Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
Nat Commun ; 13(1): 141, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013146

ABSTRACT

Prostate cancer is the second most common malignancy in men worldwide and consists of a mixture of tumor and non-tumor cell types. To characterize the prostate cancer tumor microenvironment, we perform single-cell RNA-sequencing on prostate biopsies, prostatectomy specimens, and patient-derived organoids from localized prostate cancer patients. We uncover heterogeneous cellular states in prostate epithelial cells marked by high androgen signaling states that are enriched in prostate cancer and identify a population of tumor-associated club cells that may be associated with prostate carcinogenesis. ERG-negative tumor cells, compared to ERG-positive cells, demonstrate shared heterogeneity with surrounding luminal epithelial cells and appear to give rise to common tumor microenvironment responses. Finally, we show that prostate epithelial organoids harbor tumor-associated epithelial cell states and are enriched with distinct cell types and states from their parent tissues. Our results provide diagnostically relevant insights and advance our understanding of the cellular states associated with prostate carcinogenesis.


Subject(s)
Carcinogenesis/genetics , Epithelial Cells/metabolism , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , Tumor Microenvironment/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Lineage/genetics , Epithelial Cells/classification , Epithelial Cells/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , Genetic Heterogeneity , Humans , Male , Molecular Sequence Annotation , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Organoids/metabolism , Organoids/pathology , Primary Cell Culture , Prostate/metabolism , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Signal Transduction , Single-Cell Analysis/methods , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism
2.
Nat Commun ; 13(1): 116, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013227

ABSTRACT

Glioblastoma is an aggressive form of brain cancer with well-established patterns of intra-tumoral heterogeneity implicated in treatment resistance and progression. While regional and single cell transcriptomic variations of glioblastoma have been recently resolved, downstream phenotype-level proteomic programs have yet to be assigned across glioblastoma's hallmark histomorphologic niches. Here, we leverage mass spectrometry to spatially align abundance levels of 4,794 proteins to distinct histologic patterns across 20 patients and propose diverse molecular programs operational within these regional tumor compartments. Using machine learning, we overlay concordant transcriptional information, and define two distinct proteogenomic programs, MYC- and KRAS-axis hereon, that cooperate with hypoxia to produce a tri-dimensional model of intra-tumoral heterogeneity. Moreover, we highlight differential drug sensitivities and relative chemoresistance in glioblastoma cell lines with enhanced KRAS programs. Importantly, these pharmacological differences are less pronounced in transcriptional glioblastoma subgroups suggesting that this model may provide insights for targeting heterogeneity and overcoming therapy resistance.


Subject(s)
Brain Neoplasms/genetics , Genetic Heterogeneity , Glioblastoma/genetics , Hypoxia/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Antineoplastic Agents/therapeutic use , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Cell Line, Tumor , Cohort Studies , Disease Progression , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnosis , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Hypoxia/diagnosis , Hypoxia/drug therapy , Hypoxia/mortality , Laser Capture Microdissection , Machine Learning , Models, Genetic , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Survival Analysis , Transcriptome
3.
Nat Commun ; 13(1): 178, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013316

ABSTRACT

Cancer driving mutations are difficult to identify especially in the non-coding part of the genome. Here, we present sigDriver, an algorithm dedicated to call driver mutations. Using 3813 whole-genome sequenced tumors from International Cancer Genome Consortium, The Cancer Genome Atlas Program, and a childhood pan-cancer cohort, we employ mutational signatures based on single-base substitution in the context of tri- and penta-nucleotide motifs for hotspot discovery. Knowledge-based annotations on mutational hotspots reveal enrichment in coding regions and regulatory elements for 6 mutational signatures, including APOBEC and somatic hypermutation signatures. APOBEC activity is associated with 32 hotspots of which 11 are known and 11 are putative regulatory drivers. Somatic single nucleotide variants clusters detected at hypermutation-associated hotspots are distinct from translocation or gene amplifications. Patients carrying APOBEC induced PIK3CA driver mutations show lower occurrence of signature SBS39. In summary, sigDriver uncovers mutational processes associated with known and putative tumor drivers and hotspots particularly in the non-coding regions of the genome.


