1,289 results on '"Cancer cell"'
Search Results
2. Single-cell RNA sequencing reveals distinct tumor microenvironmental patterns in lung adenocarcinoma
- Author
-
Christine Sers, Alexandra Trinks, Frederick Klauschen, David Horst, Annika Lehmann, Nils Blüthgen, Philipp Jurmeister, Markus Morkel, Jennifer Wiederspahn, Benedikt Obermayer, Jens Neudecker, Xizi Liang, Christine S. Falk, Jan Patrick Pett, Florian Uhlitz, Jens-Carsten Rückert, Aron Elsner, Philip Bischoff, Tomasz Dziodzio, and Dieter Beule
- Subjects
Cancer Research ,Lung Neoplasms ,Myeloid ,Tumour heterogeneity ,Cell ,Adenocarcinoma of Lung ,CD8-Positive T-Lymphocytes ,Biology ,tumor microenvironmental patterns ,Article ,Single-cell RNA sequencing ,Transcriptome ,Lymphocytes, Tumor-Infiltrating ,Biomarkers, Tumor ,Tumor Microenvironment ,Genetics ,medicine ,Sequencing ,Humans ,Molecular Biology ,Tumor microenvironment ,Gene Expression Profiling ,Cancer ,lung adenocarcinoma ,Prognosis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Survival Rate ,medicine.anatomical_structure ,Cancer cell ,Cancer research ,Adenocarcinoma ,Technology Platforms ,Single-Cell Analysis ,Non-small-cell lung cancer ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit::610 Medizin und Gesundheit ,CD8 - Abstract
Recent developments in immuno-oncology demonstrate that not only cancer cells, but also the tumor microenvironment can guide precision medicine. A comprehensive and in-depth characterization of the tumor microenvironment is challenging since its cell populations are diverse and can be important even if scarce. To identify clinically relevant microenvironmental and cancer features, we applied single-cell RNA sequencing to ten human lung adenocarcinomas and ten normal control tissues. Our analyses revealed heterogeneous carcinoma cell transcriptomes reflecting histological grade and oncogenic pathway activities, and two distinct microenvironmental patterns. The immune-activated CP²E microenvironment was composed of cancer-associated myofibroblasts, proinflammatory monocyte-derived macrophages, plasmacytoid dendritic cells and exhausted CD8+ T cells, and was prognostically unfavorable. In contrast, the inert N³MC microenvironment was characterized by normal-like myofibroblasts, non-inflammatory monocyte-derived macrophages, NK cells, myeloid dendritic cells and conventional T cells, and was associated with a favorable prognosis. Microenvironmental marker genes and signatures identified in single-cell profiles had progonostic value in bulk tumor profiles. In summary, single-cell RNA profiling of lung adenocarcinoma provides additional prognostic information based on the microenvironment, and may help to predict therapy response and to reveal possible target cell populations for future therapeutic approaches.
- Published
- 2021
3. Liprins in oncogenic signaling and cancer cell adhesion
- Author
-
Ivan de Curtis, Outi Monni, Henna Pehkonen, Pehkonen, H, de Curtis, I, and Monni, O.
- Subjects
Scaffold protein ,Cancer Research ,Review Article ,Protein tyrosine phosphatase ,Biology ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Nuclear Matrix-Associated Proteins ,Neoplasms ,Biomarkers, Tumor ,Cell Adhesion ,Genetics ,medicine ,Humans ,Cell adhesion ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Cancer ,Oncogenes ,medicine.disease ,Review article ,Gene Expression Regulation, Neoplastic ,Focal adhesion ,Tumor progression ,Multigene Family ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,Neuroscience ,Signal Transduction - Abstract
Liprins are a multifunctional family of scaffold proteins, identified by their involvement in several important neuronal functions related to signaling and organization of synaptic structures. More recently, the knowledge on the liprin family has expanded from neuronal functions to processes relevant to cancer progression, including cell adhesion, cell motility, cancer cell invasion, and signaling. These proteins consist of regions, which by prediction are intrinsically disordered, and may be involved in the assembly of supramolecular structures relevant for their functions. This review summarizes the current understanding of the functions of liprins in different cellular processes, with special emphasis on liprins in tumor progression. The available data indicate that liprins may be potential biomarkers for cancer progression and may have therapeutic importance.
- Published
- 2021
4. BET protein degradation triggers DR5-mediated immunogenic cell death to suppress colorectal cancer and potentiate immune checkpoint blockade
- Author
-
Lin Zhang, Kaylee Ermine, Xiao Tan, Jingshan Tong, Shaomeng Wang, Man Gao, Denise Risnik, Liangfang Shen, Xiangping Song, and Jian Yu
- Subjects
Cancer Research ,hemic and immune systems ,chemical and pharmacologic phenomena ,SPOP ,Biology ,Protein degradation ,medicine.disease_cause ,Immune checkpoint ,Bromodomain ,Cancer cell ,Genetics ,medicine ,Cancer research ,Immunogenic cell death ,Epigenetics ,Carcinogenesis ,Molecular Biology - Abstract
Bromodomain and extra-terminal domain (BET) family proteins are epigenetic readers that play a critical role in oncogenesis by controlling the expression of oncogenes such as c-Myc. Targeting BET family proteins has recently emerged as a promising anticancer strategy. However, the molecular mechanisms by which cancer cells respond to BET inhibition are not well understood. In this study, we found that inducing the degradation of BET proteins by the proteolysis targeting chimeras (PROTAC) approach potently suppressed the growth of colorectal cancer (CRC) including patient-derived tumors. Mechanistically, BET degradation transcriptionally activates Death Receptor 5 (DR5) to trigger immunogenic cell death (ICD) in CRC cells. Enhanced DR5 induction further sensitizes CRC cells with a mutation in Speckle-type POZ protein (SPOP). Furthermore, DR5 is indispensable for a striking antitumor effect of combining BET degradation and anti-PD-1 antibody, which was well tolerated in mice and almost eradicated syngeneic tumors. Our results demonstrate that BET degradation triggers DR5-mediated ICD to potently suppress CRC and potentiate immune checkpoint blockade. These results provide a rationale, mechanistic insights, and potential biomarkers for developing a precision CRC therapy by inducing BET protein degradation.
- Published
- 2021
5. Decoupling tumor cell metastasis from growth by cellular pilot protein TNFAIP8
- Author
-
Li Zhang, Xinyuan Li, Mingyue Li, Youhai H. Chen, Jason R. Goldsmith, Zienab Etwebi, Jiyeon Yu, Songlin Shi, Mayassa J. Bou-Dargham, Lin Wan, Honghong Sun, Ting Li, and Ali Zamani
- Subjects
Male ,Cancer Research ,Lung Neoplasms ,Skin Neoplasms ,Fibrosarcoma ,Biology ,Article ,Metastasis ,Cell membrane ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Movement ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Diethylnitrosamine ,Hippo Signaling Pathway ,Molecular Biology ,Cell Proliferation ,Hippo signaling pathway ,Cancer ,Chemotaxis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Second messenger system ,Cancer cell ,Cancer research ,Female ,Tumor necrosis factor alpha ,Apoptosis Regulatory Proteins ,Methylcholanthrene - Abstract
Cancer metastasis accounts for nearly 90% of all cancer deaths. Metastatic cancer progression requires both cancer cell migration to the site of the metastasis and subsequent proliferation after colonization. However, it has long been recognized that cancer cell migration and proliferation can be uncoupled; but the mechanism underlying this paradox is not well understood. Here we report that TNFAIP8 (tumor necrosis factor-α-induced protein 8), a “professional” transfer protein of phosphoinositide second messengers, promotes cancer cell migration or metastasis but inhibits its proliferation or cancer growth. TNFAIP8-deficient mice developed larger tumors, but TNFAIP8-deficient tumor cells completely lost their ability to migrate toward chemoattractants and were defective in colonizing lung tissues as compared to wild-type counterparts. Mechanistically, TNFAIP8 served as a cellular “pilot” of tumor cell migration by locally amplifying PI3K–AKT and Rac signals on the cell membrane facing chemoattractant; at the same time, TNFAIP8 also acted as a global inhibitor of tumor cell growth and proliferation by regulating Hippo signaling pathway. These findings help explain the migration–proliferation paradox of cancer cells that characterizes many cancers.
- Published
- 2021
6. Growth factor signaling predicts therapy resistance mechanisms and defines neuroblastoma subtypes
- Author
-
Vladimir S. Prassolov, Carol Stocking, Dmitry Konovalov, T D Lebedev, Pavel Spirin, Maria Suntsova, Anton Buzdin, Alexander Roumiantsev, Elmira Vagapova, Maxim Sorokin, Olga Astashkova, Petr M. Rubtsov, Alesya Mikheeva, and Uliana Vladimirova
- Subjects
MAPK/ERK pathway ,Cancer Research ,Cabozantinib ,Cell Survival ,medicine.medical_treatment ,Biology ,Article ,Paediatric cancer ,chemistry.chemical_compound ,Neuroblastoma ,Cell Line, Tumor ,Nerve Growth Factor ,Genetics ,medicine ,Cancer genomics ,Humans ,Molecular Biology ,Erythropoietin ,Protein Kinase Inhibitors ,Cell Proliferation ,Neoplasm Staging ,Crizotinib ,Growth factor ,medicine.disease ,Survival Analysis ,Axitinib ,Dasatinib ,Gene Expression Regulation, Neoplastic ,chemistry ,Drug Resistance, Neoplasm ,Cancer cell ,Mutation ,Cancer research ,medicine.drug ,Signal Transduction - Abstract
Neuroblastoma (NB) has a low frequency of recurrent mutations compared to other cancers, which hinders the development of targeted therapies and novel risk stratification strategies. Multikinase inhibitors have shown potential in treating high-risk NB, but their efficacy is likely impaired by the cancer cells’ ability to adapt to these drugs through the employment of alternative signaling pathways. Based on the expression of 48 growth factor-related genes in 1189 NB tumors, we have developed a model for NB patient survival prediction. This model discriminates between stage 4 NB tumors with favorable outcomes (>80% overall survival) and very poor outcomes (
- Published
- 2021
7. FBP1 regulates proliferation, metastasis, and chemoresistance by participating in C-MYC/STAT3 signaling axis in ovarian cancer
- Author
-
Xi Cheng, Midie Xu, Mengjiao Li, Yongdong Niu, Yufei Yang, Ziliang Wang, Haoran Li, Zihao Qi, Yangyang Pang, and Mingming Liu
- Subjects
STAT3 Transcription Factor ,Cancer Research ,Mice, Nude ,Biology ,Transfection ,medicine.disease_cause ,Article ,Metastasis ,Mice ,Downregulation and upregulation ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,STAT3 ,Molecular Biology ,Cell Proliferation ,Ovarian Neoplasms ,Gynaecological cancer ,medicine.disease ,Cancer metabolism ,Fructose-Bisphosphatase ,DNA-Binding Proteins ,DNA methylation ,Cancer cell ,Cancer research ,STAT protein ,biology.protein ,Female ,Carcinogenesis ,Ovarian cancer ,Signal Transduction ,Transcription Factors - Abstract
Fructose-1,6-bisphosphatase (FBP1) is a rate-limiting enzyme in gluconeogenesis and an important tumor suppressor in human malignancies. Here, we aimed to investigate the expression profile of FBP1 in ovarian cancer, the molecular mechanisms that regulate FBP1 expression and to examine how the FBP1 regulatory axis contributes to tumorigenesis and progression in ovarian cancer. We showed that FBP1 expression was significantly decreased in ovarian cancer tissues compared with normal ovarian tissues, and low-FBP1 expression predicted poor prognosis in patients with ovarian cancer. The enhanced expression of FBP1 in ovarian cancer cell lines suppressed proliferation and 2-D/3-D invasion, reduced aerobic glycolysis, and sensitized cancer cells to cisplatin-induced apoptosis. Moreover, DNA methylation and C-MYC binding at the promoter inhibited FBP1 expression. Furthermore, through physical interactions with signal transducer and activator of transcription 3 (STAT3), FBP1 suppressed nuclear translocation of STAT3 and exerted its non-metabolic enzymatic activity to induce the dysfunction of STAT3. Thus, our study suggests that FBP1 may be a valuable prognostic predictor for ovarian cancer. C-MYC-dependent downregulation of FBP1 acted as a tumor suppressor via modulating STAT3, and the C-MYC/FBP1/STAT3 axis could be a therapeutic target.
- Published
- 2021
8. Anoikis resistant gastric cancer cells promote angiogenesis and peritoneal metastasis through C/EBPβ-mediated PDGFB autocrine and paracrine signaling
- Author
-
Hong Chen, Zhi Yang, Wenxi Li, En Xu, Linseng Shi, Xingzhou Wang, Suhail Yousuf, Wenxian Guan, Xuefeng Xia, Ji Miao, Heng Yu, Min Zhao, Shangce Du, Xinya Zhu, and Xiaofeng Lu
- Subjects
Cancer Research ,Programmed cell death ,PDGFB ,Angiogenesis ,Cancer ,Biology ,medicine.disease ,Paracrine signalling ,Cancer cell ,Genetics ,Cancer research ,medicine ,Anoikis ,Autocrine signalling ,Molecular Biology - Abstract
Anoikis is a type of programmed cell death induced by loss of anchorage to the extracellular matrix (ECM). Anoikis resistance (AR) is crucial for the survival of metastatic cancer cells in blood, lymphatic circulation and distant organs. Compared to ordinary cancer cells, anoikis resistant cancer cells undergo various cellular and molecular alterations, probably characterizing the cells with unique features not limited to anoikis resistance. However, the molecular mechanisms connecting anoikis resistance to other metastatic properties are still poorly understood. Here, the biological interaction between anoikis resistance and angiogenesis as well as their involvement into peritoneal metastasis of gastric cancer (GC) were investigated in vitro and in vivo. The prognostic value of key components involved in this interaction was evaluated in the GC cohort. Compared to ordinary GC cells, GCAR cells exhibited stronger metastatic and pro-angiogenic traits corresponding to elevated PDGFB secretion. Mechanistically, transcription factor C/EBPβ facilitated PDGFB transcription by directly binding to and interacting with PDGFB promoter elements, subsequently increasing PDGFB secretion. Secreted PDGFB promoted the survival of detached GC cells through a C/EBPβ-dependent self-feedback loop. Moreover, secreted PDGFB promoted angiogenesis in metastases via activation of the MAPK/ERK signaling pathway in vascular endothelial cells. Both C/EBPβ activation level and PDGFB expression were significantly elevated in GC and correlated with metastatic progression and poor prognosis of patients with GC. Overall, interaction between GCAR cells and vascular endothelial cells promotes angiogenesis and peritoneal metastasis of GC based on C/EBPβ-mediated PDGFB autocrine and paracrine signaling. C/EBPβ-PDGFB-PDGFRβ-MAPK axis promises to be potential prognostic biomarkers and therapeutic targets for peritoneal metastasis of GC.
