Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Nature ; 538(7623): 109-113, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27680705

ABSTRACT

Cancer stem cells (CSCs) may be responsible for tumour dormancy, relapse and the eventual death of most cancer patients. In addition, these cells are usually resistant to cytotoxic conditions. However, very little is known about the biology behind this resistance to therapeutics. Here we investigated stem-cell death in the digestive system of adult Drosophila melanogaster. We found that knockdown of the coat protein complex I (COPI)-Arf79F (also known as Arf1) complex selectively killed normal and transformed stem cells through necrosis, by attenuating the lipolysis pathway, but spared differentiated cells. The dying stem cells were engulfed by neighbouring differentiated cells through a draper-myoblast city-Rac1-basket (also known as JNK)-dependent autophagy pathway. Furthermore, Arf1 inhibitors reduced CSCs in human cancer cell lines. Thus, normal or cancer stem cells may rely primarily on lipid reserves for energy, in such a way that blocking lipolysis starves them to death. This finding may lead to new therapies that could help to eliminate CSCs in human cancers.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Lipolysis/physiology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , ADP-Ribosylation Factor 1/antagonists & inhibitors , ADP-Ribosylation Factor 1/deficiency , Animals , Apoptosis , Autophagy , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Coat Protein Complex I/deficiency , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drug Resistance, Neoplasm/drug effects , Energy Metabolism , Enterocytes/cytology , Female , Gastrointestinal Tract/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lipolysis/drug effects , MAP Kinase Signaling System , Male , Membrane Proteins/metabolism , Necrosis/chemically induced , Neoplastic Stem Cells/drug effects , Phagocytosis , rac GTP-Binding Proteins/metabolism
2.
Br J Cancer ; 118(10): 1337-1348, 2018 05.
Article in English | MEDLINE | ID: mdl-29717200

ABSTRACT

BACKGROUND: Chemerin, a known chemoattractant, participates in multiple biological events. However, its role in cancer remains largely unknown. METHODS: Chemerin expression was evaluated by real-time PCR, western blot and immunohistochemistry. Forced expression, RNAi, immunoprecipitation, etc. were used in function and mechanism study. Mouse models of extrahepatic and intrahepatic metastasis were employed to evaluate the therapeutic potential of chemerin. RESULTS: Chemerin expression was significantly downregulated in hepatocellular carcinoma, and associated with poor prognosis of HCC patients. Forced expression of chemerin inhibited in vitro migration, invasion and in vivo metastasis of HCC cells. Administration of chemerin effectively suppressed extrahepatic and intrahepatic metastases of HCC cells, resulting in prolonged survival of tumour-bearing nude mice. Chemerin upregulated expression and phosphatase activity of PTEN by interfering with PTEN-CMKLR1 interaction, leading to weakened ubiquitination of PTEN and decreased p-Akt (Ser473) level, which was responsible for suppressed migration, invasion and metastasis of HCC cells. Positive correlation between chemerin and PTEN, and reverse correlation between chemerin and p-Akt (Ser473) were also observed in HCC clinical samples and intrahepatic mouse model in vivo. CONCLUSIONS: Our study has revealed the suppressive role and therapeutic potential of chemerin in HCC metastasis, providing both a prognostic marker and drug candidate for HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Chemokines/administration & dosage , Intercellular Signaling Peptides and Proteins/administration & dosage , Liver Neoplasms/drug therapy , PTEN Phosphohydrolase/genetics , Receptors, Chemokine/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Movement , Cell Proliferation , Chemokines/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Oncogene Protein v-akt/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
PLoS Genet ; 11(12): e1005750, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26714316

ABSTRACT

In adult Drosophila testis, asymmetric division of germline stem cells (GSCs) is specified by an oriented spindle and cortically localized adenomatous coli tumor suppressor homolog 2 (Apc2). However, the molecular mechanism underlying these events remains unclear. Here we identified Megator (Mtor), a nuclear matrix protein, which regulates GSC maintenance and asymmetric division through the spindle assembly checkpoint (SAC) complex. Loss of Mtor function results in Apc2 mis-localization, incorrect centrosome orientation, defective mitotic spindle formation, and abnormal chromosome segregation that lead to the eventual GSC loss. Expression of mitotic arrest-deficient-2 (Mad2) and monopolar spindle 1 (Mps1) of the SAC complex effectively rescued the GSC loss phenotype associated with loss of Mtor function. Collectively our results define a new role of the nuclear matrix-SAC axis in regulating stem cell maintenance and asymmetric division.