Subject(s)
APOBEC Deaminases/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , DNA, Intergenic/genetics , Gene Drive Technology , Neoplasm Proteins/genetics , Neoplasms/genetics , APOBEC Deaminases/metabolism , Atlases as Topic , Child , Class I Phosphatidylinositol 3-Kinases/metabolism , DNA, Intergenic/metabolism , Databases, Genetic , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Mutagenesis , Mutation Rate , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Nucleotide Motifs , Open Reading Frames
4.
Nucleic Acids Res ; 50(D1): D413-D420, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34570220

ABSTRACT

LncRNAs are not only well-known as non-coding elements, but also serve as templates for peptide translation, playing important roles in fundamental cellular processes and diseases. Here, we describe a database, TransLnc (http://bio-bigdata.hrbmu.edu.cn/TransLnc/), which aims to provide comprehensive experimentally supported and predicted lncRNA peptides in multiple species. TransLnc currently documents approximate 583 840 peptides encoded by 33 094 lncRNAs. Six types of direct and indirect evidences supporting the coding potential of lncRNAs were integrated, and 65.28% peptides entries were with at least one type of evidence. Considering the strong tissue-specific expression of lncRNAs, TransLnc allows users to access lncRNA peptides in any of the 34 tissues involved in. In addition, both the unique characteristic and homology relationship were also predicted and provided. Importantly, TransLnc provides computationally predicted tumour neoantigens from peptides encoded by lncRNAs, which would provide novel insights into cancer immunotherapy. There were 220 791 and 237 915 candidate neoantigens binding by major histocompatibility complex (MHC) class I or II molecules, respectively. Several flexible tools were developed to aid retrieve and analyse, particularly lncRNAs tissue expression patterns, clinical relevance across cancer types. TransLnc will serve as a valuable resource for investigating the translation capacity of lncRNAs and greatly extends the cancer immunopeptidome.


Subject(s)
Databases, Genetic , Neoplasms/genetics , Peptides/genetics , Protein Biosynthesis , RNA, Long Noncoding/genetics , Software , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Binding Sites , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunotherapy/methods , Internet , Mice , Molecular Sequence Annotation , Neoplasm Proteins/classification , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Organ Specificity , Peptides/classification , Peptides/immunology , Protein Binding , RNA, Long Noncoding/classification , RNA, Long Noncoding/immunology , Rats
5.
Nat Commun ; 12(1): 5961, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34645806

ABSTRACT

Mutations play a fundamental role in the development of cancer, and many create targetable vulnerabilities. There are both public health and basic science benefits from the determination of the proportion of all cancer cases within a population that include a mutant form of a gene. Here, we provide the first such estimates by combining genomic and epidemiological data. We estimate KRAS is mutated in only 11% of all cancers, which is less than PIK3CA (13%) and marginally higher than BRAF (8%). TP53 is the most commonly mutated gene (35%), and KMT2C, KMT2D, and ARID1A are among the ten most commonly mutated driver genes, highlighting the role of epigenetic dysregulation in cancer. Analysis of major cancer subclassifications highlighted varying dependencies upon individual cancer drivers. Overall, we find that cancer genetics is less dominated by high-frequency, high-profile cancer driver genes than studies limited to a subset of cancer types have suggested.


Subject(s)
Epigenesis, Genetic , Mutation Rate , Neoplasm Proteins/genetics , Neoplasms/epidemiology , Neoplasms/genetics , Computational Biology/methods , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genetics, Population , Humans , Incidence , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neoplasms/classification , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Terminology as Topic , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , United States/epidemiology
6.
Nat Commun ; 12(1): 5291, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489433

ABSTRACT

Esophageal squamous-cell carcinoma (ESCC), one of the most prevalent and lethal malignant disease, has a complex but unknown tumor ecosystem. Here, we investigate the composition of ESCC tumors based on 208,659 single-cell transcriptomes derived from 60 individuals. We identify 8 common expression programs from malignant epithelial cells and discover 42 cell types, including 26 immune cell and 16 nonimmune stromal cell subtypes in the tumor microenvironment (TME), and analyse the interactions between cancer cells and other cells and the interactions among different cell types in the TME. Moreover, we link the cancer cell transcriptomes to the somatic mutations and identify several markers significantly associated with patients' survival, which may be relevant to precision care of ESCC patients. These results reveal the immunosuppressive status in the ESCC TME and further our understanding of ESCC.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Neoplasm Proteins/genetics , Stromal Cells/immunology , Transcription, Genetic , Adult , Aged , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Female , Fibroblasts/immunology , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Myeloid Cells/immunology , Myeloid Cells/pathology , Neoplasm Proteins/classification , Neoplasm Proteins/immunology , Prognosis , Single-Cell Analysis , Stromal Cells/pathology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Whole Genome Sequencing
7.
Nat Commun ; 12(1): 5309, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493726

