Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.115
1.
Clin Transl Med ; 14(5): e1670, 2024 May.
Article En | MEDLINE | ID: mdl-38689429

BACKGROUND: Treatment for osteosarcoma, a paediatric bone cancer with no therapeutic advances in over three decades, is limited by a lack of targeted therapies. Osteosarcoma frequently metastasises to the lungs, and only 20% of patients survive 5 years after the diagnosis of metastatic disease. We found that WNT5B is the most abundant WNT expressed in osteosarcoma tumours and its expression correlates with metastasis, histologic subtype and reduced survival. METHODS: Using tumor-spheroids to model cancer stem-like cells, we performed qPCR, immunoblotting, and immunofluorescence to monitor changes in gene and protein expression. Additionally, we measured sphere size, migration and forming efficiency to monitor phenotypic changes. Therefore, we characterised WNT5B's relevance to cancer stem-like cells, metastasis, and chemoresistance and evaluated its potential as a therapeutic target. RESULTS: In osteosarcoma cell lines and patient-derived spheres, WNT5B is enriched in stem cells and induces the expression of the stemness gene SOX2. WNT5B promotes sphere size, sphere-forming efficiency, and cell proliferation, migration, and chemoresistance to methotrexate (but not cisplatin or doxorubicin) in spheres formed from conventional cell lines and patient-derived xenografts. In vivo, WNT5B increased osteosarcoma lung and liver metastasis and inhibited the glycosaminoglycan hyaluronic acid via upregulation of hyaluronidase 1 (HYAL1), leading to changes in the tumour microenvironment. Further, we identified that WNT5B mRNA and protein correlate with the receptor ROR1 in primary tumours. Targeting WNT5B through inhibition of WNT/ROR1 signalling with an antibody to ROR1 reduced stemness properties, including chemoresistance, sphere size and SOX2 expression. CONCLUSIONS: Together, these data define WNT5B's role in driving osteosarcoma cancer stem cell expansion and methotrexate resistance and provide evidence that the WNT5B pathway is a promising candidate for treating osteosarcoma patients. KEY POINTS: WNT5B expression is high in osteosarcoma stem cells leading to increased stem cell proliferation and migration through SOX2. WNT5B expression in stem cells increases rates of osteosarcoma metastasis to the lungs and liver in vivo. The hyaluronic acid degradation enzyme HYAL1 is regulated by WNT5B in osteosarcoma contributing to metastasis. Inhibition of WNT5B with a ROR1 antibody decreases osteosarcoma stemness.


Drug Resistance, Neoplasm , Osteosarcoma , Wnt Proteins , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Humans , Drug Resistance, Neoplasm/genetics , Wnt Proteins/metabolism , Wnt Proteins/genetics , Animals , Mice , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/drug therapy , Neoplasm Metastasis/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/drug effects , Cell Line, Tumor
2.
Nature ; 629(8012): 543-554, 2024 May.
Article En | MEDLINE | ID: mdl-38750233

Metastasis is a multistep process by which cancer cells break away from their original location and spread to distant organs, and is responsible for the vast majority of cancer-related deaths. Preventing early metastatic dissemination would revolutionize the ability to fight cancer. Unfortunately, the relatively poor understanding of the molecular underpinnings of metastasis has hampered the development of effective anti-metastatic drugs. Although it is now accepted that disseminating tumour cells need to acquire multiple competencies to face the many obstacles they encounter before reaching their metastatic site(s), whether these competencies are acquired through an accumulation of metastasis-specific genetic alterations and/or non-genetic events is often debated. Here we review a growing body of literature highlighting the importance of both genetic and non-genetic reprogramming events during the metastatic cascade, and discuss how genetic and non-genetic processes act in concert to confer metastatic competencies. We also describe how recent technological advances, and in particular the advent of single-cell multi-omics and barcoding approaches, will help to better elucidate the cross-talk between genetic and non-genetic mechanisms of metastasis and ultimately inform innovative paths for the early detection and interception of this lethal process.


