evolution and ecological diversity.


Gene Expression Regulation , Transcription Factors , Animals , Birds/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism
3.
J Mol Cell Biol ; 13(9): 622-635, 2021 12 06.
Article En | MEDLINE | ID: mdl-34097054

Tumor development is a process involving loss of the differentiation phenotype and acquisition of stem-like characteristics, which is driven by intracellular rewiring of signaling network. The measurement of network reprogramming and disorder would be challenging due to the complexity and heterogeneity of tumors. Here, we proposed signaling entropy (SR) to assess the degree of tumor network disorder. We calculated SR for 33 tumor types in The Cancer Genome Atlas database based on transcriptomic and proteomic data. The SR of tumors was significantly higher than that of normal samples and was highly correlated with cell stemness, cancer type, tumor grade, and metastasis. We further demonstrated the sensitivity and accuracy of using local SR in prognosis prediction and drug response evaluation. Overall, SR could reveal cancer network disorders related to tumor malignant potency, clinical prognosis, and drug response.


Carcinogenesis/metabolism , Models, Biological , Neoplasms/metabolism , Signal Transduction , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/pathology , Datasets as Topic , Entropy , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Proteomics , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Article En | MEDLINE | ID: mdl-33500348

ZFP57 is a master regulator of genomic imprinting. It has both maternal and zygotic functions that are partially redundant in maintaining DNA methylation at some imprinting control regions (ICRs). In this study, we found that DNA methylation was lost at most known ICRs in Zfp57 mutant embryos. Furthermore, loss of ZFP57 caused loss of parent-of-origin-dependent monoallelic expression of the target imprinted genes. The allelic expression switch occurred in the ZFP57 target imprinted genes upon loss of differential DNA methylation at the ICRs in Zfp57 mutant embryos. Specifically, upon loss of ZFP57, the alleles of the imprinted genes located on the same chromosome with the originally methylated ICR switched their expression to mimic their counterparts on the other chromosome with unmethylated ICR. Consistent with our previous study, ZFP57 could regulate the NOTCH signaling pathway in mouse embryos by impacting allelic expression of a few regulators in the NOTCH pathway. In addition, the imprinted Dlk1 gene that has been implicated in the NOTCH pathway was significantly down-regulated in Zfp57 mutant embryos. Our allelic expression switch models apply to the examined target imprinted genes controlled by either maternally or paternally methylated ICRs. Our results support the view that ZFP57 controls imprinted expression of its target imprinted genes primarily through maintaining differential DNA methylation at the ICRs.


Alleles , Genomic Imprinting , Repressor Proteins/genetics , Animals , DNA Methylation/genetics , Embryo, Mammalian/metabolism , Female , Mice , RNA-Seq , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction/genetics
5.
Zool Res ; 41(6): 599-604, 2020 Nov 18.
Article En | MEDLINE | ID: mdl-33124218

Gene regulatory networks play pivotal roles in our understanding of biological processes/mechanisms at the molecular level. Many studies have developed sample-specific or cell-type-specific gene regulatory networks from single-cell transcriptomic data based on a large amount of cell samples. Here, we review the state-of-the-art computational algorithms and describe various applications of gene regulatory networks in biological studies.


Computer Simulation , Gene Regulatory Networks/physiology , Models, Genetic , Sequence Analysis, RNA/methods , Animals , Single-Cell Analysis , Transcriptome
6.
Cancer Cell ; 38(5): 734-747.e9, 2020 11 09.
Article En | MEDLINE | ID: mdl-32888432

We integrate the genomics, proteomics, and phosphoproteomics of 480 clinical tissues from 146 patients in a Chinese colorectal cancer (CRC) cohort, among which 70 had metastatic CRC (mCRC). Proteomic profiling differentiates three CRC subtypes characterized by distinct clinical prognosis and molecular signatures. Proteomic and phosphoproteomic profiling of primary tumors alone successfully distinguishes cases with metastasis. Metastatic tissues exhibit high similarities with primary tumors at the genetic but not the proteomic level, and kinase network analysis reveals significant heterogeneity between primary colorectal tumors and their liver metastases. In vivo xenograft-based drug tests using 31 primary and metastatic tumors show personalized responses, which could also be predicted by kinase-substrate network analysis no matter whether tumors carry mutations in the drug-targeted genes. Our study provides a valuable resource for better understanding of mCRC and has potential for clinical application.


Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Genomics/methods , Neoplasm Metastasis/drug therapy , Protein Kinases/genetics , Protein Kinases/metabolism , Proteomics/methods , Animals , Antineoplastic Agents/pharmacology , China , Cohort Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Molecular Targeted Therapy , Neoplasm Metastasis/genetics , Phosphorylation , Precision Medicine , Prognosis , Protein Kinases/pharmacology , Xenograft Model Antitumor Assays
8.
EBioMedicine ; 28: 234-240, 2018 Feb.
Article En | MEDLINE | ID: mdl-29422288

Roux-en-Y Gastric bypass surgery (RYGB) is emerging as a powerful tool for treatment of obesity and may also cause remission of type 2 diabetes. However, the molecular mechanism of RYGB leading to diabetes remission independent of weight loss remains elusive. In this study, we profiled plasma metabolites and proteins of 10 normal glucose-tolerant obese (NO) and 9 diabetic obese (DO) patients before and 1-week, 3-months, 1-year after RYGB. 146 proteins and 128 metabolites from both NO and DO groups at all four stages were selected for further analysis. By analyzing a set of bi-molecular associations among the corresponding network of the subjects with our newly developed computational method, we defined the represented physiological states (called the edge-states that reflect the interactions among the bio-molecules), and the related molecular networks of NO and DO patients, respectively. The principal component analyses (PCA) revealed that the edge states of the post-RYGB NO subjects were significantly different from those of the post-RYGB DO patients. Particularly, the time-dependent changes of the molecular hub-networks differed between DO and NO groups after RYGB. In conclusion, by developing molecular network-based systems signatures, we for the first time reveal that RYGB generates a unique path for diabetes remission independent of weight loss.


Diabetes Mellitus, Type 2/surgery , Systems Biology , Blood Proteins/metabolism , Diabetes Mellitus, Type 2/blood , Gastric Bypass , Gene Regulatory Networks , Humans , Metabolome , Obesity/genetics , Principal Component Analysis , Weight Loss
...