- Published
- 2021
9. CRIP1 cooperates with BRCA2 to drive the nuclear enrichment of RAD51 and to facilitate homologous repair upon DNA damage induced by chemotherapy
- Author
-
Yannan Zheng, Min Shi, Zhenhua Huang, Jianping Bin, Huiying Sun, Jianhua Wu, Rui Zhou, Dongqiang Zeng, Xiaoxiang Rong, Na Huang, Li Sun, Dingling Zhang, Zhaowei Wen, Yulin Liao, and Wangjun Liao
- Subjects
0301 basic medicine ,Cancer Research ,DNA damage ,Poly ADP ribose polymerase ,Poly (ADP-Ribose) Polymerase-1 ,RAD51 ,Biology ,Article ,Piperazines ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Homologous recombination ,Molecular Biology ,Protein kinase B ,BRCA2 Protein ,Cell Nucleus ,Cisplatin ,Recombinational DNA Repair ,Cancer therapeutic resistance ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Phthalazines ,DNA Damage ,medicine.drug - Abstract
Homologous recombination (HR) repair is an important determinant of chemosensitivity. However, the mechanisms underlying HR regulation remain largely unknown. Cysteine-rich intestinal protein 1 (CRIP1) is a member of the LIM/double-zinc finger protein family and is overexpressed and associated with prognosis in several tumor types. However, to date, the functional role of CRIP1 in cancer biology is poorly understood. Here we found that CRIP1 downregulation causes HR repair deficiency with concomitant increase in cell sensitivity to cisplatin, epirubicin, and the poly ADP-ribose polymerase (PARP) inhibitor olaparib in gastric cancer cells. Mechanistically, upon DNA damage, CRIP1 is deubiquitinated and upregulated by activated AKT signaling. CRIP1, in turn, promotes nuclear enrichment of RAD51, which is a prerequisite step for HR commencement, by stabilizing BRCA2 to counteract FBXO5-targeted RAD51 degradation and by binding to the core domain of RAD51 (RAD51184–257) in coordination with BRCA2, to facilitate nuclear export signal masking interactions between BRCA2 and RAD51. Moreover, through mass spectrometry screening, we found that KPNA4 is at least one of the carriers controlling the nucleo-cytoplasmic distribution of the CRIP1–BRCA2–RAD51 complex in response to chemotherapy. Consistent with these findings, RAD51 inhibitors block the CRIP1-mediated HR process, thereby restoring chemotherapy sensitivity of gastric cancer cells with high CRIP1 expression. Analysis of patient specimens revealed an abnormally high level of CRIP1 expression in GC tissues compared to that in the adjacent normal mucosa and a significant negative association between CRIP1 expression and survival time in patient cohorts with different types of solid tumors undergoing genotoxic treatments. In conclusion, our study suggests an essential function of CRIP1 in promoting HR repair and facilitating gastric cancer cell adaptation to genotoxic therapy.
- Published
- 2021
10. TRIM44 mediated p62 deubiquitination enhances DNA damage repair by increasing nuclear FLNA and 53BP1 expression
- Author
-
Nami McCarty, Lin Lyu, and Tsung-Chin Lin
- Subjects
0301 basic medicine ,Genome instability ,Cancer Research ,DNA End-Joining Repair ,DNA Repair ,Cell Survival ,DNA damage ,DNA repair ,Filamins ,Protein degradation ,Biology ,Radiation Tolerance ,Genomic Instability ,Article ,Tripartite Motif Proteins ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Radiation, Ionizing ,Sequestosome-1 Protein ,Autophagy ,Genetics ,Humans ,FLNA ,Molecular Biology ,Intracellular Signaling Peptides and Proteins ,Recombinational DNA Repair ,Cell biology ,Protein Transport ,030104 developmental biology ,Gene Expression Regulation ,Cytoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Protein Multimerization ,Multiple Myeloma ,Tumor Suppressor p53-Binding Protein 1 ,DNA Damage ,Protein Binding - Abstract
Cancer cells show increases in protein degradation pathways, including autophagy, during progression to meet the increased protein degradation demand and support cell survival. On the other hand, reduced autophagy activity during aging is associated with a reduced DNA damage response and increased genomic instability. Therefore, it is a puzzling how DNA repair can be increased in cancer cells that are resistant to chemotherapies or during progression when autophagy activity is intact or increased. We discovered that tripartite motif containing 44 (TRIM44) is a pivotal element regulating the DNA damage response in cancer cells with intact autophagy. TRIM44 deubiquitinates p62, an autophagy substrate, which leads to its oligomerization. This prevents p62 localization to the nucleus upon irradiation. Increased cytoplasmic retention of p62 by TRIM44 prevents the degradation of FLNA and 53BP1, which increases DNA damage repair. Together, our data support TRIM44 a potential therapeutic target for therapy-resistant tumor cells with intact autophagy.
- Published
- 2021
11. NF-κB-activated SPRY4-IT1 promotes cancer cell metastasis by downregulating TCEB1 mRNA via Staufen1-mediated mRNA decay
- Author
-
Ming Zhang, Minjie Wei, Qiang Zhang, Longyang Jiang, Miao He, Lin Zhao, Qiutong Guan, Yalun Li, and Jingdong Zhang
- Subjects
0301 basic medicine ,Untranslated region ,Cell biology ,Cancer Research ,RNA Stability ,Elongin ,Biology ,Article ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Alu Elements ,Cell Line, Tumor ,Neoplasms ,microRNA ,Genetics ,medicine ,Humans ,Neoplasm Metastasis ,3' Untranslated Regions ,Molecular Biology ,Cancer ,Neoplasm Staging ,Messenger RNA ,Binding Sites ,Competing endogenous RNA ,NF-kappa B ,RNA-Binding Proteins ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,Cytoskeletal Proteins ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,RNA Interference ,RNA, Long Noncoding ,Ovarian cancer ,Signal Transduction - Abstract
Previous study demonstrated that most long non-coding RNAs (lncRNAs) function as competing endogenous RNAs or molecular sponges to negatively modulate miRNA and regulate tumor development. However, the molecular mechanisms of lncRNAs in cancer are not fully understood. Our study describes the role of the lncRNA SPRY4 intronic transcript 1 (SPRY4-IT1) in cancer metastasis by mechanisms related to Staufen1 (STAU1)-mediated mRNA decay (SMD). Briefly, we found that, high SPRY4-IT1 expression was associated with aggressiveness and poor outcome in human colorectal, breast and ovarian cancer tissues. In addition, functional assays revealed that SPRY4-IT1 significantly promoted colorectal, breast and ovarian cancer metastasis in vitro and in vivo. Mechanistically, microarray analyses identified several differentially-expressed genes upon SPRY4-IT1 overexpression in HCT 116 colorectal cancer cells. Among them, the 3′-UTR of transcription elongation factor B subunit 1 (TCEB1) mRNA can base-pair with the Alu element in the 3′-UTR of SPRY4-IT1. Moreover, SPRY4-IT1 was found to bind STAU1, promote STAU1 recruitment to the 3′-UTR of TCEB1 mRNA, and affect TCEB1 mRNA stability and expression, resulting in hypoxia-inducible factor 1α (HIF-1α) upregulation, and thereby affecting cancer cell metastasis. In addition, STAU1 depletion abrogated TCEB1 SMD and alleviated the pro-metastatic effect of SPRY4-IT1 overexpression. Significantly, we revealed that SPRY4-IT1 is also transactivated by NF-κB/p65, which activates SPRY4-IT1 to inhibit TCEB1 expression, and subsequently upregulate HIF-1α. In conclusion, our results highlight a novel mechanism of cytoplasmic lncRNA SPRY4-IT1 in which SPRY4-IT1 affecting TCEB1 mRNA stability via STAU1-mediated degradation during cancer metastasis.
- Published
- 2021
12. E3 ubiquitin ligase HECTD2 mediates melanoma progression and immune evasion
- Author
-
Laura Doglio, Georgina H. Cornish, George Kassiotis, Tom Lesluyes, Peter Van Loo, Anastasiya Kazachenka, Thomas Tüting, Eleonora Ottina, Veera Panova, Jan Attig, Kevin Litchfield, George R. Young, James I. MacRae, Charles Swanton, and Joanna Kirkpatrick
- Subjects
0301 basic medicine ,Cancer Research ,Cell ,Mice ,0302 clinical medicine ,Ecology,Evolution & Ethology ,Ubiquitin ,Melanoma ,Chemical Biology & High Throughput ,Human Biology & Physiology ,biology ,Genome Integrity & Repair ,Cell cycle ,Ubiquitin ligase ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Genetics & Genomics ,Cell Division ,Model organisms ,Ubiquitin-Protein Ligases ,Immunology ,Infectious Disease ,Biochemistry & Proteomics ,Article ,Signalling & Oncogenes ,03 medical and health sciences ,Immune system ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Cell Proliferation ,Immune Evasion ,Computational & Systems Biology ,Lipogenesis ,FOS: Clinical medicine ,Cancer ,Cell Biology ,Tumour Biology ,medicine.disease ,Metabolism ,030104 developmental biology ,Proteolysis ,Cancer cell ,biology.protein ,Cancer research ,Genetic markers ,Developmental Biology ,Structural Biology & Biophysics - Abstract
The ubiquitin-proteasome system maintains protein homoeostasis, underpins the cell cycle, and is dysregulated in cancer. However, the role of individual E3 ubiquitin ligases, which mediate the final step in ubiquitin-mediated proteolysis, remains incompletely understood. Identified through screening for cancer-specific endogenous retroviral transcripts, we show that the little-studied E3 ubiquitin ligase HECTD2 exerts dominant control of tumour progression in melanoma. HECTD2 cell autonomously drives the proliferation of human and murine melanoma cells by accelerating the cell cycle. HECTD2 additionally regulates cancer cell production of immune mediators, initiating multiple immune suppressive pathways, which include the cyclooxygenase 2 (COX2) pathway. Accordingly, higher HECTD2 expression is associated with weaker anti-tumour immunity and unfavourable outcome of PD-1 blockade in human melanoma and counteracts immunity against a model tumour antigen in murine melanoma. This central, multifaceted role of HECTD2 in cancer cell-autonomous proliferation and in immune evasion may provide a single target for a multipronged therapy of melanoma.
- Published
- 2021
13. Application of machine learning to large in-vitro databases to identify cancer cell characteristics: telomerase reverse transcriptase (TERT) expression
- Author
-
Jeff Sherman, Yisroel Brumer, and Grant Verstandig
- Subjects
0301 basic medicine ,Cancer Research ,Biology ,Machine learning ,computer.software_genre ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,Databases, Genetic ,Biomarkers, Tumor ,Genetics ,medicine ,Humans ,CRISPR ,Telomerase reverse transcriptase ,Epigenetics ,Telomerase ,Molecular Biology ,Gene ,business.industry ,Gene Expression Profiling ,Liver Neoplasms ,Computational Biology ,Cancer ,Molecular Sequence Annotation ,medicine.disease ,HDAC1 ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Drug development ,030220 oncology & carcinogenesis ,Cancer cell ,Artificial intelligence ,business ,computer - Abstract
Advances in biotechnology and machine learning have created an enhanced environment for unearthing and exploiting previously unrecognized relationships between genomic and epigenetic data with potential therapeutic implications. We applied advanced algorithms to data from the Cancer Dependency Map to uncover increasingly complex relationships. Specifically, we investigate characteristics of tumor cell lines with varying levels of telomerase reverse transcriptase (TERT) expression in liver cancer. The findings indicate that the effect of CRISPR knockout of Histone Deacetylase 1 (HDAC1) and numerous individual respiratory complex I genes is strongly related to the level of TERT expression, with knockout being particularly efficacious at killing or inhibiting growth of tumor cells with low levels of TERT expression for HDAC1 and high levels for Complex I genes. These findings suggest key biomarkers for therapeutic efficacy and yield novel potential pathways for drug development and provide further proof of principle for the potential of artificial intelligence in oncology.