Subject(s)
Asymmetric Cell Division , Drosophila Proteins/metabolism , Drosophila/genetics , M Phase Cell Cycle Checkpoints , Nuclear Matrix-Associated Proteins/metabolism , Stem Cells/metabolism , Testis/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Chromosome Segregation , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/genetics , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Male , Nuclear Matrix-Associated Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Stem Cells/physiology , Testis/cytology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
4.
Hepatology ; 62(6): 1791-803, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26257239

ABSTRACT

UNLABELLED: Sorafenib is a specific adenosine triphosphate-competitive RAF inhibitor used as a first-line treatment of advanced hepatocellular carcinoma (HCC). However, the responses are variable, reflecting heterogeneity of the disease, while the resistance mechanism remains poorly understood. Here, we report that sorafenib treatment can exacerbate disease progression in both patient-derived xenografts and cell line-derived xenografts and that the therapeutic effect of the drug inversely covaries to the ratio of epithelial cell adhesion molecule-positive cells, which may be tumor initiating cells in HCC. The TSC2-AKT cascade mediates this sorafenib resistance. In response to sorafenib treatment, formation of the TSC1/2 complex is enhanced, causing increased phosphorylation of AKT, which contributes to up-regulation of "stemness"-related genes in epithelial cell adhesion molecule-positive cells and enhancement of tumorigenicity. The expression of TSC2 negatively correlated with prognosis in clinical sorafenib therapy. Furthermore, all-trans retinoic acid decreased AKT activity, reduced the epithelial cell adhesion molecule-positive cell population enriched by sorafenib, and potentiated the therapeutic effect of sorafenib in the patient-derived xenograft model. CONCLUSION: Our findings suggest that a subtype of HCC is not suitable for sorafenib therapy; this resistance to sorafenib can be predicted by the status of TSC2, and agents inducing differentiation of tumor initiating cells (e.g., all-trans retinoic acid) should improve the prognosis of this subtype of HCC.


Subject(s)
Antigens, Neoplasm/drug effects , Antineoplastic Agents/adverse effects , Carcinoma, Hepatocellular/chemically induced , Cell Adhesion Molecules/drug effects , Liver Neoplasms/chemically induced , Neoplastic Stem Cells/drug effects , Niacinamide/analogs & derivatives , Oncogene Protein v-akt/physiology , Phenylurea Compounds/adverse effects , Tumor Suppressor Proteins/physiology , Animals , Carcinoma, Hepatocellular/classification , Disease Progression , Epithelial Cell Adhesion Molecule , Humans , Liver Neoplasms/classification , Mice , Niacinamide/adverse effects , Sorafenib , Tuberous Sclerosis Complex 2 Protein
5.
J Natl Cancer Cent ; 4(1): 25-35, 2024 Mar.
Article in English | MEDLINE | ID: mdl-39036388

ABSTRACT

Hepatic stellate cells (HSCs), a distinct category of non-parenchymal cells in the liver, are critical for liver homeostasis. In healthy livers, HSCs remain non-proliferative and quiescent. However, under conditions of acute or chronic liver damage, HSCs are activated and participate in the progression and regulation of liver diseases such as liver fibrosis, cirrhosis, and liver cancer. Fatty liver diseases (FLD), including nonalcoholic (NAFLD) and alcohol-related (ALD), are common chronic inflammatory conditions of the liver. These diseases, often resulting from multiple metabolic disorders, can progress through a sequence of inflammation, fibrosis, and ultimately, cancer. In this review, we focused on the activation and regulatory mechanism of HSCs in the context of FLD. We summarized the molecular pathways of activated HSCs (aHSCs) in mediating FLD and their role in promoting liver tumor development from the perspectives of cell proliferation, invasion, metastasis, angiogenesis, immunosuppression, and chemo-resistance. We aimed to offer an in-depth discussion on the reciprocal regulatory interactions between FLD and HSC activation, providing new insights for researchers in this field.