ABSTRACT

Childhood neuroblastoma has a remarkable variability in outcome. Age at diagnosis is one of the most important prognostic factors, with children less than 1 year old having favorable outcomes. Here we study single-cell and single-nuclei transcriptomes of neuroblastoma with different clinical risk groups and stages, including healthy adrenal gland. We compare tumor cell populations with embryonic mouse sympatho-adrenal derivatives, and post-natal human adrenal gland. We provide evidence that low and high-risk neuroblastoma have different cell identities, representing two disease entities. Low-risk neuroblastoma presents a transcriptome that resembles sympatho- and chromaffin cells, whereas malignant cells enriched in high-risk neuroblastoma resembles a subtype of TRKB+ cholinergic progenitor population identified in human post-natal gland. Analyses of these populations reveal different gene expression programs for worst and better survival in correlation with age at diagnosis. Our findings reveal two cellular identities and a composition of human neuroblastoma tumors reflecting clinical heterogeneity and outcome.


Subject(s)
Adrenal Gland Neoplasms/genetics , Adrenal Glands/metabolism , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Neuroblastoma/genetics , Receptor, trkB/genetics , Transcriptome , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/mortality , Adrenal Gland Neoplasms/pathology , Adrenal Glands/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Nucleus/genetics , Cell Nucleus/metabolism , Child, Preschool , Chromaffin Cells/metabolism , Chromaffin Cells/pathology , Early Diagnosis , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Male , Membrane Glycoproteins/metabolism , Mice , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Receptor, trkB/metabolism , Risk Assessment , Single-Cell Analysis , Species Specificity , Survival Analysis
8.
Bioengineered ; 12(1): 4054-4069, 2021 12.
Article in English | MEDLINE | ID: mdl-34369278

ABSTRACT

During the pandemic of the coronavirus disease 2019, there exist quite a few studies on angiotensin-converting enzyme 2 (ACE2) and SARS-CoV-2 infection, while little is known about ACE2 in hepatocellular carcinoma (HCC). The detailed mechanism among ACE2 and HCC still remains unclear, which needs to be further investigated. In the current study with a total of 6,926 samples, ACE2 expression was downregulated in HCC compared with non-HCC samples (standardized mean difference = -0.41). With the area under the curve of summary receiver operating characteristic = 0.82, ACE2 expression showed a better ability to differentiate HCC from non-HCC. The mRNA expression of ACE2 was related to the age, alpha-fetoprotein levels and cirrhosis of HCC patients, and it was identified as a protected factor for HCC patients via Kaplan-Meier survival, Cox regression analyses. The potential molecular mechanism of ACE2 may be relevant to catabolic and cell division. In all, decreasing ACE2 expression can be seen in HCC, and its protective role for HCC patients and underlying mechanisms were explored in the study.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Carcinoma, Hepatocellular/genetics , Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Neoplasm Proteins/genetics , Receptors, Virus/genetics , alpha-Fetoproteins/genetics , Age Factors , Aged , Angiotensin-Converting Enzyme 2/metabolism , Area Under Curve , COVID-19/virology , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Databases, Genetic , Datasets as Topic , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Cirrhosis/diagnosis , Liver Cirrhosis/mortality , Liver Cirrhosis/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Protective Factors , Protein Interaction Mapping , ROC Curve , Receptors, Virus/metabolism , SARS-CoV-2/pathogenicity , Survival Analysis , alpha-Fetoproteins/metabolism
9.
Genes (Basel) ; 12(7)2021 06 29.
Article in English | MEDLINE | ID: mdl-34209514