Neoplasm Metastasis , Neoplasms , Humans , Neoplasm Metastasis/genetics , Neoplasms/genetics , Neoplasms/pathology , Animals , Single-Cell Analysis
3.
PLoS One ; 19(4): e0301989, 2024.
Article En | MEDLINE | ID: mdl-38683764

Somatic Y chromosome loss in hematopoietic cells is associated with higher mortality in men. However, the status of the Y chromosome in cancer tissue is not fully known due to technical limitations, such as difficulties in labelling and sequencing DNA from the Y chromosome. We have developed a system to quantify Y chromosome gain or loss in patient-derived prostate cancer organoids. Using our system, we observed Y chromosome loss in 4 of the 13 (31%) patient-derived metastatic castration-resistant prostate cancer (mCRPC) organoids; interestingly, loss of Yq (long arm of the Y chromosome) was seen in 38% of patient-derived organoids. Additionally, potential associations were observed between mCRPC and Y chromosome nullisomy. The prevalence of Y chromosome loss was similar in primary and metastatic tissue, suggesting that Y chromosome loss is an early event in prostate cancer evolution and may not a result of drug resistance or organoid derivation. This study reports quantification of Y chromosome loss and gain in primary and metastatic prostate cancer tissue and lays the groundwork for further studies investigating the clinical relevance of Y chromosome loss or gain in mCRPC.


Chromosome Painting , Chromosomes, Human, Y , Neoplasm Metastasis , Male , Humans , Chromosomes, Human, Y/genetics , Neoplasm Metastasis/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Organoids/pathology , Chromosome Deletion
4.
Acta Neuropathol Commun ; 12(1): 67, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38671536

Melanoma is the most serious type of skin cancer that frequently spreads to other organs of the human body. Especially melanoma metastases to the brain (intracranial metastases) are hard to treat and a major cause of death of melanoma patients. Little is known about molecular alterations and altered mechanisms that distinguish intra- from extracranial melanoma metastases. So far, almost all existing studies compared intracranial metastases from one set of patients to extracranial metastases of an another set of melanoma patients. This neglects the important facts that each melanoma is highly individual and that intra- and extracranial melanoma metastases from the same patient are more similar to each other than to melanoma metastases from other patients in the same organ. To overcome this, we compared the gene expression profiles of 16 intracranial metastases to their corresponding 21 patient-matched extracranial metastases in a personalized way using a three-state Hidden Markov Model (HMM) to identify altered genes for each individual metastasis pair. This enabled three major findings by considering the predicted gene expression alterations across all patients: (i) most frequently altered pathways include cytokine-receptor interaction, calcium signaling, ECM-receptor interaction, cAMP signaling, Jak-STAT and PI3K/Akt signaling, (ii) immune-relevant signaling pathway genes were downregulated in intracranial metastases, and (iii) intracranial metastases were associated with a brain-like phenotype gene expression program. Further, the integration of all differentially expressed genes across the patient-matched melanoma metastasis pairs led to a set of 103 genes that were consistently down- or up-regulated in at least 11 of the 16 of the patients. This set of genes contained many genes involved in the regulation of immune responses, cell growth, cellular signaling and transport processes. An analysis of these genes in the TCGA melanoma cohort showed that the expression behavior of 11 genes was significantly associated with survival. Moreover, a comparison of the 103 genes to three closely related melanoma metastasis studies revealed a core set of eight genes that were consistently down- or upregulated in intra- compared to extracranial metastases in at least two of the three related studies (down: CILP, DPT, FGF7, LAMP3, MEOX2, TMEM119; up: GLDN, PMP2) including FGF7 that was also significantly associated with survival. Our findings contribute to a better characterization of genes and pathways that distinguish intra- from extracranial melanoma metastasis and provide important hints for future experimental studies to identify potential targets for new therapeutic approaches.