- Published
- 2021
14. Asporin represses gastric cancer apoptosis via activating LEF1-mediated gene transcription independent of β-catenin
- Author
-
Shutian Zhang, Zheng Zhang, Yu Zhao, Li Min, Si Liu, Shengtao Zhu, Lei Chen, Qingdong Guo, Hengcun Li, and Peng Li
- Subjects
0301 basic medicine ,Cancer Research ,Gene knockdown ,Lymphoid Enhancer-Binding Factor 1 ,Cell growth ,Wnt signaling pathway ,Asporin ,Apoptosis ,Promoter ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Stomach Neoplasms ,030220 oncology & carcinogenesis ,Catenin ,embryonic structures ,Cancer cell ,Genetics ,Humans ,Ectopic expression ,Molecular Biology - Abstract
Asporin (ASPN) presents in the tumor microenvironment and exhibits a cancer-promoting effect as a stroma protein. Even though ASPN has already been observed inside cancer cells, the functions of intracellular ASPN and its underlying mechanisms remain unknown. Here we reported that ASPN was upregulated in different stages of gastric cancer (GC), and associated with a poor prognosis. Moreover, we found that ASPN markedly inhibited GC cell apoptosis and promoted cell growth in vitro and in vivo. Further mechanism investigations revealed that ASPN directly binding to lymphoid enhancer-binding factor 1 (LEF1) and promoted LEF1-mediated gene transcription independent of β-catenin, the classic co-factor in the Wnt/LEF1 pathway. We also demonstrated that ASPN selectively facilitated LEF1 binding to and activating the promoters of PTGS2, IL6, and WISP1 to promote their transcription. The suppression of cell apoptosis by ASPN overexpression could be attenuated by LEF1 knockdown or 100 µM aspirin (PTGS2 inhibitor), and siASPN mediated apoptosis could be rescued by LEF1 ectopic expression or adding recombinant IL6. Therefore, we concluded that ASPN repressed GC cell apoptosis via activating LEF1-mediated gene transcription independent of β-catenin, which could serve as a potential prognostic biomarker in GC patients.
- Published
- 2021
15. αSMA+ fibroblasts suppress Lgr5+ cancer stem cells and restrain colorectal cancer progression
- Author
-
Raghu Kalluri, Bingrui Li, Hikaru Sugimoto, Changsoo Kwak, Karina Vázquez-Arreguín, Valerie S. LeBleu, Michelle L. Kirtley, Xiaofeng Zheng, and Kathleen M. McAndrews
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,Bone Morphogenetic Protein 4 ,CD8-Positive T-Lymphocytes ,Biology ,T-Lymphocytes, Regulatory ,Article ,Receptors, G-Protein-Coupled ,Transforming Growth Factor beta1 ,Mice ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Cell Movement ,Cancer stem cell ,Cell Line, Tumor ,Tumor Microenvironment ,Genetics ,medicine ,Animals ,Neoplasm Invasiveness ,Molecular Biology ,Tumor microenvironment ,LGR5 ,FOXP3 ,Cancer ,Forkhead Transcription Factors ,medicine.disease ,Actins ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,Neoplastic Stem Cells ,Cancer research ,Colorectal Neoplasms - Abstract
The development and progression of solid tumors is dependent on cancer cell autonomous drivers and the tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) in the TME possess both tumor-promoting and tumor-restraining functions. In the current study, we interrogated the role of αSMA(+) CAFs in a genetic mouse model of metastatic colorectal cancer (CRC). Selective depletion of αSMA(+) CAFs resulted in increased tumor invasiveness, lymph node metastasis, and reduced overall survival. Depletion of αSMA(+) CAFs reduced BMP4 and increased TGFβ1 secretion from stromal cells, and was associated with increased Lgr5(+) cancer stem-like cells (CSCs) and the generation of an immunosuppressive TME with increased frequency of Foxp3(+) regulatory T cells and suppression of CD8(+) T cells. This study demonstrates that αSMA(+) CAFs in CRC exert tumor-restraining functions via BMP4/TGFβ1 paracrine signaling that serves to suppress Lgr5(+) CSCs and promote anti-tumor immunity, ultimately limiting CRC progression.
- Published
- 2021
16. Innovative dual system approach for selective eradication of cancer cells using viral-based delivery of natural bacterial toxin–antitoxin system
- Author
-
Dina Kazanov, Ahmad Fokra, Ilana Boustanai, Nadir Arber, Shiran Shapira, and Marina Ben Shimon
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Cell Survival ,Genetic Vectors ,Gene Expression ,Biology ,medicine.disease_cause ,Response Elements ,Article ,Adenoviridae ,03 medical and health sciences ,0302 clinical medicine ,Gene therapy ,Targeted therapies ,Bacterial Proteins ,In vivo ,Cell Line, Tumor ,Neoplasms ,Gene Order ,Genetics ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Oncolytic Virotherapy ,Oncogene ,Toxin ,Gene Transfer Techniques ,Cancer ,Toxin-Antitoxin Systems ,Pancreatic cancer ,Genetic Therapy ,medicine.disease ,Flow Cytometry ,Colorectal cancer ,In vitro ,Oncolytic Viruses ,030104 developmental biology ,Genes, ras ,030220 oncology & carcinogenesis ,Cancer cell ,Viruses ,Cancer research ,Antitoxin ,Lung cancer - Abstract
The inactivation of p53, a tumor suppressor, and the activation of the RAS oncogene are the most frequent genetic alterations in cancer. We have shown that a unique E. coli MazF-MazE toxin–antitoxin (TA) system can be used for selective and effective eradication of RAS-mutated cancer cells. This out of the box strategy holds great promise for effective cancer treatment and management. We provide proof of concept for a novel platform to selectively eradicate cancer cells using an adenoviral delivery system based on the adjusted natural bacterial system. We generated adenoviral vectors carrying the mazF toxin (pAdEasy-Py4-SV40mP-mCherry-MazF) and the antitoxin mazE (pAdEasy-RGC-SV40mP-MazE-IRES-GFP) under the regulation of RAS and p53, resp. The control vector carries the toxin without the RAS-responsive element (pAdEasy-ΔPy4-SV40mP-mCherry-MazF). In vitro, the mazF-mazE TA system (Py4-SV40mP-mCherry-MazF+RGC-SV40mP-MazE-IRES-GFP) induced massive, dose-dependent cell death, at 69% compared to 19% for the control vector, in a co-infected HCT116 cell line. In vivo, the system caused significant tumor growth inhibition of HCT116 (KRASmut/p53mut) tumors at 73 and 65% compared to PBS and ΔPY4 control groups, resp. In addition, we demonstrate 65% tumor growth inhibition in HCT116 (KRASmut/p53wt) cells, compared to the other two control groups, indicating a contribution of the antitoxin in blocking system leakage in WT RAS cells. These data provide evidence of the feasibility of using mutations in the p53 and RAS pathway to efficiently kill cancer cells. The platform, through its combination of the antitoxin (mazE) with the toxin (mazF), provides effective protection of normal cells from basal low activity or leakage of mazF.
- Published
- 2021
17. Application of machine learning to large in vitro databases to identify drug–cancer cell interactions: azithromycin and KLK6 mutation status
- Author
-
Yisroel Brumer, John W. Rowe, Jeff Sherman, and Grant Verstandig
- Subjects
0301 basic medicine ,Drug ,Cancer Research ,Databases, Pharmaceutical ,media_common.quotation_subject ,Azithromycin ,Biology ,Machine learning ,computer.software_genre ,Machine Learning ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Databases, Genetic ,Drug Discovery ,Genetics ,medicine ,Humans ,CRISPR ,Molecular Biology ,Gene knockout ,media_common ,business.industry ,Drug discovery ,Cancer ,medicine.disease ,Anti-Bacterial Agents ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Cancer cell ,Mutation (genetic algorithm) ,Kallikreins ,Artificial intelligence ,business ,computer ,medicine.drug - Abstract
Recent advances in machine learning promise to yield novel insights by interrogation of large datasets ranging from gene expression and mutation data to CRISPR knockouts and drug screens. We combined existing and new algorithms with available experimental data to identify potentially clinically relevant relationships to provide a proof of principle for the promise of machine learning in oncological drug discovery. Specifically, we screened cell line data from the Cancer Dependency Map for the effects of azithromycin, which has been shown to kill cancer cells in vitro. Our findings demonstrate a strong relationship between Kallikrein Related Peptidase 6 (KLK6) mutation status and the ability of azithromycin to kill cancer cells in vitro. While the application of azithromycin showed no meaningful average effect in KLK6 wild-type cell lines, statistically significant enhancements of cell death are seen in multiple independent KLK6-mutated cancer cell lines. These findings suggest a potentially valuable clinical strategy in patients with KLK6-mutated malignancies.
- Published
- 2021
18. Integrase-derived peptides together with CD24-targeted lentiviral particles inhibit the growth of CD24 expressing cancer cells
- Author
-
Abraham Loyter, Arye Blachar, Eynat Finkelshtein, Daniel Elbirt, Dov Hershkovitz, Esmira Naftali, Shiran Shapira, Ido Wolf, Mori Hay-Levy, Irena Stepansky, Nadir Arber, Roy Dekel, Eli Brazowski, Dina Kazanov, and Faina Bedny
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,030106 microbiology ,Mice, Nude ,Apoptosis ,Biology ,Article ,03 medical and health sciences ,Mice ,Targeted therapies ,In vivo ,Cell Line, Tumor ,Neoplasms ,Genetics ,medicine ,Animals ,Humans ,Tissue Distribution ,Drug safety ,Molecular Biology ,Oncolytic Virotherapy ,Integrases ,Lentivirus ,Antibodies, Monoclonal ,CD24 Antigen ,Raltegravir ,biology.organism_classification ,Xenograft Model Antitumor Assays ,In vitro ,Peptide Fragments ,Integrase ,030104 developmental biology ,Cancer cell ,Cancer research ,biology.protein ,medicine.drug - Abstract
The integration of viral DNA into the host genome is mediated by viral integrase, resulting in the accumulation of double-strand breaks. Integrase-derived peptides (INS and INR) increase the number of integration events, leading to escalated genomic instability that induces apoptosis. CD24 is a surface protein expressed mostly in cancer cells and is very rarely found in normal cells. Here, we propose a novel targeted cancer therapeutic platform based on the lentiviral integrase, stimulated by integrase-derived peptides, that are specifically delivered to cancerous cells via CD24 antigen-antibody targeting. INS and INR were synthesized and humanized and anti-CD24 antibodies were fused to the lentivirus envelope. The activity, permeability, stability, solubility, and toxicity of these components were analyzed. Cell death was measured by fluorescent microscopy and enzymatic assays and potency were tested in vitro and in vivo. Lentivirus particles, containing non-functional DNA led to massive cell death (40–70%). Raltegravir, an antiretroviral drug, inhibited the induction of apoptosis. In vivo, single and repeated administrations of INS/INR were well tolerated without any adverse effects. Tumor development in nude mice was significantly inhibited (by 50%) as compared to the vehicle arm. In summary, a novel and generic therapeutic platform for selective cancer cell eradication with excellent efficacy and safety are presented.
- Published
- 2021
19. Enhancer rewiring in tumors: an opportunity for therapeutic intervention
- Author
-
Raphaël Margueron, Laia Richart, and François-Clément Bidard
- Subjects
0301 basic medicine ,Cancer Research ,Effector ,Cell Cycle Proteins ,Computational biology ,Biology ,Interactome ,Chromatin ,03 medical and health sciences ,Enhancer Elements, Genetic ,030104 developmental biology ,0302 clinical medicine ,Transcription (biology) ,Neoplasms ,030220 oncology & carcinogenesis ,Cancer cell ,Genetics ,Animals ,Humans ,Enhancer ,Molecular Biology ,Gene ,Transcription factor ,Transcription Factors - Abstract
Enhancers are cis-regulatory sequences that fine-tune expression of their target genes in a spatiotemporal manner. They are recognized by sequence-specific transcription factors, which in turn recruit transcriptional coactivators that facilitate transcription by promoting assembly and activation of the basal transcriptional machinery. Their functional importance is underscored by the fact that they are often the target of genetic and nongenetic events in human disease that disrupt their sequence, interactome, activation potential, and/or chromatin environment. Dysregulation of transcription and addiction to transcriptional effectors that interact with and modulate enhancer activity are common features of cancer cells and are amenable to therapeutic intervention. Here, we discuss the current knowledge on enhancer biology, the broad spectrum of mechanisms that lead to their malfunction in tumor cells, and recent progress in developing drugs that efficaciously target their dependencies.
- Published
- 2021
20. Long non-coding RNA SMASR inhibits the EMT by negatively regulating TGF-β/Smad signaling pathway in lung cancer
- Author
-
Wenzhong Liu, Lele Xu, Yu Wang, Shujuan Shao, Xuefeng Liu, Yan Wang, Qimin Zhan, Yuying Hu, Tongtong Li, and Lijie Huang
- Subjects
0301 basic medicine ,Cancer Research ,Gene knockdown ,Cancer ,SMAD ,Biology ,medicine.disease ,Long non-coding RNA ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Cancer cell ,Genetics ,medicine ,Cancer research ,Phosphorylation ,Signal transduction ,Lung cancer ,Molecular Biology - Abstract
TGF-β/Smad signaling pathway plays an important role in EMT during cancer progression. Long non-coding RNAs (lncRNAs) are involved in various behaviors of cancer cells, including EMT. Here, we report a novel lncRNA adjacent to Smad3, named Smad3-associated long non-coding RNA (SMASR). SMASR is downregulated by TGF-β via Smad2/3 in lung cancer cells. Knockdown of SMASR induces EMT and increases the migration and invasion of lung cancer cells. Moreover, knockdown of SMASR promotes the phosphorylation of Smad2/3. Mechanistically, SMASR interacts with Smad2/3 and inhibits the expression of TGFBR1, the TGF-β type I receptor responsible for phosphorylation of Smad2/3, thus leading to inactivation of TGF-β/Smad signaling pathway. Clinically, SMASR is downregulated in lung cancer tissues. Collectively, our findings prove a critical role of SMASR in EMT of lung cancer by forming a negative feedback loop with TGF-β/Smad signaling pathway.