6.
J Biol Chem ; 287(11): 7845-58, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22262830

ABSTRACT

Non-small-cell lung cancer (NSCLC) is a deadly disease due to lack of effective diagnosis biomarker and therapeutic target. Much effort has been made in defining gene defects in NSCLC, but its full molecular pathogenesis remains unexplored. Here, we found RACK1 (receptor of activated kinase 1) was elevated in most NSCLC, and its expression level correlated with key pathological characteristics including tumor differentiation, stage, and metastasis. In addition, RACK1 activated sonic hedgehog signaling pathway by interacting with and activating Smoothened to mediate Gli1-dependent transcription in NSCLC cells. And silencing RACK1 dramatically inhibited in vivo tumor growth and metastasis by blocking the sonic hedgehog signaling pathway. These results suggest that RACK1 represents a new promising diagnosis biomarker and therapeutic target for NSCLC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , GTP-Binding Proteins/metabolism , Hedgehog Proteins/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Receptors for Activated C Kinase , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor , Transcription Factors/metabolism , Transcription, Genetic , Transplantation, Heterologous , Zinc Finger Protein GLI1
7.
J Hepatol ; 59(6): 1255-63, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23867314

ABSTRACT

BACKGROUND & AIMS: Systemic chemotherapy serves as an adjuvant treatment for post-operation patients with hepatocellular carcinoma (HCC), and provides curative option for the patients with unresectable HCC. However, its efficiency is largely limited because of the high incidence of chemo-resistance. Increasing evidence has shown that tumor initiating cells (TICs) not only have the ability to self-renew and drive the initiation and progression of cancer, but also exhibit greater resistance to conventional chemo- and radio-therapies than non-TICs. It was the aim of this study to investigate the effects of ATRA with and without cisplatin on TIC differentiation and apoptosis in human HCC. METHODS: In the present study, we evaluated the TICs of HCC cell differentiation induced by all-trans retinoic acid (ATRA), and developed a novel chemotherapeutic approach to HCC, by characterizing the function of combinatorial treatment with cis-diammineplatinum(II) (cisplatin) and ATRA in vitro and in vivo. RESULTS: ATRA effectively induced differentiation of TICs, which potentiated the cytotoxic effects of cisplatin. The combinatorial treatment of ATRA acid and cisplatin reduced protein kinase B (AKT) (Thr308) phosphorylation, and promoted apoptosis of HCC cells more significantly than treatment with cisplatin alone. In addition, the combined treatment with the two drugs exerted stronger inhibition on either HCC cell migration in vitro or metastasis in vivo, when compared to the treatment with either drug alone. CONCLUSIONS: These results indicated that ATRA could significantly improve the effect of cisplatin, which is at least partially attributed to ATRA-induced differentiation of HCC TICs, and the subsequent decrease in this chemo-resistant subpopulation.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cisplatin/pharmacology , Liver Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Tretinoin/pharmacology , Animals , Antigens, Neoplasm/physiology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/physiology , Cell Differentiation/drug effects , Drug Synergism , Epithelial Cell Adhesion Molecule , Humans , Liver Neoplasms/pathology , Male , Mice , Neoplastic Stem Cells/cytology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
8.
J Hepatol ; 56(2): 389-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21888875