ABSTRACT

Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of lung cancer. However, their clinical benefit is limited to a minority of patients. To unravel immune-related factors that are predictive of sensitivity or resistance to immunotherapy, we performed a gene expression analysis by RNA-Seq using the Oncomine Immuno Response Assay (OIRRA) on a total of 33 advanced NSCLC patients treated with ICI evaluating the expression levels of 365 immune-related genes. We found four genes (CD1C, HLA-DPA1, MMP2, and TLR7) downregulated (p < 0.05) and two genes (IFNB1 and MKI67) upregulated (p < 0.05) in ICI-Responders compared to ICI-Non-Responders. The Bayesian enrichment computational analysis showed a more complex interaction network that involved 10 other genes (IFNA1, TLR4, CD40, TLR2, IL12A, IL12B, TLR9, CD1E, IFNG, and HLA-DPB1) correlated with different functional groups. Five main pathways were identified (FDR < 0.0001). High TLR7 expression levels were significantly associated with a lack of response to immunotherapy (p < 0.0001) and worse outcome in terms of both PFS (p < 0.001) and OS (p = 0.03). The multivariate analysis confirmed TLR7 RNA expression as an independent predictor for both poor PFS (HR = 2.97, 95% CI, 1.16-7.6, p = 0.023) and OS (HR = 2.2, 95% CI, 1-5.08, p = 0.049). In conclusion, a high TLR7 gene expression level was identified as an independent predictor for poor clinical benefits from ICI. These data could have important implications for the development of novel single/combinatorial strategies TLR-mediated for an efficient selection of "individualized" treatments for NSCLC in the era of immunotherapy.


Subject(s)
B7-H1 Antigen/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy , Toll-Like Receptor 7/genetics , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , B7-H1 Antigen/antagonists & inhibitors , Bayes Theorem , Biomarkers, Tumor/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Male , Middle Aged , Neoplasm Proteins/classification , Neoplasm Proteins/genetics , Progression-Free Survival , Treatment Outcome
10.
Lipids Health Dis ; 20(1): 58, 2021 Jun 02.
Article in English | MEDLINE | ID: mdl-34078402

ABSTRACT

BACKGROUND: Pancreatic cancer is the fourth leading cause of cancer deaths in the United States both in females and in males, and is projected to become the second deadliest cancer by 2030. The overall 5-year survival rate remains at around 10%. Cancer metabolism and specifically lipid metabolism plays an important role in pancreatic cancer progression and metastasis. Lipid droplets can not only store and transfer lipids, but also act as molecular messengers, and signaling factors. As lipid droplets are implicated in reprogramming tumor cell metabolism and in invasion and migration of pancreatic cancer cells, we aimed to identify lipid droplet-associated genes as prognostic markers in pancreatic cancer. METHODS: We performed a literature search on review articles related to lipid droplet-associated proteins. To select relevant lipid droplet-associated factors, bioinformatics analysis on the GEPIA platform (data are publicly available) was carried out for selected genes to identify differential expression in pancreatic cancer versus healthy pancreatic tissues. Differentially expressed genes were further analyzed regarding overall survival of pancreatic cancer patients. RESULTS: 65 factors were identified as lipid droplet-associated factors. Bioinformatics analysis of 179 pancreatic cancer samples and 171 normal pancreatic tissue samples on the GEPIA platform identified 39 deferentially expressed genes in pancreatic cancer with 36 up-regulated genes (ACSL3, ACSL4, AGPAT2, BSCL2, CAV1, CAV2, CAVIN1, CES1, CIDEC, DGAT1, DGAT2, FAF2, G0S2, HILPDA, HSD17B11, ICE2, LDAH, LIPE, LPCAT1, LPCAT2, LPIN1, MGLL, NAPA, NCEH1, PCYT1A, PLIN2, PLIN3, RAB5A, RAB7A, RAB8A, RAB18, SNAP23, SQLE, VAPA, VCP, VMP1) and 3 down-regulated genes (FITM1, PLIN4, PLIN5). Among 39 differentially expressed factors, seven up-regulated genes (CAV2, CIDEC, HILPDA, HSD17B11, NCEH1, RAB5A, and SQLE) and two down-regulation genes (BSCL2 and FITM1) were significantly associated with overall survival of pancreatic cancer patients. Multivariate Cox regression analysis identified CAV2 as the only independent prognostic factor. CONCLUSIONS: Through bioinformatics analysis, we identified nine prognostic relevant differentially expressed genes highlighting the role of lipid droplet-associated factors in pancreatic cancer.