- Published
- 2021
21. Ferroptosis as a mechanism to mediate p53 function in tumor radiosensitivity
- Author
-
Guang Lei, Anil K. Sood, Yuelong Yan, Boyi Gan, Pranavi Koppula, Chao Mao, Jinsong Liu, Xudong Zhang, Weijie Cheng, Ting Hong, Yilei Zhang, and Xiaoguang Liu
- Subjects
p53 ,0301 basic medicine ,Cancer Research ,Amino Acid Transport System y+ ,Mice, SCID ,Biology ,SLC7A11 ,Radiation Tolerance ,Article ,Lipid peroxidation ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Mice, Inbred NOD ,Cell Line, Tumor ,Neoplasms ,Radioresistance ,Genetics ,medicine ,Animals ,Humans ,Radiosensitivity ,Molecular Biology ,radiotherapy ,Effector ,Cancer ,lipid peroxidation ,medicine.disease ,Xenograft Model Antitumor Assays ,ferroptosis ,cell death ,030104 developmental biology ,chemistry ,radiosensitivity ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Female ,tumor suppression ,Tumor Suppressor Protein p53 ,Function (biology) - Abstract
Ferroptosis, a form of regulated cell death triggered by lipid peroxidation, was recently identified as an important mechanism in radiotherapy (RT)-mediated tumor suppression and radioresistance, although the exact genetic contexts in which to target ferroptosis in RT remains to be defined. p53 is the most commonly mutated gene in human cancers and a major effector to RT. Here, we identify ferroptosis as a critical mechanism to mediate p53 function in tumor radiosensitivity. Mechanistically, RT-mediated p53 activation antagonizes RT-induced SLC7A11 expression and represses glutathione synthesis, thereby promoting RT-induced lipid peroxidation and ferroptosis. p53 deficiency promotes radioresistance in cancer cells or tumors at least partly through SLC7A11-mediated ferroptosis inhibition. Ferroptosis inducers (FINs) that inhibit SLC7A11 exert significant radiosensitizing effects in tumor organoids and patient-derived xenografts with p53 mutation or deficiency. Finally, we show that RT-induced ferroptosis correlates with p53 activation and better clinical outcomes to RT in cancer patients. Together, our study uncovers a previously unappreciated role of ferroptosis in p53-mediated radiosensitization and suggest using FINs in combination with RT to treat p53-mutant cancers.
- Published
- 2021
22. LIMK1 promotes peritoneal metastasis of gastric cancer and is a therapeutic target
- Author
-
Wei Kang, Xi Kang, Sinan Zhao, Ka Fai To, Philip Wai Yan Chiu, Weixin Liu, Guiying Wang, Chi Chun Wong, Jingyu Deng, Jun Yu, Enders K.W. Ng, Weilin Li, and Han Liang
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Mice, Nude ,Biology ,Article ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,In vivo ,Cell Line, Tumor ,Oximes ,Genetics ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Neoplasm Metastasis ,Protein Kinase Inhibitors ,Molecular Biology ,Peritoneal Neoplasms ,Cell Proliferation ,Mice, Inbred BALB C ,Sequence Analysis, RNA ,Kinase ,Imidazoles ,Lim Kinases ,Cancer ,Cell migration ,Dabrafenib ,medicine.disease ,Xenograft Model Antitumor Assays ,Survival Rate ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Immunohistochemistry ,medicine.drug - Abstract
Peritoneal metastasis is a common form of metastasis among advanced gastric cancer patients. In this study, we reported the identification of LIM domain kinase 1 (LIMK1) as a promoter of gastric cancer peritoneal metastasis, and its potential to be a therapeutic target of dabrafenib (DAB). Using transcriptomic sequencing of paired gastric cancer peritoneal metastasis, primary tumors, and normal gastric tissues, we first unveiled that LIMK1 is selectively up-regulated in metastatic tumors. Increased LIMK1 in gastric cancer peritoneal metastasis was validated by immunohistochemistry analysis of an independent patient cohort. In vitro functional studies demonstrated that LIMK1 knockout or knockdown significantly inhibited cell migration and invasion of gastric cancer cells. LIMK1 knockout also abrogated peritoneal and liver metastases of gastric cancer cells in nude mice in vivo. Dabrafenib, a small molecule targeting LIMK1, was found to decrease cell migration and invasion of gastric cancer cells in vitro and abolish peritoneal and liver metastasis formation in vivo. Mechanistically, either LIMK1 knockout or Dabrafenib inhibited LIMK1 expression and phosphorylation of its downstream target cofilin. Taken together, our results demonstrated that LIMK1 functions as a metastasis promoter in gastric cancer by inhibiting LIMK1-p-cofilin and that Dabrafenib has the potential to serve as a novel treatment for gastric cancer peritoneal metastasis.
- Published
- 2021
23. Clocking cancer: the circadian clock as a target in cancer therapy
- Author
-
Meng Qu, Francesca Battaglin, Evanthia T. Roussos Torres, Erin Spiller, Shivani Soni, Yuanzhong Pan, Heinz-Josef Lenz, Sofi Castanon, Priscilla Chan, Shannon M. Mumenthaler, and Steve A. Kay
- Subjects
0301 basic medicine ,Cancer Research ,Circadian clock ,Cancer therapy ,Cancer ,Endogeny ,Oncogenes ,Biology ,medicine.disease ,Article ,03 medical and health sciences ,Therapeutic approach ,030104 developmental biology ,0302 clinical medicine ,Circadian Clocks ,Neoplasms ,030220 oncology & carcinogenesis ,Cancer cell ,Genetics ,medicine ,Humans ,Pharmacological modulation ,Cancer risk ,Molecular Biology ,Neuroscience - Abstract
Disruption of the cellular pathway modulating endogenous 24-hour rhythms, referred to as ‘the circadian clock’, has been recently proven to be associated with cancer risk, development and progression. This pathway operates through a complex network of transcription-translation feedback loops generated by a set of interplaying proteins. The expression of core circadian clock genes is frequently dysregulated in human tumors; however, the specific effects and underlying mechanisms seem to vary depending on the cancer types and are not fully understood. Additionally, specific oncogenes may differentially induce the dysregulation of the circadian clock in tumors. Pharmacological modulation of clock components has been shown to result in specific lethality in certain types of cancer cells, and thus holds great promise as a novel anti-cancer therapeutic approach. Here we present an overview of the rationale and current evidence for targeting the clock in cancer treatment.
- Published
- 2021
24. FOXO3 is a latent tumor suppressor for FOXO3-positive and cytoplasmic-type gastric cancer cells
- Author
-
Noriyuki Inaki, Kazuhiro Murakami, Yusuke Maeda, Toshikatsu Tsuji, Hiroko Oshima, Kenji Kita, Hirofumi Takemura, Masanobu Oshima, and Hideyuki Saya
- Subjects
0301 basic medicine ,Cancer Research ,Endogeny ,Biology ,medicine.disease_cause ,Article ,03 medical and health sciences ,Cell growth ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Stomach Neoplasms ,Genetics ,medicine ,Animals ,Genes, Tumor Suppressor ,Gastrointestinal cancer ,Nuclear export signal ,Molecular Biology ,Transcription factor ,Forkhead Box Protein O3 ,Cancer ,medicine.disease ,030104 developmental biology ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Cancer cell ,FOXO3 ,Cancer research ,Carcinogenesis ,Gastric cancer ,Proto-Oncogene Proteins c-akt - Abstract
FOXO3 is a member of the FOXO transcription factors thought to play a tumor-suppressor role in gastrointestinal cancer, while tumor-promoting function of FOXO3 has also been reported. These results suggest a context-dependent function of FOXO3 in tumor development. However, the relationship between the FOXO3 expression pattern and its role in tumorigenesis has not been elucidated. We examined the FOXO3 expression in 65 human primary gastric cancer and patient-derived xenograft tissues by immunohistochemistry and identified three subtypes according to subcellular localization: FOXO3-nuclear accumulated (FOXO3-Nuc), FOXO3-nuclear/cytoplasmic or cytoplasmic distributed (FOXO3-Cyt), and FOXO3-negative. In the FOXO3-Cyt gastric cancer cells, the expression of the constitutive active mutant FOXO3 (Act-ER FOXO3) induced the nuclear accumulation of FOXO3 and significantly suppressed colony formation and proliferation. The inhibition of the PI3K-AKT pathway by inhibitor treatment also suppressed the proliferation of FOXO3-Cyt gastric cancer cells, which was associated with the nuclear accumulation of endogenous FOXO3. Furthermore, the expression of Act-ER FOXO3 by an endogenous promoter significantly suppressed gastric tumorigenesis in Gan mice, a model of gastric cancer. Finally, treatment of FOXO3-Cyt human gastric cancer-derived organoids with an AKT inhibitor significantly suppressed the survival and proliferation. These results indicate that FOXO3 is a latent tumor suppressor for FOXO3-Cyt-type gastric cancer cells and that activation of the PI3K-AKT pathway protects this type of gastric cancer cell from FOXO3-mediated growth suppression via constitutive nuclear export. Thus, the inhibition of the PI3K-AKT pathway and nuclear translocation of endogenous FOXO3 may have therapeutic applications in the treatment of FOXO3-positive and cytoplasmic-type gastric cancer.
- Published
- 2021
25. Energy stress-induced linc01564 activates the serine synthesis pathway and facilitates hepatocellular carcinogenesis
- Author
-
Qingfa Wu, Guang Zhang, Yide Mei, Yang Yang, Zhongyu Wang, Bingyan Li, Yu Zhu, Kaiyue Liu, and Hao Hu
- Subjects
0301 basic medicine ,Cancer Research ,Carcinoma, Hepatocellular ,Carcinogenesis ,Biology ,Transfection ,medicine.disease_cause ,Serine ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Humans ,Phosphoglycerate dehydrogenase ,Molecular Biology ,Transcription factor ,Competing endogenous RNA ,Liver Neoplasms ,ATF4 ,Cancer ,medicine.disease ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,RNA, Long Noncoding - Abstract
Cancer cells undergo metabolic adaption to sustain their survival and growth under metabolic stress conditions, yet the underlying mechanism remains largely unclear. It is also not known if lncRNAs contribute to this metabolic adaption of cancer cells. Here we show that linc01564 is induced in response to glucose deprivation by the transcription factor ATF4. Linc01564 functions to facilitate hepatocellular carcinoma cell survival under glucose deprivation by activating the serine synthesis pathway. Mechanistically, linc01564 acts as a competing endogenous RNA for miR-107/103a-3p and attenuates the inhibitory effect of miR-107/103a-3p on PHGDH, the rate-limiting enzyme of the serine synthesis pathway, thereafter leading to increased PHGDH expression. Furthermore, linc01564 is able to promote hepatocellular carcinogenesis via PHGDH. Together, these findings suggest that linc01564 is an important player in the regulation of metabolic adaption of cancer cells and also implicate linc01564 as a potential therapeutic target for cancer.
- Published
- 2021
26. POU1F1 transcription factor induces metabolic reprogramming and breast cancer progression via LDHA regulation
- Author
-
Noemí Gómez-Lado, Manuel Macia, Efigenia Arias, Anxo Martinez-Ordoñez, Noemi Eiro, Tomás García-Caballero, Fabio Pereira, Samuel Seoane, Pablo Aguiar, Leandro Avila, Roman Perez-Fernandez, Francisco J. Vizoso, Universidade de Santiago de Compostela. Centro de Investigación en Medicina Molecular e Enfermidades Crónicas, Universidade de Santiago de Compostela. Departamento de Ciencias Forenses, Anatomía Patolóxica, Xinecoloxía e Obstetricia, e Pediatría, Universidade de Santiago de Compostela. Departamento de Ciencias Morfolóxicas, and Universidade de Santiago de Compostela. Departamento de Fisioloxía
- Subjects
0301 basic medicine ,Cancer Research ,Lactate dehydrogenase A ,Mice, Nude ,Biology ,Transfection ,Article ,Mice ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Immune system ,Genetics ,medicine ,Animals ,Humans ,education ,Molecular Biology ,Transcription factor ,Mice, Inbred BALB C ,Gene knockdown ,education.field_of_study ,L-Lactate Dehydrogenase ,Cancer ,Cellular Reprogramming ,medicine.disease ,Mechanisms of disease ,030104 developmental biology ,Anaerobic glycolysis ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,MCF-7 Cells ,Cancer research ,Heterografts ,Transcription Factor Pit-1 - Abstract
Metabolic reprogramming is considered hallmarks of cancer. Aerobic glycolysis in tumors cells has been well-known for almost a century, but specific factors that regulate lactate generation and the effects of lactate in both cancer cells and stroma are not yet well understood. In the present study using breast cancer cell lines, human primary cultures of breast tumors, and immune deficient murine models, we demonstrate that the POU1F1 transcription factor is functionally and clinically related to both metabolic reprogramming in breast cancer cells and fibroblasts activation. Mechanistically, we demonstrate that POU1F1 transcriptionally regulates the lactate dehydrogenase A (LDHA) gene. LDHA catalyzes pyruvate into lactate instead of leading into the tricarboxylic acid cycle. Lactate increases breast cancer cell proliferation, migration, and invasion. In addition, it activates normal-associated fibroblasts (NAFs) into cancer-associated fibroblasts (CAFs). Conversely, LDHA knockdown in breast cancer cells that overexpress POU1F1 decreases tumor volume and [18F]FDG uptake in tumor xenografts of mice. Clinically, POU1F1 and LDHA expression correlate with relapse- and metastasis-free survival. Our data indicate that POU1F1 induces a metabolic reprogramming through LDHA regulation in human breast tumor cells, modifying the phenotype of both cancer cells and fibroblasts to promote cancer progression This study was supported by FEDER/Ministerio de Ciencia, Innovación y Universidades- Agencia Estatal de Investigación-PGC2018-100776-B-I00 and from Conselleria de Cultura, Educación e Ordenacion Universitaria (GPC2014/001), AM-O was supported by an FPU grant (Ministerio de Educación—FPU14/00548) SI
- Published
- 2021
27. Epiregulin confers EGFR-TKI resistance via EGFR/ErbB2 heterodimer in non-small cell lung cancer
- Author
-
Kun Mi, Juan Zeng, Shiqi Ma, Yuan Ren, Bangrong Cao, Tingqing Chen, Wei Dai, Jinyi Lang, Lu Zhang, and Liping Luo
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Biology ,Epiregulin ,03 medical and health sciences ,0302 clinical medicine ,Gefitinib ,Carcinoma, Non-Small-Cell Lung ,Genetics ,medicine ,Humans ,Lung cancer ,Protein Kinase Inhibitors ,Molecular Biology ,Tumor microenvironment ,Gene knockdown ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,Apoptosis ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Erlotinib ,medicine.drug - Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are effective against non-small cell lung cancer (NSCLC) with EGFR-activating mutations. The mechanisms underlying EGFR-TKI resistance are not fully understood. This study aimed to analyze the effects of seven EGFR ligands on EGFR-TKI sensitivity in NSCLC cells and patients. Cells with EGFR E746-A750del mutation were treated with recombinant EGFR ligands, and analyzed for cell viability, proliferation, and apoptosis. shRNA knockdown of endogenous Epiregulin (EREG) or overexpression of exogenous EREG and immunofluorescence experiments were carried out. Public gene expression datasets were used for tumor microenvironment and clinical assessment. Among the EGFR ligands, EREG significantly diminished cellular sensitivity to TKIs and was associated with decreased response to erlotinib in NSCLC patients. EREG induced AKT phosphorylation and attenuated TKI-induced cellular apoptosis in an ErbB2-dependent manner. EREG induced the formation of the EGFR/ErbB2 heterodimer regardless of gefitinib treatment. However, overexpression or knockdown of EREG in cancer cells had little impact on TKI sensitivity. Single-cell RNA sequencing data revealed that EREG was predominantly expressed in macrophages in the tumor microenvironment. In addition, EREG-enriched macrophage conditional medium induced EGFR-TKI resistance. These findings shed new light on the mechanism underlying EGFR-TKI resistance, and suggest macrophage-produced intratumoral EREG as a novel regulator and biomarker for EGFR-TKI therapy in NSCLC.