ABSTRACT

BACKGROUND & AIMS: Portal vein tumor thrombus (PVTT) has previously been demonstrated to correlate with poor prognosis of hepatocellular carcinoma. Approximately 50-80% of HCC is accompanied by portal or hepatic vein invasion. The underlying mechanisms of PVTT development remain unclear. This study aimed to elucidate the role of miR-135a in PVTT tumorigenesis. METHODS: In the present study, we investigated the expression of microRNAs and mRNAs in PVTT tissues using advanced microRNA and cDNA microarray techniques. MicroRNA (miR)-135a was noted to be highly over-expressed in PVTT and the cell line CSQT-2 and was selected for further study. We characterized the function of miR-135a in vitro and in vivo. We also analyzed the clinical relevance of miR-135a in relation to the prognosis and survival of HCC patients with PVTT. RESULTS: Our analyses found that the miRNA and mRNA expression profiles of PVTT were distinct from the parenchyma tumor. Overexpression of miR-135a favors invasive and metastatic behavior in vitro. Furthermore, in a CSQT-2 orthotopic transplantation nude mouse model, blockade of miR-135a significantly reduced PVTT incidence. We also found that miR-135a was transcribed by forkhead box M1 (FOXM1), and metastasis suppressor 1 (MTSS1) was identified as the direct and functional target of miR-135a. Additionally, the cohort analysis revealed the relevance of miR-135a with respect to the prognosis and survival of HCC patients with PVTT. CONCLUSIONS: Our data suggest an important role for miR-135a in promoting PVTT tumorigenesis and indicate the potential application of miR-135a in PVTT therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Portal Vein , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Venous Thrombosis/etiology , Animals , Carcinoma, Hepatocellular/complications , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Humans , Liver Neoplasms/complications , Mice , Mice, Nude , MicroRNAs/antagonists & inhibitors , Microfilament Proteins/genetics , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Mol Cell Proteomics ; 9(12): 2617-28, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20699381

ABSTRACT

Gastric cardia cancer (GCC), which occurs at the gastric-esophageal boundary, is one of the most malignant tumors. Despite its high mortality and morbidity, the molecular mechanism of initiation and progression of this disease is largely unknown. In this study, using proteomics and metabolomics approaches, we found that the level of several enzymes and their related metabolic intermediates involved in glucose metabolism were deregulated in GCC. Among these enzymes, two subunits controlling pyruvic acid efflux, lactate dehydrogenase A (LDHA) and pyruvate dehydrogenase B (PDHB), were further analyzed in vitro. Either down-regulation of LDH subunit LDHA or overexpression of PDH subunit PDHB could force pyruvic acid into the Krebs cycle rather than the glycolysis process in AGS gastric cancer cells, which inhibited cell growth and cell migration. Our results reflect an important glucose metabolic signature, especially the dysregulation of pyruvic acid efflux in the development of GCC. Forced transition from glycolysis to the Krebs cycle had an inhibitory effect on GCC progression, providing potential therapeutic targets for this disease.


Subject(s)
Glucose/metabolism , Metabolomics , Proteomics , Stomach Neoplasms/metabolism , Base Sequence , Cell Line, Tumor , Chromatography, High Pressure Liquid , Citric Acid Cycle , DNA Primers , Electrophoresis, Gel, Two-Dimensional , Female , Glycolysis , Humans , L-Lactate Dehydrogenase/genetics , Male , Middle Aged , Polymerase Chain Reaction , Pyruvate Dehydrogenase Complex/genetics , RNA Interference , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology
10.
Nat Metab ; 4(2): 239-253, 2022 02.
Article in English | MEDLINE | ID: mdl-35145325

ABSTRACT

Tumors can reprogram the functions of metabolic enzymes to fuel malignant growth; however, beyond their conventional functions, key metabolic enzymes have not been found to directly govern cell mitosis. Here, we report that glutamine synthetase (GS) promotes cell proliferation by licensing mitotic progression independently of its metabolic function. GS depletion, but not impairment of its enzymatic activity, results in mitotic arrest and multinucleation across multiple lung and liver cancer cell lines, patient-derived organoids and xenografted tumors. Mechanistically, GS directly interacts with the nuclear pore protein NUP88 to prevent its binding to CDC20. Such interaction licenses activation of the CDC20-mediated anaphase-promoting complex or cyclosome to ensure proper metaphase-to-anaphase transition. In addition, GS is overexpressed in human non-small cell lung cancer and its depletion reduces tumor growth in mice and increases the efficacy of microtubule-targeted chemotherapy. Our findings highlight a moonlighting function of GS in governing mitosis and illustrate how an essential metabolic enzyme promotes cell proliferation and tumor development, beyond its main metabolic function.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Cell Cycle Proteins/metabolism , Cell Proliferation , Glutamate-Ammonia Ligase , Humans , Mice , Mitosis
11.
Hepatology ; 51(2): 535-44, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19918976