Brain Neoplasms , Melanoma , Skin Neoplasms , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/secondary , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Female , Male , Middle Aged , Aged , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Adult , Gene Expression Profiling , Neoplasm Metastasis/genetics
5.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38673800

Clear-cell renal-cell carcinoma (ccRCC) is a silent-development pathology with a high rate of metastasis in patients. The activity of coding genes in metastatic progression is well known. New studies evaluate the association with non-coding genes, such as competitive endogenous RNA (ceRNA). This study aims to build a ceRNA network and a gene signature for ccRCC associated with metastatic development and analyze their biological functions. Using data from The Cancer Genome Atlas (TCGA), we constructed the ceRNA network with differentially expressed genes, assembled nine preliminary gene signatures from eight feature selection techniques, and evaluated the classification metrics to choose a final signature. After that, we performed a genomic analysis, a risk analysis, and a functional annotation analysis. We present an 11-gene signature: SNHG15, AF117829.1, hsa-miR-130a-3p, hsa-mir-381-3p, BTBD11, INSR, HECW2, RFLNB, PTTG1, HMMR, and RASD1. It was possible to assess the generalization of the signature using an external dataset from the International Cancer Genome Consortium (ICGC-RECA), which showed an Area Under the Curve of 81.5%. The genomic analysis identified the signature participants on chromosomes with highly mutated regions. The hsa-miR-130a-3p, AF117829.1, hsa-miR-381-3p, and PTTG1 were significantly related to the patient's survival and metastatic development. Additionally, functional annotation resulted in relevant pathways for tumor development and cell cycle control, such as RNA polymerase II transcription regulation and cell control. The gene signature analysis within the ceRNA network, with literature evidence, suggests that the lncRNAs act as "sponges" upon the microRNAs (miRNAs). Therefore, this gene signature presents coding and non-coding genes and could act as potential biomarkers for a better understanding of ccRCC.


Carcinoma, Renal Cell , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Kidney Neoplasms , Machine Learning , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Biomarkers, Tumor/genetics , Neoplasm Metastasis/genetics , MicroRNAs/genetics , Gene Expression Profiling/methods , Transcriptome , RNA, Competitive Endogenous
6.
Comput Biol Med ; 175: 108496, 2024 Jun.
Article En | MEDLINE | ID: mdl-38657466

Distant metastasis of cancer is a significant contributor to cancer-related complications, and early identification of unidentified stomach adenocarcinoma is crucial for a positive prognosis. Changes inDNA methylation are being increasingly recognized as a crucial factor in predicting cancer progression. Within this research, we developed machine learning and deep learning models for distinguishing distant metastasis in samples of stomach adenocarcinoma based on DNA methylation profile. Employing deep neural networks (DNN), support vector machines (SVM), random forest (RF), Naive Bayes (NB) and decision tree (DT), and models for forecasting distant metastasis in stomach adenocarcinoma. The results show that the performance of DNN is better than that of other models, AUC and AUPR achieving 99.9 % and 99.5 % respectively. Additionally, a weighted random sampling technique was utilized to address the issue of imbalanced datasets, enabling the identification of crucial methylation markers associated with functionally significant genes in stomach distant metastasis tumors with greater performance.


DNA Methylation , Deep Learning , Neoplasm Metastasis , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Humans , DNA Methylation/genetics , Neoplasm Metastasis/genetics , Machine Learning , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Female
7.
Adv Sci (Weinh) ; 11(16): e2308531, 2024 Apr.
Article En | MEDLINE | ID: mdl-38380551