Subject(s)
Caveolin 2/genetics , Gene Expression Regulation, Neoplastic , Lipid Droplets/metabolism , Neoplasm Proteins/genetics , Pancreatic Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Caveolin 2/metabolism , Computational Biology/methods , Female , Gene Expression Profiling , Humans , Lipid Droplets/chemistry , Lipid Metabolism/genetics , Male , Neoplasm Invasiveness , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Signal Transduction , Survival Analysis , Pancreatic Neoplasms
11.
Gene ; 792: 145728, 2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34022297

ABSTRACT

TNBC is the most aggressive and hormone receptor-negative subtype of breast cancer with molecular heterogeneity in bulk tumors hindering effective treatment. Toll-like receptors (TLRs) have the potential to ignite diverse immune responses in the tumor microenvironment (TME). This encouraged us to screen their transcript expression in the publically available TCGA datasets. Reported molecular subtypes of TNBC may represent different TMEs and we observed differentially expressed TLRs (DETs) i.e. TLR3/4/6/8/9 have unique expression pattern in the TNBC subtypes, particularly in Immunomodulatory (IM) TNBC subtype. We then dissected expression of the DETs in immune and other components of the TME. TLR4 and TLR8 showed significant (p-value ≤ 0.05) negative partial correlation with tumor purity compared to other DETs. Interestingly, TLR4 and TLR8 expression showed a significant (adjusted p-value ≤ 0.05) correlation with different subsets of immune infiltrating cells having the highest correlation with monocytes/macrophage/dendritic cell populations mediating both innate and adaptive response in TNBC. The co-expression network identified genes correlated with these immune cells. Further, GSEA analysis of co-expressed genes showed a significant association of TLR8 partners with 'Peptide ligand binding', 'Gά-signaling', and 'Cytokine-cytokine interaction' while TLR4 associated genes correlated with 'Adaptive immune system' and 'Systemic lupus erythematosus' interactome. Finally, the expression of TLR4 protein was validated in a panel of TNBC cell lines. TLR4 expression in chemoresponsive TNBC was also validated in TNBC cell lines upon Paclitaxel (PTX) treatment. Collectively, the present study identified specific DETs in TNBC and discovered a prospective role of TLR4 and TLR8 in the maintenance of tumor-immune-microenvironment.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphocytes, Tumor-Infiltrating/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 8/genetics , Triple Negative Breast Neoplasms/genetics , Tumor Microenvironment/genetics , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Computational Biology/methods , Databases, Factual , Female , Gene Expression Profiling , Gene Ontology , Gene Regulatory Networks , Humans , Lymphocytes, Tumor-Infiltrating/classification , Lymphocytes, Tumor-Infiltrating/pathology , Molecular Sequence Annotation , Neoplasm Proteins/classification , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Paclitaxel/therapeutic use , Signal Transduction , Toll-Like Receptor 4/immunology , Toll-Like Receptor 8/immunology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/mortality , Tumor Microenvironment/immunology
12.
Biochem J ; 478(9): 1809-1825, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33988704

ABSTRACT

Epithelial plasticity involved the terminal and transitional stages that occur during epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET), both are essential at different stages of early embryonic development that have been co-opted by cancer cells to undergo tumor metastasis. These processes are regulated at multiple instances, whereas the post-transcriptional regulation of key genes mediated by microRNAs is gaining major attention as a common and conserved pathway. In this review, we focus on discussing the latest findings of the cellular and molecular basis of the less characterized process of MET during embryonic development, with special attention to the role of microRNAs. Although we take in consideration the necessity of being cautious when extrapolating the obtained evidence, we propose some commonalities between early embryonic development and cancer progression that can shed light into our current understanding of this complex event and might aid in the design of specific therapeutic approaches.


Subject(s)
Embryonic Development/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Disease Progression , Embryo, Mammalian , Gene Expression Regulation, Neoplastic , Germ Layers/cytology , Germ Layers/growth & development , Germ Layers/metabolism , Humans , MicroRNAs/classification , MicroRNAs/metabolism , Neoplasm Metastasis , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Somites/cytology , Somites/growth & development , Somites/metabolism
13.
Molecules ; 26(8)2021 Apr 11.
Article in English | MEDLINE | ID: mdl-33920405