ABSTRACT

UNLABELLED: Eph/Ephrin family, one of the largest receptor tyrosine kinase families, has been extensively studied in morphogenesis and neural development. Recently, growing attention has been paid to its role in the initiation and progression of various cancers. However, the role of Eph/Ephrins in hepatocellular carcinoma (HCC) has been rarely investigated. In this study, we found that the expression of EphrinA2 was significantly up-regulated in both established cell lines and clinical tissue samples of HCC, and the most significant increase was observed in the tumors invading the portal veins. Forced expression of EphrinA2 in HCC cells significantly promoted in vivo tumorigenicity, whereas knockdown of this gene inhibited this oncogenic effect. We further found that suppression of apoptosis, rather than accelerating proliferation, was responsible for EphrinA2-enhanced tumorigenicity. In addition, EphrinA2 endowed cancer cells with resistance to tumor necrosis factor alpha (TNF-alpha)-induced apoptosis, thus facilitating their survival. Furthermore, we disclosed a novel EphrinA2/ras-related c3 botulinum toxin substrate 1 (Rac1)/V-akt murine thymoma viral oncogene homolog (Akt)/nuclear factor-kappa B (NF-kappaB) pathway contributing to the inhibitory effect on apoptosis in HCC cells. CONCLUSION: This study revealed that EphrinA2 played an important role in the development and progression of HCC by promoting the survival of cancer cells, indicating its role as a potential therapeutic target in HCC.


Subject(s)
Carcinoma, Hepatocellular/etiology , Ephrin-A2/physiology , Liver Neoplasms/etiology , NF-kappa B/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , rac1 GTP-Binding Protein/physiology , Cell Line, Tumor , Female , Humans , Male , Middle Aged
12.
Front Oncol ; 11: 770843, 2021.
Article in English | MEDLINE | ID: mdl-34746012

ABSTRACT

As a central cellular program to sense and transduce stress signals, the integrated stress response (ISR) pathway has been implicated in cancer initiation and progression. Depending on the genetic mutation landscape, cellular context, and differentiation states, there are emerging pieces of evidence showing that blockage of the ISR can selectively and effectively shift the balance of cancer cells toward apoptosis, rendering the ISR a promising target in cancer therapy. Going beyond its pro-survival functions, the ISR can also influence metastasis, especially via proteostasis-independent mechanisms. In particular, ISR can modulate metastasis via transcriptional reprogramming, in the help of essential transcription factors. In this review, we summarized the current understandings of ISR in cancer metastasis from the perspective of transcriptional regulation.

13.
Nat Commun ; 11(1): 220, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31924786

ABSTRACT

Cancer stem cells (CSCs) may be responsible for treatment resistance, tumor metastasis, and disease recurrence. Here we demonstrate that the Arf1-mediated lipid metabolism sustains cells enriched with CSCs and its ablation induces anti-tumor immune responses in mice. Notably, Arf1 ablation in cancer cells induces mitochondrial defects, endoplasmic-reticulum stress, and the release of damage-associated molecular patterns (DAMPs), which recruit and activate dendritic cells (DCs) at tumor sites. The activated immune system finally elicits antitumor immune surveillance by stimulating T-cell infiltration and activation. Furthermore, TCGA data analysis shows an inverse correlation between Arf1 expression and T-cell infiltration and activation along with patient survival in various human cancers. Our results reveal that Arf1-pathway knockdown not only kills CSCs but also elicits a tumor-specific immune response that converts dying CSCs into a therapeutic vaccine, leading to durable benefits.


Subject(s)
ADP-Ribosylation Factor 1/metabolism , Antineoplastic Agents/immunology , Lipid Metabolism/physiology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , ADP-Ribosylation Factor 1/genetics , ADP-Ribosylation Factor 1/pharmacology , Alarmins , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dendritic Cells/immunology , Endoplasmic Reticulum Stress/drug effects , Female , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/therapy , Gene Knockdown Techniques , Humans , Lipolysis/drug effects , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Neoplastic Stem Cells/cytology , T-Lymphocytes/immunology , Tamoxifen/pharmacology , Vaccination
14.
Oncotarget ; 8(48): 83602-83618, 2017 Oct 13.
Article in English | MEDLINE | ID: mdl-29137367