Gallbladder cancer (GBC) is an extremely lethal malignancy with aggressive behaviors, including liver or distant metastasis; however, the underlying mechanisms driving the metastasis of GBC remain poorly understood. In this study, it is found that DNA methyltransferase DNMT3A is highly expressed in GBC tumor tissues compared to matched adjacent normal tissues. Clinicopathological analysis shows that DNMT3A is positively correlated with liver metastasis and poor overall survival outcomes in patients with GBC. Functional analysis confirms that DNMT3A promotes the metastasis of GBC cells in a manner dependent on its DNA methyltransferase activity. Mechanistically, DNMT3A interacts with and is recruited by YAP/TAZ to recognize and access the CpG island within the CDH1 promoter and generates hypermethylation of the CDH1 promoter, which leads to transcriptional silencing of CDH1 and accelerated epithelial-to-mesenchymal transition. Using tissue microarrays, the association between the expression of DNMT3A, YAP/TAZ, and CDH1 is confirmed, which affects the metastatic ability of GBC. These results reveal a novel mechanism through which DNMT3A recruitment by YAP/TAZ guides DNA methylation to drive GBC metastasis and provide insights into the treatment of GBC metastasis by targeting the functional connection between DNMT3A and YAP/TAZ.


DNA Methyltransferase 3A , Gallbladder Neoplasms , Animals , Female , Humans , Male , Mice , Middle Aged , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Antigens, CD , Cadherins , Cell Line, Tumor , Disease Models, Animal , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , DNA Methyltransferase 3A/metabolism , DNA Methyltransferase 3A/genetics , Epithelial-Mesenchymal Transition/genetics , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Metastasis/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics
8.
Genes Cells ; 29(4): 290-300, 2024 Apr.
Article En | MEDLINE | ID: mdl-38339971

Lung cancer frequently metastasizes to the bones. An in vivo model is urgently required to identify potential therapeutic targets for the prevention and treatment of lung cancer with bone metastasis. We established a lung adenocarcinoma cell subline (H322L-BO4) that specifically showed metastasis to the leg bones and adrenal glands. This was achieved by repeated isolation of metastatic cells from the leg bones of mice. The cells were intracardially injected into nude mice. Survival was prolonged for mice that received H322L-BO4 cells versus original cells (H322L). H322L-BO4 cells did not exhibit obvious changes in general in vitro properties associated with the metastatic potential (e.g., cell growth, migration, and invasion) compared with H322L cells. However, the phosphorylation of chromosome 9 open reading frame 10/oxidative stress-associated Src activator (C9orf10/Ossa) was increased in H322L-BO4 cells. This result confirmed the increased anchorage independence through C9orf10/Ossa-mediated activation of Src family tyrosine kinase. Reduction of C9orf10/Ossa by shRNA reduced cells' metastasis to the leg bone and prolonged survival in mice. These findings indicate that H322L-BO4 cells can be used to evaluate the effect of candidate therapeutic targets against bone metastatic lung cancer cells. Moreover, C9orf10/Ossa may be a useful target for treatment of lung cancer with bone metastasis.


Adenocarcinoma of Lung , Bone Neoplasms , Lung Neoplasms , Animals , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Neoplasm Metastasis/genetics , src-Family Kinases/therapeutic use , Humans
9.
Adv Sci (Weinh) ; 11(16): e2306915, 2024 Apr.
Article En | MEDLINE | ID: mdl-38357830

Recent studies suggest that circular RNA (circRNA)-mediated post-translational modification of RNA-binding proteins (RBP) plays a pivotal role in metastasis of hepatocellular carcinoma (HCC). However, the specific mechanism and potential clinical therapeutic significance remain vague. This study attempts to profile the regulatory networks of circRNA and RBP using a multi-omics approach. Has_circ_0006646 (circ0006646) is an unreported circRNA in HCC and is associated with a poor prognosis. Silencing of circ0006646 significantly hinders metastasis in vivo. Mechanistically, circ0006646 prevents the interaction between nucleolin (NCL) and the E3 ligase tripartite motif-containing 21 to reduce the proteasome-mediated degradation of NCL via K48-linked polyubiquitylation. Furthermore, the change of NCL expression is proven to affect the phosphorylation levels of multiple proteins and inhibit p53 translation. Moreover, patient-derived tumor xenograft and lentivirus injection, which is conducted to simulate clinical treatment confirmed the potential therapeutic value. Overall, this study describes the integrated multi-omics landscape of circRNA-mediated NCL ubiquitination degradation in HCC metastasis and provides a novel therapeutic target.


Carcinoma, Hepatocellular , Liver Neoplasms , RNA, Circular , Ubiquitination , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , Ubiquitination/genetics , Mice , Animals , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Cell Line, Tumor , Nucleolin , Neoplasm Metastasis/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Disease Models, Animal , Multiomics
10.
Nature ; 627(8004): 586-593, 2024 Mar.
Article En | MEDLINE | ID: mdl-38355797

Over half of hepatocellular carcinoma (HCC) cases diagnosed worldwide are in China1-3. However, whole-genome analysis of hepatitis B virus (HBV)-associated HCC in Chinese individuals is limited4-8, with current analyses of HCC mainly from non-HBV-enriched populations9,10. Here we initiated the Chinese Liver Cancer Atlas (CLCA) project and performed deep whole-genome sequencing (average depth, 120×) of 494 HCC tumours. We identified 6 coding and 28 non-coding previously undescribed driver candidates. Five previously undescribed mutational signatures were found, including aristolochic-acid-associated indel and doublet base signatures, and a single-base-substitution signature that we termed SBS_H8. Pentanucleotide context analysis and experimental validation confirmed that SBS_H8 was distinct to the aristolochic-acid-associated SBS22. Notably, HBV integrations could take the form of extrachromosomal circular DNA, resulting in elevated copy numbers and gene expression. Our high-depth data also enabled us to characterize subclonal clustered alterations, including chromothripsis, chromoplexy and kataegis, suggesting that these catastrophic events could also occur in late stages of hepatocarcinogenesis. Pathway analysis of all classes of alterations further linked non-coding mutations to dysregulation of liver metabolism. Finally, we performed in vitro and in vivo assays to show that fibrinogen alpha chain (FGA), determined as both a candidate coding and non-coding driver, regulates HCC progression and metastasis. Our CLCA study depicts a detailed genomic landscape and evolutionary history of HCC in Chinese individuals, providing important clinical implications.


Carcinoma, Hepatocellular , Genome, Human , High-Throughput Nucleotide Sequencing , Liver Neoplasms , Mutation , Whole Genome Sequencing , Humans , Aristolochic Acids/metabolism , Carcinogenesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , China , Chromothripsis , Disease Progression , DNA, Circular/genetics , East Asian People/genetics , Evolution, Molecular , Genome, Human/genetics , Hepatitis B virus/genetics , INDEL Mutation/genetics , Liver/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/virology , Mutation/genetics , Neoplasm Metastasis/genetics , Open Reading Frames/genetics , Reproducibility of Results
13.
Exp Cell Res ; 434(1): 113864, 2024 01 01.
Article En | MEDLINE | ID: mdl-38040050

Metastasis is the primary cause of cancer-related deaths and remains poorly understood. Deubiquitinase OTU domain containing 4 (OTUD4) has been reported to regulate antiviral immune responses and resistance to radio- or chemo-therapies in certain cancers. However, the role of OTUD4 in cancer metastasis remain unknown. Here, we demonstrate that the depletion of OTUD4 in triple-negative breast cancer (TNBC) cells markedly suppress cell clonogenic ability, migration, invasion and cancer stem cell population in vitro as well as metastasis in vivo. Mechanistically, the tumor promoting function of OTUD4 is mainly mediated by deuiquitinating and stabilizing Snail1, one key transcriptional factor in the epithelial-mesenchymal transition. The inhibitory effect of targeting OTUD4 could be largely reversed by the reconstitution of Snail1 in OTUD4-deficient cells. Overall, our study establishes the OTUD4-Snail1 axis as an important regulatory mechanism of breast cancer metastasis and provides a rationale for potential therapeutic interventions in the treatment of TNBC.


Snail Family Transcription Factors , Triple Negative Breast Neoplasms , Ubiquitin-Specific Proteases , Ubiquitin-Specific Proteases/metabolism , MDA-MB-231 Cells , HEK293 Cells , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/physiopathology , Neoplasm Metastasis/genetics , Snail Family Transcription Factors/metabolism , Humans , Female , Animals , Mice , Cell Movement/genetics , Neoplasm Invasiveness/genetics , Protein Stability
14.
Gut ; 73(3): 470-484, 2024 Feb 23.
Article En | MEDLINE | ID: mdl-38050068

OBJECTIVE: Metastasis is the major cause of cancer death. However, what types of heterogenous cancer cells in primary tumour and how they metastasise to the target organs remain largely undiscovered. DESIGN: We performed single-cell RNA sequencing and spatial transcriptomic analysis in primary colorectal cancer (CRC) and metastases in the liver (lCRC) or ovary (oCRC). We also conducted immunofluorescence staining and functional experiments to examine the mechanism. RESULTS: Integrative analyses of epithelial cells reveal a stem-like cell cluster with high protein tyrosine phosphatase receptor type O (PTPRO) and achaete scute-like 2 (ASCL2) expression as the metastatic culprit. This cell cluster comprising distinct subpopulations shows distinct liver or ovary metastatic preference. Population 1 (P1) cells with high delta-like ligand 4 (DLL4) and MAF bZIP transcription factor A (MAFA) expression are enriched in primary CRC and oCRC, thus may be associated with ovarian metastasis. P3 cells having a similar expression pattern as cholangiocytes are found mainly in primary CRC and lCRC, presuming to be likely the culprits that specifically metastasise to the liver. Stem-like cells interacted with cancer-associated fibroblasts and endothelial cells via the DLL4-NOTCH signalling pathway to metastasise from primary CRC to the ovary. In the oCRC microenvironment, myofibroblasts provide cancer cells with glutamine and perform a metabolic reprogramming, which may be essential for cancer cells to localise and develop in the ovary. CONCLUSION: We uncover a mechanism for organ-specific CRC metastasis.


Colorectal Neoplasms , Liver Neoplasms , Female , Humans , Colorectal Neoplasms/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Liver Neoplasms/pathology , Gene Expression Profiling , Signal Transduction/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis/genetics , Tumor Microenvironment/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism
15.
Nat Commun ; 14(1): 6569, 2023 10 17.
Article En | MEDLINE | ID: mdl-37848444

While macrophage phagocytosis is an immune defense mechanism against invading cellular organisms, cancer cells expressing the CD47 ligand send forward signals to repel this engulfment. Here we report that the reverse signaling using CD47 as a receptor additionally enhances a pro-survival function of prostate cancer cells under phagocytic attack. Although low CD47-expressing cancer cells still allow phagocytosis, the reverse signaling delays the process, leading to incomplete digestion of the entrapped cells and subsequent tumor hybrid cell (THC) formation. Viable THCs acquire c-Myc from parental cancer cells to upregulate both M1- and M2-like macrophage polarization genes. Consequently, THCs imitating dual macrophage features can confound immunosurveillance, gaining survival advantage in the host. Furthermore, these cells intrinsically express low levels of androgen receptor and its targets, resembling an adenocarcinoma-immune subtype of metastatic castration-resistant prostate cancer. Therefore, phagocytosis-generated THCs may represent a potential target for treating the disease.


CD47 Antigen , Macrophages , Neoplasm Metastasis , Phagocytosis , Proto-Oncogene Proteins c-myc , Tumor Escape , Humans , Male , Carrier Proteins , CD47 Antigen/metabolism , Macrophages/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Signal Transduction , Tumor Escape/genetics , Tumor Escape/immunology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Tumor Cells, Cultured
16.
Oncogene ; 42(40): 2956-2970, 2023 09.
Article En | MEDLINE | ID: mdl-37612524