ABSTRACT

The bioassay-guided fractionation of a CHCl3-MeOH extract from the stems of Cissus trifoliata identified an active fraction against PC3 prostate cancer cells. The treatment for 24 h showed an 80% reduction in cell viability (p ≤ 0.05) by a WST-1 assay at a concentration of 100 µg/mL. The HPLC-QTOF-MS analysis of the fraction showed the presence of coumaric and isoferulic acids, apigenin, kaempferol, chrysoeriol, naringenin, ursolic and betulinic acids, hexadecadienoic and octadecadienoic fatty acids, and the stilbene resveratrol. The exposure of PC3 cells to resveratrol (IC25 = 23 µg/mL) for 24 h induced significant changes in 847 genes (Z-score ≥ ±2). The functional classification tool of the DAVID v6.8 platform indicates that the underlying molecular mechanisms against the proliferation of PC3 cells were associated (p ≤ 0.05) with the process of differentiation and metabolism. These findings provide experimental evidence suggesting the potential of C. trifoliata as a promising natural source of anticancer compounds.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Cell Proliferation/drug effects , Cissus/chemistry , Neoplasm Proteins/genetics , Transcriptome , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Apigenin/chemistry , Apigenin/isolation & purification , Apigenin/pharmacology , Biological Assay , Cell Survival/drug effects , Flavanones/chemistry , Flavanones/isolation & purification , Flavanones/pharmacology , Flavones/chemistry , Flavones/isolation & purification , Flavones/pharmacology , Gene Expression Profiling , Humans , Kaempferols/chemistry , Kaempferols/isolation & purification , Kaempferols/pharmacology , Male , Microarray Analysis , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , PC-3 Cells , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/isolation & purification , Pentacyclic Triterpenes/pharmacology , Plant Extracts/chemistry , Resveratrol/chemistry , Resveratrol/isolation & purification , Resveratrol/pharmacology , Betulinic Acid
14.
J Endocrinol Invest ; 44(11): 2375-2386, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33646556

ABSTRACT

BACKGROUND: This study aimed to identify the potential circulating biomarkers of protein, mRNAs, and long non-coding RNAs (lncRNAs) to differentiate the papillary thyroid cancers from benign thyroid tumors. METHODS: The study population of 100 patients was classified into identification (10 patients with papillary thyroid cancers and 10 patients with benign thyroid tumors) and validation groups (45 patients with papillary thyroid cancers and 35 patients with benign thyroid tumors). The Sengenics Immunome Protein Array-combined data mining approach using the Open Targets Platform was used to identify the putative protein biomarkers, and their expression validated using the enzyme-linked immunosorbent assay. Next-generation sequencing by Illumina HiSeq was used for the detection of dysregulated mRNAs and lncRNAs. The website Timer v2.0 helped identify the putative mRNA biomarkers, which were significantly over-expressed in papillary thyroid cancers than in adjacent normal thyroid tissue. The mRNA and lncRNA biomarker expression was validated by a real-time polymerase chain reaction. RESULTS: Although putative protein and mRNA biomarkers have been identified, their serum expression could not be confirmed in the validation cohorts. In addition, seven lncRNAs (TCONS_00516490, TCONS_00336559, TCONS_00311568, TCONS_00321917, TCONS_00336522, TCONS_00282483, and TCONS_00494326) were identified and validated as significantly downregulated in patients with papillary thyroid cancers compared to those with benign thyroid tumors. These seven lncRNAs showed moderate accuracy based on the area under the curve (AUC = 0.736) of receiver operating characteristic in predicting the occurrence of papillary thyroid cancers. CONCLUSIONS: We identified seven downregulated circulating lncRNAs with the potential for predicting the occurrence of papillary thyroid cancers.


Subject(s)
Neoplasm Proteins , Neoplasms , RNA, Long Noncoding/blood , Thyroid Cancer, Papillary , Thyroid Neoplasms , Area Under Curve , Biomarkers, Tumor/blood , Biomarkers, Tumor/classification , Cell-Free Nucleic Acids/blood , Diagnosis, Differential , Down-Regulation , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Neoplasm Proteins/blood , Neoplasm Proteins/classification , Neoplasms/blood , Neoplasms/diagnosis , Predictive Value of Tests , Thyroid Cancer, Papillary/blood , Thyroid Cancer, Papillary/diagnosis , Thyroid Neoplasms/blood , Thyroid Neoplasms/diagnosis
15.
Nat Genet ; 53(4): 529-538, 2021 04.
Article in English | MEDLINE | ID: mdl-33753930

ABSTRACT

Exciting therapeutic targets are emerging from CRISPR-based screens of high mutational-burden adult cancers. A key question, however, is whether functional genomic approaches will yield new targets in pediatric cancers, known for remarkably few mutations, which often encode proteins considered challenging drug targets. To address this, we created a first-generation pediatric cancer dependency map representing 13 pediatric solid and brain tumor types. Eighty-two pediatric cancer cell lines were subjected to genome-scale CRISPR-Cas9 loss-of-function screening to identify genes required for cell survival. In contrast to the finding that pediatric cancers harbor fewer somatic mutations, we found a similar complexity of genetic dependencies in pediatric cancer cell lines compared to that in adult models. Findings from the pediatric cancer dependency map provide preclinical support for ongoing precision medicine clinical trials. The vulnerabilities observed in pediatric cancers were often distinct from those in adult cancer, indicating that repurposing adult oncology drugs will be insufficient to address childhood cancers.