- Published
- 2021
28. Oncogenic functions and therapeutic targeting of EphA2 in cancer
- Author
-
Eileen Shiuan, Kalin L. Wilson, and Dana M. Brantley-Sieders
- Subjects
0301 basic medicine ,Cancer Research ,EphA2 ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Neoplasms ,Genetics ,medicine ,cancer ,Humans ,Lung cancer ,Molecular Biology ,experimental therapeutics ,clinical trials ,Kinase ,Receptor, EphA2 ,Melanoma ,small molecule inhibitor ,Cancer ,Oncogenes ,medicine.disease ,targeting antibody ,ephrin ,Clinical trial ,Dasatinib ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,medicine.drug - Abstract
More than twenty-five years of research and pre-clinical validation have defined EphA2 receptor tyrosine kinase as a promising molecular target for clinical translation in cancer treatment. Molecular, genetic, biochemical, and pharmacological targeting strategies have been extensively tested in vitro and in vivo, and drugs like dasatinib, initially designed to target SRC family kinases, have been found to also target EphA2 activity. Other small molecules, therapeutic targeting antibodies, and peptide-drug conjugates are being tested, and more recently, approaches harnessing anti-tumor immunity against EphA2-expressing cancer cells have emerged as a promising strategy. This review will summarize pre-clinical studies supporting the oncogenic role of EphA2 in breast cancer, lung cancer, glioblastoma, and melanoma, while delineating the differing roles of canonical and noncanonical EphA2 signaling in each setting. This review also summarizes completed and ongoing clinical trials, highlighting the promise and challenges of targeting EphA2 in cancer.
- Published
- 2021
29. PSPH promotes melanoma growth and metastasis by metabolic deregulation-mediated transcriptional activation of NR4A1
- Author
-
Suresh Bugide, Romi Gupta, Xuchen Zhang, Deborah L. Della Manna, Eddy S. Yang, Vipin Rawat, Parmanand Malvi, and Narendra Wajapeyee
- Subjects
Transcriptional Activation ,Jumonji Domain-Containing Histone Demethylases ,Cancer Research ,Article ,Glutarates ,Small hairpin RNA ,Mice ,Cell Line, Tumor ,Gene expression ,Nuclear Receptor Subfamily 4, Group A, Member 1 ,Serine ,Genetics ,medicine ,Animals ,Humans ,RNA, Messenger ,Neoplasm Metastasis ,Melanoma ,Molecular Biology ,Cell Proliferation ,Histone Demethylases ,Gene knockdown ,biology ,medicine.disease ,Phosphoric Monoester Hydrolases ,Gene Expression Regulation, Neoplastic ,Histone ,Cell culture ,Gene Knockdown Techniques ,Cancer cell ,biology.protein ,Cancer research ,Heterografts ,H3K4me3 - Abstract
Metabolic deregulation, a hallmark of cancer, fuels cancer cell growth and metastasis. Here, we show that phosphoserine phosphatase (PSPH), an enzyme of the serine metabolism pathway, is upregulated in patient-derived melanoma samples. PSPH knockdown using short hairpin RNAs (shRNAs) blocks melanoma tumor growth and metastasis in both cell culture and mice. To elucidate the mechanism underlying PSPH action, we evaluated PSPH shRNA-expressing melanoma cells using global metabolomics and targeted mRNA expression profiling. Metabolomics analysis showed an increase in 2-hydroxyglutarate (2-HG) levels in PSPH knockdown cells. 2-HG inhibits the TET family of DNA demethylases and the Jumonji family of histone demethylases (KDM and JMJD), which is known to impact gene expression. Consistent with these data, PSPH knockdown in melanoma cells showed reduced DNA 5-hydroxymethylcytosine (5hmC) and increased histone H3K4me3 modifications. 2-HG treatment also inhibited melanoma growth. The nCounter PanCancer Pathways Panel-based mRNA expression profiling revealed attenuation of a number of cancer-promoting pathways upon PSPH knockdown. In particular, PSPH was necessary for nuclear receptor NR4A1 expression. Ectopic NR4A1 expression partly rescued the growth of melanoma cells expressing PSPH shRNA. Collectively, these results link PSPH to the facilitation of melanoma growth and metastasis through suppression of 2-HG and thus activation of pro-oncogenic gene expression.
- Published
- 2021
30. HuR up-regulates cell surface PD-L1 via stabilizing CMTM6 transcript in cancer
- Author
-
Yanbin Liu, Yanli Wei, Mingming Zhang, Hui Zhang, and Xingzhi Li
- Subjects
0301 basic medicine ,Cancer Research ,T-Lymphocytes ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Cell ,Naphthols ,B7-H1 Antigen ,Article ,ELAV-Like Protein 1 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Neoplasms ,PD-L1 ,Genetics ,medicine ,Animals ,Humans ,RNA, Messenger ,Furans ,3' Untranslated Regions ,Molecular Biology ,MARVEL Domain-Containing Proteins ,biology ,Immunity ,Cancer ,medicine.disease ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Heterografts ,Interleukin-2 ,Tumour immunology ,Antibody ,Myelin Proteins - Abstract
Despite the well-established role of CMTM6 in the stabilization of cell surface PD-L1 in cancer cells, the mechanisms underlying CMTM6 expression and regulation are still largely unknown. Here we unexpectedly find a strikingly positive correlation between CMTM6 and Hu-Antigen R (HuR) expression in most types of cancer. Mechanistically, we elucidate HuR stabilizes CMTM6 mRNA via direct association with AU-rich elements (AREs) in its 3′UTR and predominantly up-regulates CMTM6, which is readily abolished by HuR-specific inhibitor, MS-444. Phenotypically, we notice abundant cell surface PD-L1 in HuR-high cancer cells, which significantly inhibits immune activation of co-cultured T cells as indicated by IL-2 production. Treatment with MS-444 completely relieves immune suppression imposed by HuR-overexpression and further stimulates immune responses. Ectopic HuR accelerates allograft tumor progression in vivo, which is greatly compromised by simultaneous administration with MS-444. Our study uncovers a novel mechanism in control of CMTM6 and therefore PD-L1 expression, and suggests the potential of combining HuR inhibitor with PD-1/PD-L1 antibodies for cancer immunotherapy.
- Published
- 2021
31. IL-17B/IL-17RB signaling cascade contributes to self-renewal and tumorigenesis of cancer stem cells by regulating Beclin-1 ubiquitination
- Author
-
Hui Song, Xinke Chen, Rou Zhao, Lihua Zhang, Li Wei, Xiao Yang, Huabao Xiong, Shuo Shi, Bin Zhang, Qingli Bie, and Baogui Zhang
- Subjects
0301 basic medicine ,Cancer Research ,Carcinogenesis ,Angiogenesis ,Biology ,medicine.disease_cause ,Article ,Metastasis ,Gastrointestinal cancer ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,Cancer stem cell ,Autophagy ,Morphogenesis ,Genetics ,medicine ,Homeostasis ,Humans ,Cell Self Renewal ,Neoplasm Metastasis ,Molecular Biology ,Receptors, Interleukin-17 ,Neovascularization, Pathologic ,Cancer stem cells ,Interleukin-17 ,HEK 293 cells ,Ubiquitination ,Cell Differentiation ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Neoplastic Stem Cells ,Cancer research ,Beclin-1 ,Tumor necrosis factor alpha ,Signal Transduction - Abstract
Cancer stem cells (CSCs) are characterized by robust self-renewal and tumorigenesis and are responsible for metastasis, drug resistance, and angiogenesis. However, the molecular mechanisms for the regulation of CSC homeostasis are incompletely understood. This study demonstrated that the interleukin-17 (IL-17)B/IL-17RB signaling cascade promotes the self-renewal and tumorigenesis of CSCs by inducing Beclin-1 ubiquitination. We found that IL-17RB expression was significantly upregulated in spheroid cells and Lgr5-positive cells from the same tumor tissues of patients with gastric cancer (GC), which was closely correlated with the degree of cancer cell differentiation. Recombinant IL-17B (rIL-17B) promoted the sphere-formation ability of CSCs in vitro and enhanced tumor growth and metastasis in vivo. Interestingly, IL-17B induced autophagosome formation and cleavage-mediated transformation of LC3 in CSCs and 293T cells. Furthermore, inhibition of autophagy activation by ATG7 knockdown reversed rIL-17B-induced self-renewal of GC cells. In addition, we showed that IL-17B also promoted K63-mediated ubiquitination of Beclin-1 by mediating the binding of tumor necrosis factor receptor-associated factor 6 to Beclin-1. Silencing IL-17RB expression abrogated the effects of IL-17B on Beclin-1 ubiquitination and autophagy activation in GC cells. Finally, we showed that IL-17B level in the serum of GC patients was positively correlated with IL-17RB expression in GC tissues, and IL-17B could induce IL-17RB expression in GC cells. Overall, the results elucidate the novel functions of IL-17B for CSCs and suggest that the intervention of the IL-17B/IL-17RB signaling pathway may provide new therapeutic targets for the treatment of cancer.
- Published
- 2021
32. Mitochondrial uncoupler MB1-47 is efficacious in treating hepatic metastasis of pancreatic cancer in murine tumor transplantation models
- Author
-
Bin Cao, Shengkan Jin, Victor M. Tan, Amer Alasadi, Jingjing Guo, Juan Collantes, Hanlin Tao, Xiaoyang Su, and David Augeri
- Subjects
0301 basic medicine ,Cancer Research ,Citric Acid Cycle ,Adenocarcinoma ,Mitochondrion ,Biology ,Mice ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,Cell Line, Tumor ,Pancreatic cancer ,Pyruvic Acid ,Genetics ,medicine ,Animals ,Humans ,Glycolysis ,Molecular Biology ,Cell Proliferation ,Futile cycle ,Liver Neoplasms ,Cell Cycle Checkpoints ,Cell cycle ,medicine.disease ,Warburg effect ,Adenosine Monophosphate ,Mitochondria ,Adenosine Diphosphate ,Transplantation ,Disease Models, Animal ,Glucose ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Heterografts ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDA) is aggressive cancer characterized by rapid progression, metastatic recurrence, and highly resistant to treatment. PDA cells exhibit aerobic glycolysis, or the Warburg effect, which reduces the flux of pyruvate into mitochondria. As a result, more glycolytic metabolites are shunted to pathways for the production of building blocks (e.g., ribose) and reducing agents (e.g., NADPH) for biosynthesis that are necessary for cell proliferation. In addition, PDA cells are highly addicted to glutamine for both maintaining biosynthetic pathways and achieving redox balance. Mitochondrial uncoupling facilitates proton influx across the mitochondrial inner membrane without generating ATP, leading to a futile cycle that consumes glucose metabolites and glutamine. We synthesized a new mitochondrial uncoupler MB1-47 and tested its effect on cancer cell metabolism and the anticancer activity in pancreatic cancer cell models and murine tumor transplantation models. MB1-47 uncouples mitochondria in the pancreatic cancer cells, resulting in: (1) the acceleration of pyruvate oxidation and TCA turnover; (2) increases in AMP/ATP and ADP/AMP ratios; and (3) a decrease in the synthesis rate of nucleotides and sugar nucleotides. Moreover, MB1-47 arrests cell cycle at G0-G1 phase, reduces clonogenicity, and inhibits cell growth of murine and human pancreatic cancer cells. In vivo studies showed that MB1-47 inhibits tumor growth in murine tumor transplantation models, and inhibits the hepatic metastasis when tumor cells were transplanted intrasplenically. Our results provide proof of concept for a potentially new strategy of treating PDA, and a novel prototype experimental drug for future studies and development.
- Published
- 2021
33. Tumor-derived osteopontin drives the resident fibroblast to myofibroblast differentiation through Twist1 to promote breast cancer progression
- Author
-
Anupama Mane, Ramakrishna Nimma, Tushar Patil, Dhiraj Kumar, Satyajit S. Gill, Georg F. Weber, Totakura V. S. Kumar, Vinoth Prasanna Gunasekaran, Ramesh Butti, Deepti Tomar, Gautam Kundu, Anuradha Bulbule, and Gopal C. Kundu
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Stromal cell ,Carcinogenesis ,Breast Neoplasms ,Mammary Neoplasms, Animal ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,stomatognathic system ,Genetics ,medicine ,Animals ,Humans ,Osteopontin ,Epithelial–mesenchymal transition ,Myofibroblasts ,Molecular Biology ,biology ,Twist-Related Protein 1 ,CD44 ,Nuclear Proteins ,Cancer ,Cell Differentiation ,medicine.disease ,Chemokine CXCL12 ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,Carcinogens ,biology.protein ,Cancer research ,Heterografts ,Female ,Myofibroblast - Abstract
Tumor-stroma interactions are important determinants for the disease course in cancer. While stromal influence has been known to often play a tumor-promoting role, incomplete mechanistic insight into this phenomenon has prevented its therapeutic targeting. Stromal fibroblasts can be activated by tumor cells to differentiate into cancer-associated fibroblasts (CAFs), that exhibit the traits of myofibroblasts, and in turn, they increase cancer aggressiveness. Here, we report the crosstalk between the cancer cells and stromal fibroblasts that leads to tumor progression. The process is initiated by secretion of a chemokine like protein, osteopontin (OPN) from the cancer cells that differentiates the fibroblasts to myofibroblasts. Tumor-derived OPN achieves this transition by engaging CD44 and αvβ3 integrins on the fibroblast surface, which mediates signaling via Akt and ERK to induce Twist1-dependent gene expression. The OPN-driven CAFs then secrete CXCL12, which in turn triggers epithelial to mesenchymal transition (EMT) in the tumor cells. OPN, produced by the cancer cells, and CXCL12, secreted by activated fibroblasts, are necessary and sufficient to perpetuate the crosstalk. Knocking out OPN in carcinogen-induced mammary tumors or knocking down OPN in cancer cells and fibroblast co-implanted xenografts abrogates myofibroblast differentiation, Twist1, and CXCL12 expression. OPN expression is correlated with CAF-specific gene signature as shown by breast tumor tissue microarray consisting of 100 patient specimens. Bioinformatics analyses have confirmed that the expression of OPN is significantly correlated with the expression of myofibroblast-specific markers as demonstrated in human breast carcinoma dataset of 2509 patients. Our findings describe OPN and CXCL12 act as compelling targets to curb the tumor-promoting features of the stromal components and further suggested that OPN-regulated CXCL12 network might act as potential therapeutic target for the management of CAF-mediated breast cancer progression.
- Published
- 2021
34. Long-chain fatty acyl-CoA synthetase 1 promotes prostate cancer progression by elevation of lipogenesis and fatty acid beta-oxidation
- Author
-
Kaixiong Ye, Amelia Yin, Qianjin Li, Qingfu Zhang, Junyi Zha, Zanna Beharry, Yongjie Ma, Jiaoti Huang, Xiangkun Yang, Hanwen Huang, Houjian Cai, Hang Yin, and Michael G. Bartlett
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Biology ,Fatty acid beta-oxidation ,Mice ,03 medical and health sciences ,Prostate cancer ,chemistry.chemical_compound ,Adenosine Triphosphate ,0302 clinical medicine ,Biosynthesis ,Cell Line, Tumor ,Coenzyme A Ligases ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Cell Proliferation ,chemistry.chemical_classification ,Fatty acid metabolism ,Lipogenesis ,Fatty Acids ,Prostatic Neoplasms ,medicine.disease ,030104 developmental biology ,Enzyme ,Biochemistry ,chemistry ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,Heterografts ,lipids (amino acids, peptides, and proteins) ,Oxidation-Reduction - Abstract
Fatty acid metabolism is essential for the biogenesis of cellular components and ATP production to sustain proliferation of cancer cells. Long-chain fatty acyl-CoA synthetases (ACSLs), a group of rate-limiting enzymes in fatty acid metabolism, catalyze the bioconversion of exogenous or de novo synthesized fatty acids to their corresponding fatty acyl-CoAs. In this study, systematical analysis of ACSLs levels and the amount of fatty acyl-CoAs illustrated that ACSL1 were significantly associated with the levels of a broad spectrum of fatty acyl-CoAs, and were elevated in human prostate tumors. ACSL1 increased the biosynthesis of fatty acyl-CoAs including C16:0-, C18:0-, C18:1-, and C18:2-CoA, triglycerides and lipid accumulation in cancer cells. Mechanistically, ACSL1 modulated mitochondrial respiration, β-oxidation, and ATP production through regulation of CPT1 activity. Knockdown of ACSL1 inhibited the cell cycle, and suppressed the proliferation and migration of prostate cancer cells in vitro, and growth of prostate xenograft tumors in vivo. Our study implicates ACSL1 as playing an important role in prostate tumor progression, and provides a therapeutic strategy of targeting fatty acid metabolism for the treatment of prostate cancer.
- Published
- 2021
35. HACE1 blocks HIF1α accumulation under hypoxia in a RAC1 dependent manner
- Author
-
Amy Li, Daniel Martinez, Busra Turgu, Luigi Tortola, Christopher S. Hughes, Fan Zhang, Tony Ng, Poul H. Sorensen, Melanie Kogler, Fraser Johnson, Josef M. Penninger, John M. Maris, William W. Lockwood, Amal El-Naggar, Gian Luca Negri, Donald T. T. Yapp, and Mads Daugaard
- Subjects
0301 basic medicine ,HECT domain ,rac1 GTP-Binding Protein ,Cancer Research ,Lung Neoplasms ,Ubiquitin-Protein Ligases ,RAC1 ,Article ,Metastasis ,law.invention ,03 medical and health sciences ,Mice ,0302 clinical medicine ,law ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Molecular Biology ,chemistry.chemical_classification ,Mice, Knockout ,DNA ligase ,biology ,Protein Stability ,Tumor Suppressor Proteins ,Ubiquitination ,Cancer ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Ubiquitin ligase ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Suppressor ,Tumor Hypoxia ,Cell signalling ,Signal Transduction - Abstract
Uncovering the mechanisms that underpin how tumor cells adapt to microenvironmental stress is essential to better understand cancer progression. The HACE1 (HECT domain and ankyrin repeat-containing E3 ubiquitin-protein ligase) gene is a tumor suppressor that inhibits the growth, invasive capacity, and metastasis of cancer cells. However, the direct regulatory pathways whereby HACE1 confers this tumor-suppressive effect remain to be fully elucidated. In this report, we establish a link between HACE1 and the major stress factor, hypoxia-inducible factor 1 alpha (HIF1α). We find that HACE1 blocks the accumulation of HIF1α during cellular hypoxia through decreased protein stability. This property is dependent on HACE1 E3 ligase activity and loss of Ras-related C3 botulinum toxin substrate 1 (RAC1), an established target of HACE1 mediated ubiquitinylation and degradation. In vivo, genetic deletion of Rac1 reversed the increased HIF1α expression observed in Hace1–/– mice in murine KRasG12D-driven lung tumors. An inverse relationship was observed between HACE1 and HIF1α levels in tumors compared to patient-matched normal kidney tissues, highlighting the potential pathophysiological significance of our findings. Together, our data uncover a previously unrecognized function for the HACE1 tumor suppressor in blocking HIF1α accumulation under hypoxia in a RAC1-dependent manner., Oncogene, 40 (11), ISSN:0950-9232, ISSN:1476-5594
- Published
- 2021
36. Repurposing screen identifies Amlodipine as an inducer of PD-L1 degradation and antitumor immunity
- Author
-
Han Yao, Jie Xu, Jing-Yuan Fang, Chushu Li, and Huanbin Wang
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Autophagy ,Immunotherapy ,Biology ,Calcium in biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,PD-L1 ,Cancer cell ,Calcium flux ,Genetics ,Cancer research ,medicine ,biology.protein ,Amlodipine ,Molecular Biology ,medicine.drug - Abstract
Cancer cell expression of PD-L1 leads to T cells exhaustion by transducing co-inhibitory signal, and further understanding the regulation of PD-L1 in cancer cells may provide additional therapeutic strategies. Here by drug repurposing screen, we identified amlodipine as a potent inhibitor of PD-L1 expression in cancer cells. Further survey of calcium-associated pathways revealed calpain-dependent stabilization of the PD-L1 protein. Intracellular calcium delivered an operational signal to calpain-dependent Beclin-1 cleavage, blocking autophagic degradation of PD-L1 accumulated on recycling endosome (RE). Blocking calcium flux by amlodipine depleted PD-L1 expression and increased CD8+ T-cell infiltration in tumor tissues but not in myocardium, causing dose-dependent tumor suppression in vivo. Rescuing PD-L1 expression eliminated the effects of amlodipine, suggesting the PD-L1-dependent effect of amlodipine. These results reveal a calcium-dependent mechanism controlling PD-L1 degradation, and highlight calcium flux blockade as a potential strategy for combinatorial immunotherapy.
- Published
- 2020
37. Pten-NOLC1 fusion promotes cancers involving MET and EGFR signalings
- Author
-
Silvia Liu, Deqin Ma, James D. Luketich, Arjun Pennathur, Donald B. DeFranco, David F. Jarrard, Qi Chen, Shi Yuan Cheng, Satdarshan Paul Monga, Tianzhou Ma, Joel B. Nelson, Junyan Tao, Rohit Bhargava, Yanping Yu, Zhang-Hui Chen, Katherine L. Luo, Michael A. Nalesnik, George K. Michalopoulos, Jianhua Luo, George C. Tseng, Baoguo Ren, Jun Zhang, and Kathleen Cieply
- Subjects
0301 basic medicine ,Cancer Research ,Cell signaling ,Oncogene Proteins, Fusion ,Article ,Metastasis ,Fusion gene ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,PTEN ,Nuclear protein ,Molecular Biology ,Cell Proliferation ,biology ,Genome, Human ,Liver Neoplasms ,PTEN Phosphohydrolase ,Nuclear Proteins ,Cancer ,Proto-Oncogene Proteins c-met ,Phosphoproteins ,medicine.disease ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Heterografts ,Signal transduction ,Signal Transduction - Abstract
Inactivation of Pten gene through deletions and mutations leading to excessive pro-growth signaling pathway activations frequently occurs in cancers. Here, we report a Pten derived pro-cancer growth gene fusion Pten-NOLC1 originated from a chr10 genome rearrangement and identified through a transcriptome sequencing analysis of human cancers. Pten-NOLC1 fusion is present in primary human cancer samples and cancer cell lines from different organs. The product of Pten-NOLC1 is a nuclear protein that interacts and activates promoters of EGFR, c-MET, and their signaling molecules. Pten-NOLC1 promotes cancer proliferation, growth, invasion, and metastasis, and reduces the survival of animals xenografted with Pten-NOLC1-expressing cancer cells. Genomic disruption of Pten-NOLC1 induces cancer cell death, while genomic integration of this fusion gene into the liver coupled with somatic Pten deletion produces spontaneous liver cancers in mice. Our studies indicate that Pten-NOLC1 gene fusion is a driver for human cancers.
- Published
- 2020
38. VEGF-C mediates tumor growth and metastasis through promoting EMT-epithelial breast cancer cell crosstalk
- Author
-
Deguang Kong, Connor J. Hughes, Hengbo Zhou, Michael T. Lewis, Ramakrishnan R. Srinivasan, Deepika Neelakantan, Jessica Y. Hsu, and Heide L. Ford
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Vascular Endothelial Growth Factor C ,VEGF-C ,Breast Neoplasms ,Biology ,Article ,Metastasis ,03 medical and health sciences ,Paracrine signalling ,breast cancer ,0302 clinical medicine ,Breast cancer ,Cell Movement ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Hedgehog Proteins ,Secretion ,Neoplasm Metastasis ,Molecular Biology ,Cell Proliferation ,Homeodomain Proteins ,Regulation of gene expression ,EMT ,medicine.disease ,Hedgehog signaling pathway ,Neuropilin-2 ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer cell ,Cancer research ,Female ,GLI ,Signal Transduction - Abstract
It is well established that a subset of cells within primary breast cancers can undergo an epithelial-to-mesenchymal transition (EMT), although the role of EMT in metastasis remains controversial. We previously demonstrated that breast cancer cells that had undergone an oncogenic EMT could increase metastasis of neighboring cancer cells via non-canonical paracrine-mediated activation of GLI activity that is dependent on SIX1 expression in the EMT cancer cells. However, the mechanism by which these SIX1-expressing EMT cells activate GLI signaling remained unclear. In this study, we demonstrate a novel mechanism for activation of GLI-mediated signaling in epithelial breast tumor cells via EMT cell-induced production and secretion of VEGF-C. We show that VEGF-C, secreted by breast cancer cells that have undergone an EMT, promotes paracrine-mediated increases in proliferation, migration, and invasion of epithelial breast cancer cells, via non-canonical activation of GLI-signaling. We further show that the aggressive phenotypes, including metastasis, imparted by EMT cells on adjacent epithelial cancer cells can be disrupted by either inhibiting VEGF-C in EMT cells or by knocking down NRP2, a receptor which interacts with VEGF-C, in neighboring epithelial cancer cells. Interrogation of TCGA and GEO public datasets supports the relevance of this pathway in human breast cancer, demonstrating that VEGF-C strongly correlates with activation of Hedgehog signaling and EMT in the human disease. Our study suggests that the VEGF-C/NRP2/GLI axis is a novel and conserved paracrine means by which EMT cells enhance metastasis, and provides potential targets for therapeutic intervention in this heterogeneous disease.