Lymphatic metastasis is recognized as the leading manner of metastasis in bladder cancer (BLCa), but hematogenous metastasis accounts for a majority of cancer-associated deaths. The past two decades have witnessed tremendous attention in long non-coding RNAs (lncRNAs), which are a new hope for the development of targeted drug therapy for metastatic cancers; however, the underlying mechanism of lncRNAs involved in BLCa hematogenous metastasis remains to be elucidated. Here, we identified BLCa-associated transcript 3 (BLACAT3), a lncRNA, which was aberrantly upregulated in BLCa and corelated with poor prognosis of patients with muscle-invasive bladder cancer. Methodologically, m6A epitranscriptomic microarray, RNA sequencing and mass spectrometry (MS) were used to screen the key molecules of the regulatory axis. Functional assays, animal models and clinical samples were used to explore the roles of BLACAT3 in BLCa in vitro and in vivo. Mechanistically, m6A modification contributes to BLACAT3 upregulation by stabilizing RNA structure. BLACAT3 recruits YBX3 to shuttle into the nucleus, synergistically enhances NCF2 transcription, and promotes BLCa angiogenesis and hematogenous metastasis by activating downstream NF-κB signaling. Our findings will develop prognosis prediction tools for BLCa patients and discover novel therapeutic biological targets for metastatic BLCa.


RNA, Long Noncoding , Urinary Bladder Neoplasms , Animals , Humans , NADPH Oxidases/genetics , NF-kappa B/genetics , RNA, Long Noncoding/genetics , Signal Transduction/genetics , Up-Regulation , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neoplasm Metastasis/genetics
17.
Arch. esp. urol. (Ed. impr.) ; 76(6): 425-438, 28 aug. 2023. ilus, graf
Article En | IBECS | ID: ibc-224895

Background: Based on publicly available transcriptome and single-cell sequencing data, the current study aimed to explore the molecular mechanisms underlying the involvement of hepatocellular carcinoma-derived growth factor-like 3 (HDGFL3) in prostate cancer (PCA) growth and metastasis. Methods: The Gene Expression Omnibus database was used to download the single cell transcriptome of PCA (GSE193337). Single-cell RNA sequencing (scRNA-seq) data were examined to identify which genes are essential for endothelial cell function. The Cancer Genome Atlas Prostate Adenocarcinoma database provided the RNA sequencing data, and univariate COX regression analysis was introduced to identify the genes that were associated with the prognosis of patients with PCA. Human PCA cell lines PC-3 and DU145 were used in in vitro cellular studies to test the effect of silencing HDGFL3. The results were validated using Transwell® assay, scratch assay, and cell counting kit-8 assay. To support the role of HDGFL3 in PCA, an in vivo animal model of PCA transplantation tumor in nude mice was established. Quantitative reverse transcription polymerase chain reaction was introduced to measure HDGFL3 messenger ribonucleic acid (mRNA) expression levels in tumor tissues from nude mice, and Hematoxylin and Eosin staining was used to identify lung metastasis. Immunohistochemical staining was employed to identify the expression levels of HDGFL3 and hematopoietic progenitor cell antigen CD34+. Results: It was discovered through analysis of the scRNA-seq dataset that HDGFL3, a gene specific to endothelial cells, is linked to a poor prognosis in men with PCA. In addition, HDGFL3 and the expression of genes linked to angiogenesis have a substantial association (AU)


Humans , Male , Gene Expression Regulation, Neoplastic , RNA/genetics , Exome Sequencing , Prostatic Neoplasms/genetics , Neoplasm Metastasis/genetics , Transcription Factors
18.
BMC Gastroenterol ; 23(1): 193, 2023 Jun 05.
Article En | MEDLINE | ID: mdl-37277698

BACKGROUND: Advances in molecular biology have improved understanding of the molecular features of carcinogenesis and progression of colorectal cancer. It is clear that the efficacy of anti-EGFR depends upon the RAS mutational status, since any mutation in RAS is associated with resistance to anti-EGFR therapy. The aim of this study is to report the largest North African description of KRAS and NRAS status in metastatic colorectal cancer and to describe the association of these mutations with clinicopathological characteristics. METHODS: This is a prospective study of all consecutive unselected metastatic colorectal cancer samples, collected from the Laboratory of Pathology at the National Institute of Oncology of Rabat, Morocco, from January 1st 2020 to December 31st 2021. The molecular analysis was performed on the Idylla™ platform (fully automated real-time polymerase chain reaction-based assay) for KRAS and NRAS mutations in exons 2, 3 and 4. These mutations were correlated to gender, primary tumor site, histological type and degree of differentiation of tumor using adequate statistical methods. RESULTS: Four hundred fourteen colorectal tumors were screened for KRAS and NRAS mutations. These mutations occurred in 51.7% of tumors for KRAS (mainly in exon 12) and in 3% of tumors for NRAS. There was a significant correlation between NRAS mutation and age of colorectal patients in this study. The low rate of invalid RAS tests (1.7% for KRAS and 3.1% for NRAS) was certainly obtained due to the strict respect of pre-analytical factors such as cold ischemia time and formalin fixation. CONCLUSION: We report the largest North African analysis of NRAS and KRAS status in colorectal metastatic patients. This study showed the ability in low middle income countries to perform a high rate of valid tests and the unusual trend towards older patients for NRAS mutations.


Colonic Neoplasms , Colorectal Neoplasms , Rectal Neoplasms , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Morocco , Mutation , North African People , Prospective Studies , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Neoplasm Metastasis/genetics
19.
Cells ; 12(12)2023 06 14.
Article En | MEDLINE | ID: mdl-37371090

Ezrin is the cytoskeletal organizer and functions in the modulation of membrane-cytoskeleton interaction, maintenance of cell shape and structure, and regulation of cell-cell adhesion and movement, as well as cell survival. Ezrin plays a critical role in regulating tumor metastasis through interaction with other binding proteins. Notably, Ezrin has been reported to interact with immune cells, allowing tumor cells to escape immune attack in metastasis. Here, we review the main functions of Ezrin, the mechanisms through which it acts, its role in tumor metastasis, and its potential as a therapeutic target.


Cell Adhesion , Cytoskeletal Proteins , Cytoskeleton , Neoplasm Metastasis , Cell Adhesion/genetics , Cell Adhesion/physiology , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Neoplasm Metastasis/genetics , Neoplasm Metastasis/physiopathology
20.
Nature ; 618(7964): 333-341, 2023 Jun.
Article En | MEDLINE | ID: mdl-37165194

Metastatic cancer remains an almost inevitably lethal disease1-3. A better understanding of disease progression and response to therapies therefore remains of utmost importance. Here we characterize the genomic differences between early-stage untreated primary tumours and late-stage treated metastatic tumours using a harmonized pan-cancer analysis (or reanalysis) of two unpaired primary4 and metastatic5 cohorts of 7,108 whole-genome-sequenced tumours. Metastatic tumours in general have a lower intratumour heterogeneity and a conserved karyotype, displaying only a modest increase in mutations, although frequencies of structural variants are elevated overall. Furthermore, highly variable tumour-specific contributions of mutational footprints of endogenous (for example, SBS1 and APOBEC) and exogenous mutational processes (for example, platinum treatment) are present. The majority of cancer types had either moderate genomic differences (for example, lung adenocarcinoma) or highly consistent genomic portraits (for example, ovarian serous carcinoma) when comparing early-stage and late-stage disease. Breast, prostate, thyroid and kidney renal clear cell carcinomas and pancreatic neuroendocrine tumours are clear exceptions to the rule, displaying an extensive transformation of their genomic landscape in advanced stages. Exposure to treatment further scars the tumour genome and introduces an evolutionary bottleneck that selects for known therapy-resistant drivers in approximately half of treated patients. Our data showcase the potential of pan-cancer whole-genome analysis to identify distinctive features of late-stage tumours and provide a valuable resource to further investigate the biological basis of cancer and resistance to therapies.


Genome, Human , Genomics , Neoplasm Metastasis , Neoplasms , Female , Humans , Male , Disease Progression , Mutation , Neoplasm Metastasis/genetics , Neoplasms/genetics , Genome, Human/genetics , Cohort Studies , Karyotyping , APOBEC Deaminases/metabolism
...