Subject(s)
Chromosome Mapping/methods , Gene Expression Regulation, Neoplastic , Genome, Human , Mutation , Neoplasm Proteins/genetics , Neoplasms/genetics , Adult , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Child , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
16.
Sci Rep ; 11(1): 3128, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542382

ABSTRACT

Computational approaches to predict drug sensitivity can promote precision anticancer therapeutics. Generalizable and explainable models are of critical importance for translation to guide personalized treatment and are often overlooked in favor of prediction performance. Here, we propose PathDSP: a pathway-based model for drug sensitivity prediction that integrates chemical structure information with enrichment of cancer signaling pathways across drug-associated genes, gene expression, mutation and copy number variation data to predict drug response on the Genomics of Drug Sensitivity in Cancer dataset. Using a deep neural network, we outperform state-of-the-art deep learning models, while demonstrating good generalizability a separate dataset of the Cancer Cell Line Encyclopedia as well as provide explainable results, demonstrated through case studies that are in line with current knowledge. Additionally, our pathway-based model achieved a good performance when predicting unseen drugs and cells, with potential utility for drug development and for guiding individualized medicine.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Drugs, Investigational/therapeutic use , Metabolic Networks and Pathways/genetics , Neoplasm Proteins/genetics , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Cell Line, Tumor , DNA Copy Number Variations , Datasets as Topic , Drug Resistance, Neoplasm/drug effects , Drugs, Investigational/chemistry , Gene Expression Regulation, Neoplastic , Humans , Metabolic Networks and Pathways/drug effects , Mutation , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neural Networks, Computer , Precision Medicine/methods , Signal Transduction
17.
Sci Rep ; 11(1): 3176, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542435

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype, lacking effective therapy. Many TNBCs show remarkable response to carboplatin-based chemotherapy, but often develop resistance over time. With increasing use of carboplatin in the clinic, there is a pressing need to identify vulnerabilities of carboplatin-resistant tumors. In this study, we generated carboplatin-resistant TNBC MDA-MB-468 cell line and patient derived TNBC xenograft models. Mass spectrometry-based proteome profiling demonstrated that carboplatin resistance in TNBC is linked to drastic metabolism rewiring and upregulation of anti-oxidative response that supports cell replication by maintaining low levels of DNA damage in the presence of carboplatin. Carboplatin-resistant cells also exhibited dysregulation of the mitotic checkpoint. A kinome shRNA screen revealed that carboplatin-resistant cells are vulnerable to the depletion of the mitotic checkpoint regulators, whereas the checkpoint kinases CHEK1 and WEE1 are indispensable for the survival of carboplatin-resistant cells in the presence of carboplatin. We confirmed that pharmacological inhibition of CHEK1 by prexasertib in the presence of carboplatin is well tolerated by mice and suppresses the growth of carboplatin-resistant TNBC xenografts. Thus, abrogation of the mitotic checkpoint by CHEK1 inhibition re-sensitizes carboplatin-resistant TNBCs to carboplatin and represents a potential strategy for the treatment of carboplatin-resistant TNBCs.


Subject(s)
Carboplatin/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Checkpoint Kinase 1/genetics , Drug Resistance, Neoplasm/drug effects , Protein-Tyrosine Kinases/genetics , Pyrazines/pharmacology , Pyrazoles/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1/metabolism , DNA Damage , Drug Resistance, Neoplasm/genetics , Drug Synergism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Proteins/classification , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteome/classification , Proteome/genetics , Proteome/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
18.
Curr Cancer Drug Targets ; 21(4): 274-282, 2021.
Article in English | MEDLINE | ID: mdl-33413063

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is the most common histological subtype of non-Hodgkin B cell lymphoma (NHL), and manifests highly heterogeneous genetic/phenotypic characteristics as well as variable responses to conventional immunochemotherapy. Genetic profiling of DLBCL patients has revealed highly recurrent mutations of epigenetic regulator genes such as CREBBP, KMT2D, EZH2 and TET2. These mutations drive malignant transformation through aberrant epigenetic programming of B-cells and may influence clinical outcomes. These and other chromatin modifier genes also play critical roles in normal B-cells, as they undergo the various phenotypic transitions characteristic of the humoral immune response. Many of these functions have to do with impairing immune surveillance and may critically mediate resistance to immunotherapies. In this review, we describe how epigenetic dysfunction induces lymphomagenesis and discuss ways of implementing precision epigenetic therapies to reverse these immune resistant phenotypes.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Drug Resistance, Neoplasm/genetics , Lymphoma, Large B-Cell, Diffuse , Neoplasm Proteins , Epigenesis, Genetic , Genetic Heterogeneity , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Mutation , Neoplasm Proteins/classification , Neoplasm Proteins/genetics
19.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33406529

ABSTRACT

Glioblastoma (GBM) is a common malignant brain tumor which often presents as a comorbidity with central nervous system (CNS) disorders. Both CNS disorders and GBM cells release glutamate and show an abnormality, but differ in cellular behavior. So, their etiology is not well understood, nor is it clear how CNS disorders influence GBM behavior or growth. This led us to employ a quantitative analytical framework to unravel shared differentially expressed genes (DEGs) and cell signaling pathways that could link CNS disorders and GBM using datasets acquired from the Gene Expression Omnibus database (GEO) and The Cancer Genome Atlas (TCGA) datasets where normal tissue and disease-affected tissue were examined. After identifying DEGs, we identified disease-gene association networks and signaling pathways and performed gene ontology (GO) analyses as well as hub protein identifications to predict the roles of these DEGs. We expanded our study to determine the significant genes that may play a role in GBM progression and the survival of the GBM patients by exploiting clinical and genetic factors using the Cox Proportional Hazard Model and the Kaplan-Meier estimator. In this study, 177 DEGs with 129 upregulated and 48 downregulated genes were identified. Our findings indicate new ways that CNS disorders may influence the incidence of GBM progression, growth or establishment and may also function as biomarkers for GBM prognosis and potential targets for therapies. Our comparison with gold standard databases also provides further proof to support the connection of our identified biomarkers in the pathology underlying the GBM progression.


Subject(s)
Brain Neoplasms/genetics , Central Nervous System/metabolism , Gene Regulatory Networks , Glioblastoma/genetics , Machine Learning , Neoplasm Proteins/genetics , Atlases as Topic , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Central Nervous System/pathology , Computational Biology/methods , Datasets as Topic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , Glioblastoma/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Glutamic Acid/metabolism , Humans , Kaplan-Meier Estimate , Molecular Sequence Annotation , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Proportional Hazards Models , Signal Transduction
20.
Protein J ; 40(1): 108-118, 2021 02.
Article in English | MEDLINE | ID: mdl-33387250

ABSTRACT

Patients with triple-negative breast cancer (TNBC) have a relatively poor prognosis and cannot benefit from endocrine and/or targeted therapy. Considerable effort has been devoted toward the elucidation of the molecular mechanisms and potential diagnostic/therapeutic targets. However, it is inefficient and often ineffective to study the biological nuances of TNBC in large-scale clinical trials. In contrast, the investigation of the association between molecular alterations induced through controlled variables and relevant physiochemical characteristics of TNBC cells in laboratory settings is simple, definite, and efficient in exploring the molecular mechanisms. In this study, microgravity was selected as the sole variable of study as it can inhibit cancer cell viability, proliferation, metastasis, and chemoresistance. Identifying the key molecules that shift cancer cells toward a less aggressive phenotype may facilitate future TNBC studies. We focused on extracellular vesicles (EV) derived from TNBC MDA-MB-231 cells in microgravity, which mediate intercellular communication by transporting signaling molecules between cells. Our results show that in comparison with cells in full gravity, EV release rate decreased in microgravity while average EV size increased. In addition, we found EVs may be superior to cells in analyzing differentially expressed proteins, especially those that are down-regulated ones and usually unidentified or neglected in analysis of intact cellular contents. Proteomic analysis of both EVs and cells further revealed a significant correlation with GTPases and proliferation of MDA-MB-231 cells in microgravity. Altogether, our findings would further inspire in-depth correlative cancer biological studies and subsequent clinical research.


Subject(s)
Cell Communication/genetics , Epithelial Cells/metabolism , Extracellular Vesicles/metabolism , GTP Phosphohydrolases/genetics , Neoplasm Proteins/genetics , Weightlessness Simulation/methods , Biological Transport , Cell Line, Tumor , Cell Proliferation , Epithelial Cells/pathology , Extracellular Vesicles/chemistry , GTP Phosphohydrolases/classification , GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Molecular Sequence Annotation , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Proteomics/methods , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...