ABSTRACT

Tumor-initiating cells (TICs) play important roles in tumor progression and metastasis. Identifying the factors regulating TICs may open new avenues in cancer therapy. Here, we show that TIC-enriched prostate cancer cell clones use more glucose and secrete more lactate than TIC-low clones. We determined that elevated levels of phosphoenolpyruvate carboxykinase isoform 2 (PCK2) are critical for the metabolic switch and the maintenance of TICs in prostate cancer. Information from prostate cancer patient databases revealed that higher PCK2 levels correlated with more aggressive tumors and lower survival rates. PCK2 knockdown resulted in low TIC numbers, increased cytosolic acetyl-CoA and cellular protein acetylation. Our data suggest PCK2 promotes tumor initiation by lowering acetyl-CoA level through reducing the mitochondrial tricarboxylic acid (TCA) cycle. Thus, PCK2 is a potential therapeutic target for aggressive prostate tumors.

16.
Nat Commun ; 7: 10473, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26792023

ABSTRACT

Stem cell competition has emerged as a mechanism for selecting fit stem cells/progenitors and controlling tumourigenesis. However, little is known about the underlying molecular mechanism. Here we identify Mlf1-adaptor molecule (Madm), a novel tumour suppressor that regulates the competition between germline stem cells (GSCs) and somatic cyst stem cells (CySCs) for niche occupancy. Madm knockdown results in overexpression of the EGF receptor ligand vein (vn), which further activates EGF receptor signalling and integrin expression non-cell autonomously in CySCs to promote their overproliferation and ability to outcompete GSCs for niche occupancy. Conversely, expressing a constitutively activated form of the Drosophila JAK kinase (hop(Tum-l)) promotes Madm nuclear translocation, and suppresses vn and integrin expression in CySCs that allows GSCs to outcompete CySCs for niche occupancy and promotes GSC tumour formation. Tumour suppressor-mediated stem cell competition presented here could be a mechanism of tumour initiation in mammals.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Germ Cells/metabolism , Stem Cell Niche , Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Movement , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Female , Gene Expression Regulation, Developmental , Germ Cells/cytology , Male , Testis/cytology , Testis/metabolism , Tumor Suppressor Proteins/genetics
17.
Cancer Cell ; 22(3): 291-303, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22975373

ABSTRACT

Portal vein tumor thrombus (PVTT) is strongly correlated to a poor prognosis for patients with hepatocellular carcinoma (HCC). In this study, we uncovered a causative link between hepatitis B virus (HBV) infection and development of PVTT. Mechanistically, elevated TGF-ß activity, associated with the persistent presence of HBV in the liver tissue, suppresses the expression of microRNA-34a, leading to enhanced production of chemokine CCL22, which recruits regulatory T (Treg) cells to facilitate immune escape. These findings strongly suggest that HBV infection and activity of the TGF-ß-miR-34a-CCL22 axis serve as potent etiological factors to predispose HCC patients for the development of PVTT, possibly through the creation of an immune-subversive microenvironment to favor colonization of disseminated HCC cells in the portal venous system.


Subject(s)
Carcinoma, Hepatocellular/secondary , Chemokine CCL22/metabolism , Hepatitis B/complications , Liver Neoplasms/pathology , MicroRNAs/metabolism , Portal Vein , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/metabolism , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Female , Hepatitis B virus/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/immunology , Liver Neoplasms/virology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , MicroRNAs/biosynthesis , Middle Aged , Neoplasm Metastasis , Signal Transduction , Tumor Microenvironment
18.
Nat Commun ; 3: 667, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22314363