- Published
- 2020
39. Nerve fibers in the tumor microenvironment in neurotropic cancer—pancreatic cancer and cholangiocarcinoma
- Author
-
Jan Bednarsch, Steven W.M. Olde Damink, Judith de Vos-Geelen, Merel R. Aberle, Jakob Nikolas Kather, Jan M. Niehues, Svetlana Kintsler, Marcel den Dulk, Liselot B.J. Valkenburg-van Iersel, Guangshan Hao, Xiuxiang Tan, Nadine T. Gaisa, Anjali A. Roeth, Jarne Koolen, Georg Wiltberger, Shivan Sivakumar, Marielle M.E. Coolsen, Sven Arke Lang, Lara R. Heij, and Ulf P. Neumann
- Subjects
EXPRESSION ,Cancer microenvironment ,0301 basic medicine ,GROWTH-FACTOR ,STELLATE CELLS ,Cancer Research ,Neurotropism ,Perineural invasion ,PROGRESSION ,Nerve fiber ,Review Article ,Adenocarcinoma ,Biology ,PERINEURAL INVASION ,NEUROGENESIS ,Cholangiocarcinoma ,03 medical and health sciences ,Nerve Fibers ,0302 clinical medicine ,Pancreatic cancer ,Tumor Microenvironment ,Genetics ,medicine ,Carcinoma ,INTRAHEPATIC CHOLANGIOCARCINOMA ,Humans ,PREDICTS POOR-PROGNOSIS ,CLINICAL-SIGNIFICANCE ,Molecular Biology ,Tumor microenvironment ,MUSCARINIC ACETYLCHOLINE-RECEPTOR ,Prognosis ,medicine.disease ,Combined Modality Therapy ,Crosstalk (biology) ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Carcinoma, Pancreatic Ductal ,Signal Transduction - Abstract
Oncogene : including Oncogene reviews 40(5), 899-908 (2021). doi:10.1038/s41388-020-01578-4, Published by Springer Nature, London
- Published
- 2020
40. BAP1 mutant uveal melanoma is stratified by metabolic phenotypes with distinct vulnerability to metabolic inhibitors
- Author
-
Andrew E. Aplin, David W. Speicher, Erin L. Seifert, Timothy J. Purwin, Anna Han, Zachary T. Schug, Vivian Chua, Nelisa Bechtel, Takami Sato, J. William Harbour, and Connie Liao
- Subjects
Uveal Neoplasms ,0301 basic medicine ,Cancer Research ,Mutant ,Biology ,Article ,Oxidative Phosphorylation ,Metastasis ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Gene expression ,Genetics ,medicine ,Humans ,Melanoma ,Molecular Biology ,BAP1 ,Tumor Suppressor Proteins ,Cancer ,medicine.disease ,Phenotype ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Cancer cell ,Cancer research ,Ubiquitin Thiolesterase - Abstract
Cancer cell metabolism is a targetable vulnerability; however, a precise understanding of metabolic heterogeneity is required. Inactivating mutations in BRCA1-associated protein 1 (BAP1) are associated with metastasis in uveal melanoma (UM), the deadliest adult eye cancer. BAP1 functions in UM remain unclear. UM patient sample analysis divided BAP1 mutant UM tumors into two subgroups based on oxidative phosphorylation (OXPHOS) gene expression suggesting metabolic heterogeneity. Consistent with patient data, transcriptomic analysis of BAP1 mutant UM cell lines also showed OXPHOS(high) or OXPHOS(low) subgroups. Integrated RNA sequencing, metabolomics and molecular analyses showed that OXPHOS(high) BAP1 mutant UM cells utilize glycolytic and nucleotide biosynthesis pathways, whereas OXPHOS(low) BAP1 mutant UM cells employ fatty acid oxidation. Furthermore, the two subgroups responded to different classes of metabolic suppressors. Our findings indicate that targeting cancer metabolism is a promising therapeutic option for BAP1 mutant UM; however, tailored approaches may be required to due metabolic heterogeneities.
- Published
- 2020
41. The p53/p73 - p21CIP1 tumor suppressor axis guards against chromosomal instability by restraining CDK1 in human cancer cells
- Author
-
Maik Kschischo, Ann-Kathrin Schmidt, Holger Bastians, Jan-Eric Boekenkamp, Karoline Pudelko, and Katharina Berger
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,0301 basic medicine ,Cancer Research ,Mitosis ,Aneuploidy ,Apoptosis ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Microtubule ,Chromosomal Instability ,Chromosome instability ,CDC2 Protein Kinase ,Cancer genomics ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Genetics ,medicine ,Humans ,neoplasms ,Molecular Biology ,Cell Proliferation ,Anaphase ,Tumor Protein p73 ,medicine.disease ,female genital diseases and pregnancy complications ,Microtubule plus-end ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Cancer cell ,Tumor Suppressor Protein p53 ,CDK inhibitor - Abstract
Whole chromosome instability (W-CIN) is a hallmark of human cancer and contributes to the evolvement of aneuploidy. W-CIN can be induced by abnormally increased microtubule plus end assembly rates during mitosis leading to the generation of lagging chromosomes during anaphase as a major form of mitotic errors in human cancer cells. Here, we show that loss of the tumor suppressor genes TP53 and TP73 can trigger increased mitotic microtubule assembly rates, lagging chromosomes, and W-CIN. CDKN1A, encoding for the CDK inhibitor p21CIP1, represents a critical target gene of p53/p73. Loss of p21CIP1 unleashes CDK1 activity which causes W-CIN in otherwise chromosomally stable cancer cells. Consequently, induction of CDK1 is sufficient to induce abnormal microtubule assembly rates and W-CIN. Vice versa, partial inhibition of CDK1 activity in chromosomally unstable cancer cells corrects abnormal microtubule behavior and suppresses W-CIN. Thus, our study shows that the p53/p73 - p21CIP1 tumor suppressor axis, whose loss is associated with W-CIN in human cancer, safeguards against chromosome missegregation and aneuploidy by preventing abnormally increased CDK1 activity.
- Published
- 2020
42. Mesenchymal stem cells-derived exosomal microRNA-139-5p restrains tumorigenesis in bladder cancer by targeting PRC1
- Author
-
Lihua Zhou, Xuecheng Yang, Li Zhang, Yuefeng Jia, and Xuemei Ding
- Subjects
0301 basic medicine ,Cancer Research ,Bladder cancer ,Mesenchymal stem cell ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Microvesicles ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,microRNA ,Cancer cell ,Genetics ,medicine ,Cancer research ,Gene silencing ,Carcinogenesis ,Molecular Biology - Abstract
microRNAs (miRNAs) can be delivered to tumor cells where they exert their function via mesenchymal stem cells (MSCs)-derived exosomes. This study investigated exosomal transfer of miR-139-5p to bladder cancer cells and their role in the regulation of tumorigenesis. The dysregulation of polycomb repressor complex 1 (PRC1) in bladder cancer was characterized by RNA quantification, and its functional significance in bladder cancer cells was identified by loss-of-function experiments. We predicted the miR-139-5p that could play a role in regulating PRC1, which was further verified using dual-luciferase reporter gene assay. Next, we altered the expression of miR-139-5p and PRC1 in bladder cancer cells to identify their functions in cancer progression. Bladder cancer cells were co-cultured with exosomes isolated from human umbilical cord mesenchymal stem cells (hUCMSCs) over-expressing miR-139-5p. The intercellular transfer of miR-139-5p along with in vitro and in vivo functions was determined using gain- and loss-of-function approaches. Our results revealed that PRC1 levels were increased in bladder cancer tissues and cells, and silencing PRC1 appeared to impede the cell proliferation, migration, and invasion potentials. In addition, miR-139-5p was observed to be down-regulated in bladder cancer, which targeted PRC1 and reduced its expression, hereby resulting in ameliorated tumorigenic characteristics of bladder cancer cells in vitro. Furthermore, we noted that miR-139-5p from hUCMSCs-derived exosomes could be transferred into bladder cancer cells to down-regulate the PRC1 expression. Moreover, hUCMSCs-derived exosomal miR-139-5p conferred a suppressive role on bladder cancer development in vitro and in vivo. These data together supported the tumor-inhibiting role of MSCs-derived exosomal miR-139-5p in bladder cancer, highlighting a promising therapeutic strategy.
- Published
- 2020
43. Metabolic reprogramming of ovarian cancer involves ACSL1-mediated metastasis stimulation through upregulated protein myristoylation
- Author
-
Wei Zhou, Kin Yip Tam, Shan Yu, Qingyu Zhang, Alice S.T. Wong, Terence C.W. Poon, Yufei Jiao, Leo T. O. Lee, Yaojun Ju, and Sally K. Y. To
- Subjects
Proteomics ,0301 basic medicine ,Cancer Research ,Carcinoma, Ovarian Epithelial ,Biology ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Coenzyme A Ligases ,Tumor Microenvironment ,Genetics ,medicine ,Animals ,Humans ,Protein myristoylation ,Neoplasm Metastasis ,Protein kinase A ,Molecular Biology ,Myristoylation ,Ovarian Neoplasms ,Tumor microenvironment ,Fatty Acids ,Prognosis ,medicine.disease ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,030220 oncology & carcinogenesis ,Lipidomics ,Cancer cell ,Cancer research ,Female ,Signal transduction ,Neoplasm Transplantation ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
As a result of the hostile microenvironment, metabolic alterations are required to enable the malignant growth of cancer cells. To understand metabolic reprogramming during metastasis, we conducted shotgun proteomic analysis of highly metastatic (HM) and non-metastatic (NM) ovarian cancer cells. The results suggest that the genes involved in fatty-acid (FA) metabolism are upregulated, with consequent increases of phospholipids with relatively short FA chains (myristic acid, MA) in HM cells. Among the upregulated proteins, ACSL1 expression could convert the lipid profile of NM cells to that similar of HM cells and make them highly aggressive. Importantly, we demonstrated that ACSL1 activates the AMP-activated protein kinase and Src pathways via protein myristoylation and finally enhances FA beta oxidation. Patient samples and tissue microarray data also suggested that omentum metastatic tumours have higher ACSL1 expression than primary tumours and a strong association with poor clinical outcome. Overall, our data reveal that ACSL1 enhances cancer metastasis by regulating FA metabolism and myristoylation.
- Published
- 2020
44. Multi-color clonal tracking reveals intra-stage proliferative heterogeneity during mammary tumor progression
- Author
-
James Canales Murillo, Ravi Kiran Reddy Kalathur, Mathias Hess, Barbara Maria Szczerba, Stefanie Tiede, Benjamin Müller, Fabiana Lüönd, Gerhard Christofori, Tatjana Vlajnic, Nicola Aceto, and Luca von Allmen
- Subjects
0301 basic medicine ,Cancer Research ,Mammary tumor ,Cancer ,Biology ,medicine.disease ,Somatic evolution in cancer ,Metastatic breast cancer ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Mammary tumor virus ,030220 oncology & carcinogenesis ,Cancer cell ,Genetics ,medicine ,Cancer research ,Molecular Biology - Abstract
Despite major progress in breast cancer research, the functional contribution of distinct cancer cell clones to malignant tumor progression and metastasis remains largely elusive. We have assessed clonal heterogeneity within individual primary tumors and metastases and also during the distinct stages of malignant tumor progression using clonal tracking of cancer cells in the MMTV-PyMT mouse model of metastatic breast cancer. Comparative gene expression analysis of clonal subpopulations reveals a substantial level of heterogeneity across and also within the various stages of breast carcinogenesis. The intra-stage heterogeneity is primarily manifested by differences in cell proliferation, also found within invasive carcinomas of luminal A-, luminal B-, and HER2-enriched human breast cancer. Surprisingly, in the mouse model of clonal tracing of cancer cells, chemotherapy mainly targets the slow-proliferative clonal populations and fails to efficiently repress the fast-proliferative populations. These insights may have considerable impact on therapy selection and response in breast cancer patients.
- Published
- 2020
45. Ephrin-B2–EphB4 communication mediates tumor–endothelial cell interactions during hematogenous spread to spinal bone in a melanoma metastasis model
- Author
-
Thomas Broggini, Ralf H. Adams, Marcus Czabanka, Christian J. Hoffmann, Andras Piffko, Peter Vajkoczy, Christoph Harms, and Adnan Ghori
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Skin Neoplasms ,Intravital Microscopy ,Receptor, EphB2 ,Receptor, EphB4 ,Melanoma, Experimental ,Cell Communication ,Biology ,Bone tissue ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,Spinal cord compression ,Cell Line, Tumor ,Cell Adhesion ,Genetics ,medicine ,Animals ,Cell adhesion ,Molecular Biology ,Mice, Knockout ,Microscopy, Video ,Osteoblasts ,Spinal Neoplasms ,Melanoma ,Skull ,Endothelial Cells ,Bone metastasis ,Cancer ,medicine.disease ,Magnetic Resonance Imaging ,Spine ,Extravasation ,Pyrimidines ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Pyrazoles ,Female ,Spinal Cord Compression - Abstract
Metastases account for the majority of cancer deaths. Bone represents one of the most common sites of distant metastases, and spinal bone metastasis is the most common source of neurological morbidity in cancer patients. During metastatic seeding of cancer cells, endothelial-tumor cell interactions govern extravasation to the bone and potentially represent one of the first points of action for antimetastatic treatment. The ephrin-B2-EphB4 pathway controls cellular interactions by inducing repulsive or adhesive properties, depending on forward or reverse signaling. Here, we report that in an in vivo metastatic melanoma model, ephrin-B2-mediated activation of EphB4 induces tumor cell repulsion from bone endothelium, translating in reduced spinal bone metastatic loci and improved neurological function. Selective ephrin-B2 depletion in endothelial cells or EphB4 inhibition increases bone metastasis and shortens the time window to hind-limb locomotion deficit from spinal cord compression. EphB4 overexpression in melanoma cells ameliorates the metastatic phenotype and improves neurological outcome. Timely harvesting of bone tissue after tumor cell injection and intravital bone microscopy revealed less tumor cells attached to ephrin-B2-positive endothelial cells. These results suggest that ephrin-B2-EphB4 communication influences bone metastasis formation by altering melanoma cell repulsion/adhesion to bone endothelial cells, and represents a molecular target for therapeutic intervention.
- Published
- 2020
46. Multifaceted role of branched-chain amino acid metabolism in cancer
- Author
-
Hui Peng, Yingfei Wang, and Weibo Luo
- Subjects
0301 basic medicine ,Cancer Research ,Carcinogenesis ,Nitrogen ,Branched-chain amino acid ,Glutamic Acid ,Biology ,BCAT ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Valine ,Neoplasms ,Tumor Microenvironment ,Genetics ,medicine ,Animals ,Humans ,metabolic reprogramming ,BCAA ,Molecular Biology ,chemistry.chemical_classification ,Cancer ,BCKDH ,Metabolism ,medicine.disease ,cancer progression ,Amino acid ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,Metabolic pathway ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,Cancer research ,Ketoglutaric Acids ,Leucine ,Reactive Oxygen Species ,Amino Acids, Branched-Chain ,Metabolic Networks and Pathways - Abstract
Metabolic reprogramming fulfils increased nutrient demands and regulates numerous oncogenic processes in tumors, leading to tumor malignancy. Branched-chain amino acids (BCAAs, i.e., valine, leucine, and isoleucine) function as nitrogen donors to generate macromolecules such as nucleotides and are indispensable for human cancer cell growth. The cell-autonomous and non-autonomous roles of altered BCAA metabolism have been implicated in cancer progression and the key proteins in the BCAA metabolic pathway serve as possible prognostic and diagnostic biomarkers in human cancers. Here we summarize how BCAA metabolic reprogramming is regulated in cancer cells and how it influences cancer progression.
- Published
- 2020
47. RSK2 protects human breast cancer cells under endoplasmic reticulum stress through activating AMPKα2-mediated autophagy
- Author
-
Dong-Sheng Cao, Xi-Sha Chen, Kuansong Wang, Jin-Ming Yang, Yi-Di Guan, Lan-ya Li, Xin-luan Wang, Yongguang Tao, Yi Zhang, Minghua Wu, Mingzhu Yin, Xin-Yuan Sun, Xingcong Ren, Yan Cheng, and Ao-Xue Li
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Paclitaxel ,MAP Kinase Signaling System ,Antineoplastic Agents ,Apoptosis ,Breast Neoplasms ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Biology ,Ribosomal Protein S6 Kinases, 90-kDa ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Endoribonucleases ,Autophagy ,Genetics ,medicine ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Cell Nucleus ,Gene knockdown ,Endoplasmic reticulum ,Cancer ,Endoplasmic Reticulum Stress ,medicine.disease ,Xenograft Model Antitumor Assays ,Cell biology ,030104 developmental biology ,Drug Resistance, Neoplasm ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Cancer cell ,MCF-7 Cells ,Unfolded protein response ,Female - Abstract
Autophagy can protect stressed cancer cell by degradation of damaged proteins and organelles. However, the regulatory mechanisms behind this cellular process remain incompletely understood. Here, we demonstrate that RSK2 (p90 ribosomal S6 kinase 2) plays a critical role in ER stress-induced autophagy in breast cancer cells. We demonstrated that the promotive effect of RSK2 on autophagy resulted from directly binding of AMPKα2 in nucleus and phosphorylating it at Thr172 residue. IRE1α, an ER membrane-associated protein mediating unfolded protein response (UPR), is required for transducing the signal for activation of ERK1/2-RSK2 under ER stress. Suppression of autophagy by knockdown of RSK2 enhanced the sensitivity of breast cancer cells to ER stress both in vitro and in vivo. Furthermore, we demonstrated that inhibition of RSK2-mediated autophagy rendered breast cancer cells more sensitive to paclitaxel, a chemotherapeutic agent that induces ER stress-mediated cell death. This study identifies RSK2 as a novel controller of autophagy in tumor cells and suggests that targeting RSK2 can be exploited as an approach to reinforce the efficacy of ER stress-inducing agents against cancer.
- Published
- 2020
48. LncRNA SLC26A4-AS1 suppresses the MRN complex-mediated DNA repair signaling and thyroid cancer metastasis by destabilizing DDX5
- Author
-
Nasha Zhang, Changming An, Jiandong Liu, Yankang Li, Wenting Pan, Yue Shen, Yeyang Xu, Jupeng Yuan, Ming Yang, Yemei Song, Hui Hua, Mengyu Xie, and Bowen Wang
- Subjects
Male ,0301 basic medicine ,Cancer Research ,DNA Repair ,Thyroid Gland ,Cell Cycle Proteins ,Kaplan-Meier Estimate ,Metastasis ,DEAD-box RNA Helicases ,Tripartite Motif Proteins ,Mice ,0302 clinical medicine ,E2F1 ,DNA Breaks, Double-Stranded ,RNA-Seq ,Thyroid cancer ,MRE11 Homologue Protein ,Protein Stability ,Nuclear Proteins ,Middle Aged ,Prognosis ,Acid Anhydride Hydrolases ,Ubiquitin ligase ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Gene Knockdown Techniques ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,Female ,RNA, Long Noncoding ,Signal Transduction ,Adult ,DNA repair ,Ubiquitin-Protein Ligases ,Down-Regulation ,Biology ,03 medical and health sciences ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Gene silencing ,Thyroid Neoplasms ,Molecular Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,MRN complex ,Cancer cell ,biology.protein ,Cancer research ,Lymph Nodes ,E2F1 Transcription Factor ,Transcription Factors - Abstract
Lymph node metastasis is the major adverse feature for recurrence and death of thyroid cancer patients. To identify lncRNAs involved in thyroid cancer metastasis, we systemically screened differentially expressed lncRNAs in lymph node metastasis, thyroid cancer, and normal tissues via RNAseq. We found that lncRNA SLC26A4-AS1 was continuously, significantly down-regulated in normal tissues, thyroid cancer, and lymph node metastasis specimens. Low SLC26A4-AS1 levels in tissues were significantly associated with poor prognosis of thyroid cancer patients. LncRNA SLC26A4-AS1 markedly inhibited migration, invasion, and metastasis capability of cancer cells in vitro and in vivo. Intriguingly, SLC26A4-AS1 could simultaneously interact with DDX5 and the E3 ligase TRIM25, which promoting DDX5 degradation through the ubiquitin-proteasome pathway. In particular, SLC26A4-AS1 inhibited expression of multiple DNA double-strand breaks (DSBs) repair genes, especially genes coding proteins in the MRE11/RAS50/NBS1 (MRN) complex. Enhanced interaction between DDX5 and transcriptional factor E2F1 due to silencing of SLC26A4-AS1 promoted binding of the DDX5-E2F1 complex at promoters of the MRN genes and, thus, stimulate the MRN/ATM dependent DSB signaling and thyroid cancer metastasis. Our study uncovered new insights into the biology driving thyroid cancer metastasis and highlights potentials of lncRNAs as future therapeutic targets again cancer metastasis.
- Published
- 2020
49. High-metastatic cancer cells derived exosomal miR92a-3p promotes epithelial-mesenchymal transition and metastasis of low-metastatic cancer cells by regulating PTEN/Akt pathway in hepatocellular carcinoma
- Author
-
Changbiao Li, Jian Wu, Wenxuan Wu, Hua Liu, Rongliang Tong, Xiaode Feng, Junru Chen, Qiyang Cheng, Yunhao Chen, Xianlei Cai, Jingbang Wu, Shusen Zheng, Zhen Lv, Yuejie Lu, Hanxi Yu, Kejiong Liang, Jiating Hu, and Beng Yang
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Exosomes ,Metastasis ,Mice ,0302 clinical medicine ,Cell Movement ,Neoplasm Metastasis ,Promoter Regions, Genetic ,biology ,Liver Neoplasms ,Middle Aged ,Prognosis ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Female ,Signal Transduction ,Carcinoma, Hepatocellular ,Epithelial-Mesenchymal Transition ,Article ,Disease-Free Survival ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,PTEN ,Epithelial–mesenchymal transition ,neoplasms ,Molecular Biology ,PI3K/AKT/mTOR pathway ,PTEN Phosphohydrolase ,Cancer ,Oncogenes ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,Microvesicles ,MicroRNAs ,030104 developmental biology ,MRNA Sequencing ,Cancer cell ,Cancer research ,biology.protein ,Proto-Oncogene Proteins c-akt ,E2F1 Transcription Factor - Abstract
Exosomes play an important role in intercellular communication and metastatic progression of hepatocellular carcinoma (HCC). However, cellular communication between heterogeneous HCC cells with different metastatic potentials and the resultant cancer progression are not fully understood in HCC. Here, HCC cells with high-metastatic capacity (97hm and Huhm) were constructed by continually exerting selective pressure on primary HCC cells (MHCC-97H and Huh7). Through performing exosomal miRNA sequencing in HCC cells with different metastatic potentials (MHCC-97H and 97hm), many significantly different miRNA candidates were found. Among these miRNAs, miR-92a-3p was the most abundant miRNA in the exosomes of highly metastatic HCC cells. Exosomal miR92a-3p was also found enriched in the plasma of HCC patient-derived xenograft mice (PDX) model with high-metastatic potential. Exosomal miR-92a-3p promotes epithelial-mesenchymal transition (EMT) in recipient cancer cells via targeting PTEN and regulating its downstream Akt/Snail signaling. Furthermore, through mRNA sequencing in HCC cells with different metastatic potentials and predicting potential transcription factors of miR92a-3p, upregulated transcript factors E2F1 and c-Myc were found in high-metastatic HCC cells promote the expression of cellular and exosomal miR-92a-3p in HCC by directly binding the promoter of its host gene, miR17HG. Clinical data showed that a high plasma exosomal miR92a-3p level was correlated with shortened overall survival and disease-free survival, indicating poor prognosis in HCC patients. In conclusion, hepatoma-derived exosomal miR92a-3p plays a critical role in the EMT progression and promoting metastasis by inhibiting PTEN and activating Akt/Snail signaling. Exosomal miR92a-3p is a potential predictive biomarker for HCC metastasis, and this may provoke the development of novel therapeutic and preventing strategies against metastasis of HCC.
- Published
- 2020
50. The cell-surface anchored serine protease TMPRSS13 promotes breast cancer progression and resistance to chemotherapy
- Author
-
Lauren M. Tanabe, Carly E. Martin, Karin List, Thomas E. Hyland, Jacob R. Mackinder, Fausto A. Varela, Andrew S. Murray, and Kimberley E. Sala-Hamrick
- Subjects
0301 basic medicine ,Cancer Research ,Proteases ,Cell Survival ,medicine.medical_treatment ,Datasets as Topic ,Apoptosis ,Triple Negative Breast Neoplasms ,Biology ,Article ,Targeted therapy ,Extracellular matrix ,Mice ,03 medical and health sciences ,Mammary Glands, Animal ,0302 clinical medicine ,Breast cancer ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Genetics ,medicine ,Animals ,Humans ,Gene silencing ,Breast ,Molecular Biology ,Mice, Knockout ,Carcinoma, Ductal, Breast ,Serine Endopeptidases ,Mammary Neoplasms, Experimental ,Membrane Proteins ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Drug Resistance, Neoplasm ,Tumor progression ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Zymogen activation ,Cancer cell ,Disease Progression ,Cancer research ,Female - Abstract
Breast cancer progression is accompanied by increased expression of extracellular and cell-surface proteases capable of degrading the extracellular matrix as well as cleaving and activating downstream targets. The type II transmembrane serine proteases (TTSPs) are a family of cell-surface proteases that play critical roles in numerous types of cancers. Therefore, the aim of this study was to identify novel and uncharacterized TTSPs with differential expression in breast cancer and to determine their potential roles in progression. Systematic in silico data analysis followed by immunohistochemical validation identified increased expression of the TTSP family member, TMPRSS13 (transmembrane protease, serine 13), in invasive ductal carcinoma patient tissue samples compared to normal breast tissue. To test whether loss of TMPRSS13 impacts tumor progression, TMPRSS13 was genetically ablated in the oncogene-induced transgenic MMTV-PymT tumor model. TMPRSS13 deficiency resulted in a significant decrease in overall tumor burden and growth rate, as well as a delayed formation of detectable mammary tumors, thus suggesting a causal relationship between TMPRSS13 expression and the progression of breast cancer. Complementary studies using human breast cancer cell culture models revealed that siRNA-mediated silencing of TMPRSS13 expression decreases proliferation, induces apoptosis, and attenuates invasion. Importantly, targeting TMPRSS13 expression renders aggressive triple-negative breast cancer cell lines highly responsive to chemotherapy. At the molecular level, knockdown of TMPRSS13 in breast cancer cells led to increased protein levels of the tumor-suppressive protease prostasin. TMPRSS13/prostasin co-immunoprecipitation and prostasin zymogen activation experiments identified prostasin as a potential novel target for TMPRSS13. Regulation of prostasin levels may be a mechanism that contributes to the pro-oncogenic properties of TMPRSS13 in breast cancer. TMPRSS13 represents a novel candidate for targeted therapy in combination with standard of care chemotherapy agents in patients with hormone receptor-negative breast cancer or in patients with tumors refractory to endocrine therapy.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.