ABSTRACT

Eph receptors are implicated in regulating the malignant progression of cancer. Here we find that despite overexpression of EphB3 in human non-small-cell lung cancer, as reported previously, the expression of its cognate ligands, either ephrin-B1 or ephrin-B2, is significantly downregulated, leading to reduced tyrosine phosphorylation of EphB3. Forced activation of EphB3 kinase in EphB3-overexpressing non-small-cell lung cancer cells inhibits cell migratory capability in vitro as well as metastatic seeding in vivo. Furthermore, we identify a novel EphB3-binding protein, the receptor for activated C-kinase 1, which mediates the assembly of a ternary signal complex comprising protein phosphatase 2A, Akt and itself in response to EphB3 activation, leading to reduced Akt phosphorylation and subsequent inhibition of cell migration. Our study reveals a novel tumour-suppressive signalling pathway associated with kinase-activated EphB3 in non-small-cell lung cancer, and provides a potential therapeutic strategy by activating EphB3 signalling, thus inhibiting tumour metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Neuropeptides/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphB3/metabolism , Receptors, Cell Surface/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement , HEK293 Cells , Humans , Ligands , Lung Neoplasms/pathology , Mice , Mice, Nude , Models, Biological , Mutation , Neoplasm Metastasis , Neoplasm Transplantation , Phosphorylation , Photons , Receptors for Activated C Kinase , Signal Transduction , Tyrosine/chemistry
19.
Cancer Res ; 71(3): 1156-66, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21266352

ABSTRACT

Eph receptors, the largest subfamily of transmembrane tyrosine kinase receptors, have been increasingly implicated in various physiologic and pathologic processes, and the roles of the Eph family members during tumorigenesis have recently attracted growing attention. Until now, research on EphB3 function in cancer is limited to focusing on tumor suppression by EphB receptors in colorectal cancer. However, its function in other types of cancer remains poorly investigated. In this study, we explored the function of EphB3 in non-small-cell lung cancer (NSCLC). We found that the expression of EphB3 was significantly upregulated in clinical samples and cell lines, and the expression level correlated with the patient pathologic characteristics, including tumor size, differentiation, and metastasis. Overexpression of EphB3 in NSCLC cell lines accelerated cell growth and migration and promoted tumorigenicity in xenografts in a kinase-independent manner. In contrast, downregulation of EphB3 inhibited cell proliferation and migration and suppressed in vivo tumor growth and metastasis. Furthermore, we showed that silencing of EphB3 inhibited cell growth by reducing DNA synthesis and caspase-8-mediated apoptosis and suppressed cell migration by increasing accumulation of focal adhesion formation. Taken together, our findings suggest that EphB3 provides critical support to the development and progression of NSCLC by stimulating cell growth, migration, and survival, thereby implicating EphB3 as a potential therapeutic target in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Receptor, EphB3/biosynthesis , Animals , Apoptosis/physiology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 8/metabolism , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Cell Survival/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Disease Progression , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Nude , Neoplasm Metastasis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, EphB3/genetics
20.
PLoS One ; 6(6): e21419, 2011.
Article in English | MEDLINE | ID: mdl-21731740

ABSTRACT

BACKGROUND: Esophageal Squamous Cell Carcinoma (ESCC) is a major subtype of esophageal cancer causing significant morbility and mortality in Asia. Mechanism of initiation and progression of this disease is unclear. Tumor initiating cells (TICs) are the subpopulation of cells which have the ability to self-renew, as well as, to drive initiation and progression of cancer. Increasing evidence has shown that TICs exist in a variety of tumors. However, the identification and characterization of TICs in esophageal carcinoma has remained elusive. METHODOLOGY/PRINCIPAL FINDINGS: to identify TICs in ESCC, ESCC cell lines including two primary cells were used for screening suitable surface marker. Then colony formation assay, drug resistant assay and tumorigenicity assay in immune deficient mice were used to characterize TICs in ESCC. We found that just the CD44 expression correlated with tumorigenicity in ESCC cell lines. And then induced differentiation of ESCC cells by all-trans retinoic acid treatment led to decreased expression of CD44. The FACS isolated cell subpopulations with high CD44 expression showed increased colony formation and drug resistance in vitro, as well as significantly enhanced tumorigenicity in NOD/SICD mice, as compared to the low expressing CD44 ESCC cells. CONCLUSIONS/SIGNIFICANCE: our study has discovered a novel TIC surface marker, CD44, which can be utilized to enrich efficiently the TICs in ESCC. These findings will be useful for further studies of these cells and exploring therapeutic approaches.


Subject(s)
Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/pathology , Adult , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Esophageal Neoplasms/drug therapy , Female , Humans , Male , Mice , Middle Aged , Neoplastic Stem Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL