Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Cell Death Dis ; 15(6): 417, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879509

ABSTRACT

Chemotherapy is a crucial treatment for colorectal tumors. However, its efficacy is restricted by chemoresistance. Recently, Golgi dispersal has been suggested to be a potential response to chemotherapy, particularly to drugs that induce DNA damage. However, the underlying mechanisms by which Golgi dispersal enhances the capacity to resist DNA-damaging agents remain unclear. Here, we demonstrated that DNA-damaging agents triggered Golgi dispersal in colorectal cancer (CRC), and cancer stem cells (CSCs) possessed a greater degree of Golgi dispersal compared with differentiated cancer cells (non-CSCs). We further revealed that Golgi dispersal conferred resistance against the lethal effects of DNA-damaging agents. Momentously, Golgi dispersal activated the Golgi stress response via the PKCα/GSK3α/TFE3 axis, resulting in enhanced protein and vesicle trafficking, which facilitated drug efflux through ABCG2. Identification of Golgi dispersal indicated an unexpected pathway regulating chemoresistance in CRC.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm , Golgi Apparatus , Neoplastic Stem Cells , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Humans , Golgi Apparatus/metabolism , Golgi Apparatus/drug effects , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Animals , Cell Line, Tumor , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , DNA Damage , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Int J Biol Sci ; 20(7): 2748-2762, 2024.
Article in English | MEDLINE | ID: mdl-38725859

ABSTRACT

Abnormal nuclear enlargement is a diagnostic and physical hallmark of malignant tumors. Large nuclei are positively associated with an increased risk of developing metastasis; however, a large nucleus is inevitably more resistant to cell migration due to its size. The present study demonstrated that the nuclear size of primary colorectal cancer (CRC) cells at an advanced stage was larger than cells at an early stage. In addition, the nuclei of CRC liver metastases were larger than those of the corresponding primary CRC tissues. CRC cells were sorted into large-nucleated cells (LNCs) and small-nucleated cells (SNCs). Purified LNCs exhibited greater constricted migratory and metastatic capacity than SNCs in vitro and in vivo. Mechanistically, ErbB4 was highly expressed in LNCs, which phosphorylated lamin A/C at serine 22 via the ErbB4-Akt1 signaling pathway. Furthermore, the level of phosphorylated lamin A/C was a negative determinant of nuclear stiffness. Taken together, CRC LNCs possessed greater constricted migratory and metastatic potential than SNCs due to ErbB4-Akt1-mediated lamin A/C phosphorylation and nuclear softening. These results may provide a potential treatment strategy for tumor metastasis by targeting nuclear stiffness in patients with cancer, particularly CRC.


Subject(s)
Cell Nucleus , Colorectal Neoplasms , Lamin Type A , Proto-Oncogene Proteins c-akt , Receptor, ErbB-4 , Signal Transduction , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Lamin Type A/metabolism , Mice, Nude , Neoplasm Metastasis , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-4/metabolism , Receptor, ErbB-4/genetics
4.
iScience ; 26(9): 107606, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37664607

ABSTRACT

Invadopodia, being actin-rich membrane protrusions, play a vital role in tumor cell invasion and metastasis. Our previous studies have revealed some functions of the DOC-2/DAB2 interacting protein (DAB2IP) as a tumor suppressor. Nevertheless, the specific role and mechanism of DAB2IP in invadopodia formation remain unclear. Here, we find that DAB2IP effectively suppresses invadopodia formation and metastasis in breast cancer, both in vitro and in vivo. Additionally, DAB2IP could downregulate anaplastic lymphoma kinase (ALK), resulting in the inhibition of tyrosine phosphorylation of Cortactin and the prevention of invadopodia formation. DAB2IP competitively antagonizes the interaction between the deubiquitinating enzyme Ubiquitin-specific peptidase 10 (USP10) and ALK, leading to a decrease in the abundance of ALK protein. In summary, DAB2IP impairs the stability of ALK through USP10-dependent deubiquitination, suppressing Cortactin phosphorylation, thereby inhibiting invadopodia formation and metastasis of breast cancer cells. Furthermore, this study suggests a potential therapeutic strategy for breast cancer treatment.

5.
Cell Death Dis ; 13(7): 651, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35896535

ABSTRACT

Uneven oxygen supply in solid tumors leads to hypoxic and normoxic regions. Hypoxic cells exhibit increased secretion of lactate, which creates an acidic tumor microenvironment (TME). This acidic TME is positively associated with tumor metastasis. Despite the increased metastatic capacity of hypoxic cells, they are located relatively further away from the blood vessels and have limited access to the circulatory system. Studies have shown that cancer stem cells (CSCs) are enriched for tumor metastasis-initiating cells and generally undergo aerobic respiration, which could be enhanced by lactate. We therefore hypothesized that TME-derived lactate may promote the metastasis of normoxic CSCs. In the present study, the abundance of hypoxic and normoxic CSCs was analyzed in primary CRC tumors. It was found that the proportion of normoxic CSCs was positively associated with tumor stage. Using two human CRC cell lines, LoVo and SW480, and a patient-derived xenograft (XhCRC), it was found that treatment with lactate promoted normoxic CSC metastasis. Metabolism analysis indicated that, upon treatment with lactate, oxidative phosphorylation (OXPHOS) activity in normoxic CSCs was enhanced, whereas hypoxic CSCs were rarely altered. At the molecular level, the expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a master regulator of lactate oxidation, was found to be elevated in normoxic CSCs. Furthermore, PGC-1α knockdown markedly reduced the metastatic potential of normoxic CSCs. Notably, both the PGC-1α-mediated OXPHOS activity and metastatic potential were impaired when hypoxia-inducible factor-1α (HIF-1α) was activated in normoxic CSCs. Together, these findings provide a therapeutic strategy against tumor metastasis through the targeting of PGC-1α and, thus, the suppression of lactate-feeding OXPHOS in normoxic CSCs may improve the therapeutic benefit of patients with cancer, particularly CRC.


Subject(s)
Colorectal Neoplasms , Oxidative Phosphorylation , Cell Line , Colorectal Neoplasms/pathology , Humans , Hypoxia/pathology , Lactic Acid , Neoplastic Stem Cells/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Tumor Microenvironment
6.
Oncogene ; 41(31): 3830-3845, 2022 07.
Article in English | MEDLINE | ID: mdl-35773411

ABSTRACT

Yes-associated protein 1 (YAP1), a central component of the Hippo pathway, plays an important role in tumor metastasis; however, the underlying mechanism remains to be elucidated. Invadopodia are actin-rich protrusions containing multiple proteases and have been widely reported to promote cell invasiveness by degrading the extracellular matrix. In the present study, we report that YAP1 induces invadopodia formation and promotes tumor metastasis in breast cancer cells. We also identify TIAM1, a guanine nucleotide exchange factor, as a target of the YAP1-TEAD4 complex. Our results demonstrate that YAP1 could promote TEAD4 binding to the enhancer region of TIAM1, which activates TIAM1 expression, subsequently increasing RAC1 activity and inducing invadopodia formation. These findings reveal the functional role of Hippo signaling in the regulation of invadopodia and provide potential molecular targets for preventing tumor metastasis in breast cancer.


Subject(s)
Breast Neoplasms , Podosomes , Actins/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Female , Guanine Nucleotide Exchange Factors/metabolism , Humans , Muscle Proteins/metabolism , Neoplasm Invasiveness , Podosomes/metabolism , T-Lymphoma Invasion and Metastasis-inducing Protein 1/metabolism , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
7.
Int J Pharm ; 590: 119963, 2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33039492

ABSTRACT

BACKGROUND: RNAi-based technology has achieved good results in both in vitro and in vivo applications, and it is expected to become a good genetic treatment for some diseases, especially neoplastic diseases. But there are still many obstacles in the in vivo application, the most important thing is the lack of an efficient and safe carrier. METHODS: In this study, we designed and constructed a new siRNA delivery, which was named as aptamer-protamine-siRNA nanoparticle (APR). APR was consisted of ErbB3 aptamer, protamine and siRNA. We used Zeta nanosize to detect the size of APR to verify whether it is a nano-scale compound. We use the FAMRNA to replace the siRNA to detect whether APR could recognize and enter ErbB3 positive MCF-7 cells. Then we replaced the siRNA as oncogene suvivin siRNA to detect whether APR could inhibit tumor growth by silence surviving, and replaced siRNA to CDK1 siRNA to detect the cell cycle blocking effect. At last we tested the anticancer effect and safety of APR by carrying survivin siRNA in MCF-7 bearing nude mice. RESULTS: APR was identified as a nanoscale compound. It showed specific targeting for ErbB3-positive MCF-7 cancer cells. APR has demonstrated the characteristics of inhibiting tumor growth by carrying siRNA against oncogene survivin. APR could also block cell cycle of MCF-7 cells by delivering CDK1 siRNAs. In the ErbB3 positive breast cancer xenograft mice model, APR nanoparticles could inhibit tumor growth and cause tumor regression without any toxicity. CONCLUSIONS: In both in vivo and in vitro applications, APR nanoparticles could be targeted to recognize and enter ErbB3 positive tumor cells, and play a corresponding role by silencing targeted gene expression. APR nanoparticle is expected to become a good tumor treatment option.


Subject(s)
Breast Neoplasms , Nanoparticles , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Protamines , RNA, Small Interfering , Receptor, ErbB-3
8.
Cancer Lett ; 493: 236-244, 2020 11 28.
Article in English | MEDLINE | ID: mdl-32898601

ABSTRACT

Tumors harbor diverse compartments of cells with distinct metabolic properties and phenotypes, but the mechanism by which metabolic commensalism among distinct subsets of cancer cells affects tumor progression remains unclear. Colorectal cancer (CRC) has been reported to consist of cancer stem cells (CSCs) and differentiated cancer cells (non-CSCs). In the present study, organoid models were employed to show that CSCs and non-CSCs in CRC were characterized by distinct metabolic phenotypes. Treatment with either non-CSC-derived conditioned medium or exogenous lactate enhanced organoid-forming and tumor-initiating capacity of CSCs. In tumor regeneration assays with co-implanted CSCs and non-CSCs, the tumor-initiating activity was reduced when either monocarboxylate transporter (MCT)4 in non-CSCs or MCT1 in CSCs was silenced or inhibited. Mechanistically, oxiadative phosphorylation-derived reactive oxygen species in CSCs activated AKT-Wnt/ß-catenin signaling, which could be induced by lactate from non-CSCs. Overall, these results suggest that CSCs and non-CSCs possess distinct metabolic profiles and, unexpectedly, non-CSC-originated lactate promotes self-renewal of CSCs and thus contributes to CRC progression. Our findings establish a rationale for developing novel therapies targeting the metabolic commensalism between different cell populations in CRC.


Subject(s)
Colorectal Neoplasms/pathology , Lactic Acid/metabolism , Neoplastic Stem Cells/pathology , Organoids/transplantation , Animals , Cell Differentiation , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Organoids/cytology , Organoids/metabolism , Oxidative Phosphorylation , Tumor Cells, Cultured , Wnt Signaling Pathway
9.
Cell Death Dis ; 11(7): 610, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737283

ABSTRACT

Tumor heterogeneity is an important feature of malignant tumors, and cell subpopulations may positively interact to facilitate tumor progression. Studies have shown that hypoxic cancer cells possess enhanced metastatic capacity. However, it is still unclear whether hypoxic cancer cells may promote the metastasis of normoxic cells, which have greater access to the blood circulation. When cocultured with hypoxic CRC cells or treated with hypoxic CRC cell-derived CM, normoxic CRC cells possessed increased metastatic capacity. Furthermore, hypoxic CRC cell-derived CM was enriched in interleukin 8. Hypoxic CRC cell-derived CM and recombinant human IL-8 both enhanced the metastatic capacity of normoxic cells by increasing the phosphorylation of p65 and then by inducing epithelial-mesenchymal transition. Knockdown of IL-8 in hypoxic CRC cells or the use of an anti-IL-8 antibody attenuated the CM- or rhIL-8-induced prometastatic capacity of normoxic CRC cells. Inhibition or knockdown of p65 abrogated IL-8-induced prometastatic effects. Most importantly, hypoxia-treated xenograft tumors enhanced the metastasis of normoxic CRC cells. Hypoxic CRC cell-derived IL-8 promotes the metastatic capacity of normoxic cells, and novel therapies targeting the positive interactions between hypoxic and normoxic cells should be developed.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Interleukin-8/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Tumor Hypoxia , Cell Hypoxia , Cell Line, Tumor , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Paracrine Communication
10.
Int J Oncol ; 54(3): 893-904, 2019 03.
Article in English | MEDLINE | ID: mdl-30664193

ABSTRACT

Three­dimensional (3D) cultures are indispensable for capturing tumor heterogeneity in colorectal cancer (CRC) in vitro. Although 3D cultures (such as sphere­forming assay and organoid culture) can partially preserve the morphological and molecular characteristics of primary CRC, whether these 3D cultures maintain the long­term stemness of cancer stem cells (CSCs) remains largely unknown. In the present study, spheres and organoids were generated side by side using individual primary CRC specimens, then respectively processed as serial passages. The results revealed that during serial passages, the percentage of CSCs (such as cluster of differentiation­133+ and Wnt+ cells) in organoids and the tumor­initiating capacity of organoid­derived cells were constant, while they gradually increased in the sphere­derived cells. Furthermore, during serial passages, resistance to chemotherapeutic agents (including 5­fluorouracil and oxaliplatin) in sphere­ and organoid­derived cells was evaluated. The results indicated that the percentage of chemoresistant cells was constant in serial organoid cultures; however, it gradually increased in the serial sphere­forming assays. Taken together, the results of the present study comprehensively demonstrate that, with regard to long­term culture in vitro, organoid culture may be useful in maintaining tumor heterogeneity and the levels of chemoresistant cells, while the sphere formation assay enriches for CSCs and chemoresistant cells.


Subject(s)
Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Neoplastic Stem Cells/pathology , Organoids/pathology , Tissue Culture Techniques/standards , Animals , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Female , Humans , Mice , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Organoids/drug effects , Organoids/metabolism , Tumor Cells, Cultured , Wnt Proteins/metabolism
11.
J Exp Clin Cancer Res ; 37(1): 175, 2018 Jul 28.
Article in English | MEDLINE | ID: mdl-30055645

ABSTRACT

BACKGROUND: Focal adhesion plays an essential role in tumour invasiveness and metastasis. Hippo component YAP has been widely reported to be involved in many aspects of tumour biology. However, its role in focal adhesion regulation in breast cancer remains unexplored. METHODS: Tissue microarray was used to evaluate YAP expression in clinical breast cancer specimens by immunohistochemical staining. Cell migration and invasion abilities were measured by Transwell assay. A cell adhesion assay was used to measure the ability of cell adhesion to gelatin. The focal adhesion was visualized through immunofluorescence. Phosphorylated FAK and other proteins were detected by Western blot analysis. Gene expression profiling was used to screen differently expressed genes, and gene ontology enrichment was performed using DAVID software. The gene mRNA levels were measured by quantitative real-time PCR. The activity of the THBS1-promoter was evaluated by dual luciferase assay. Chromatin immunoprecipitation (ChIP) was used to verify whether YAP could bind to the THBS1-promoter region. The prediction of potential protein-interaction was performed with the String program. The ChIP sequence data of TEAD was obtained from the ENCODE database and analysed via the ChIP-seek tool. The gene expression dataset (GSE30480) of purified tumour cells from primary breast tumour tissues and metastatic lymph nodes was used in the gene set enrichment analysis. Prognostic analysis of the TCGA dataset was performed by the SurvExpress program. Gene expression correlation of the TCGA dataset was analysed via R2: Genomics Analysis and Visualization Platform. RESULTS: Our study provides evidence that YAP acts as a promoter of focal adhesion and tumour invasiveness via regulating FAK phosphorylation in breast cancer. Further experiments reveal that YAP could induce FAK phosphorylation through a TEAD-dependent manner. Using gene expression profiling and bioinformatics analysis, we identify the FAK upstream gene, thrombospondin 1, as a direct transcriptional target of YAP-TEAD. Silencing THBS1 could reverse the YAP-induced FAK activation and focal adhesion. CONCLUSION: Our results unveil a new signal axis, YAP/THBS1/FAK, in the modulation of cell adhesion and invasiveness, and provides new insights into the crosstalk between Hippo signalling and focal adhesion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Focal Adhesions/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Thrombospondin 1/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/physiology , Female , Focal Adhesions/genetics , Focal Adhesions/pathology , HEK293 Cells , Hippo Signaling Pathway , Humans , Lymphatic Metastasis , MCF-7 Cells , Neoplasm Invasiveness , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Thrombospondin 1/genetics , Transcription Factors , Transfection , YAP-Signaling Proteins
12.
Oncotarget ; 8(64): 107907-107919, 2017 Dec 08.
Article in English | MEDLINE | ID: mdl-29296212

ABSTRACT

Colorectal cancer (CRC) is heterogeneous and contains different-sized cells. Recent studies have shown that tumor-initiating cells (TICs) are involved in cancer initiation, recurrence and metastasis. However, connections between cancer cell size and stem-like properties are largely unknown. Here we purified large- and small-sized CRC cells by fluorescence-activated cell sorting (FACS) based on forward scatter (FSC), and demonstrated that small CRC cells possess higher holoclone- and sphere-forming capacity in vitro, tumor-initiating capacity in vivo and form more lung metastases compared with large CRC cells. Furthermore, we found that down-regulated YAP1 (yes-associated protein 1) decreased tumor-initiating and metastatic capacity in small CRC cells but not in large CRC cells. More importantly, our results showed that the expression of YAP1 positively correlated with the poor prognosis in CRCs. Collectively, our findings suggest that small CRC cells enrich for metastatic TICs, and YAP1 is one of the potential therapeutic targets of metastatic TICs, the small CRC cells.

13.
Oncotarget ; 7(49): 80700-80715, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27813496

ABSTRACT

Serum carcinoembryonic antigen (CEA) is the most commonly used tumor marker in a variety of cancers including colorectal cancer (CRC) for tumor diagnosis and monitoring. Recent studies have shown that colonic crypt cells expressing little or no CEA may enrich for stem cells. Numerous studies have clearly shown that there exist CRC patients with normal serum CEA levels during tumor progression or even tumor relapse, although CEA itself is considered to promote metastasis and block cell differentiation. These seemingly contradictory observations prompted us to investigate, herein, the biological properties as well as tumorigenic and metastatic capacity of CRC cells that express high (CEA+) versus low CEA (CEA-/lo) levels of CEA. Our findings show that the abundance of CEA-/lo cells correlate with poor differentiation and poor prognosis, and moreover, CEA-/lo cells form more spheres in vitro, generate more tumors and exhibit a higher potential in developing liver and lung metastases than corresponding CEA+ cells. Applying RNAi-mediated approach, we found that IGF1R mediated tumorigenic and capacity of CEA-/lo cells but did not mediate those of CEA+ cells. Notably, our data demonstrated that CEA molecule was capable of protecting CEA-/lo cells from anoikis, implying that CEA+ cells, although themselves possessing less tumorigenic and metastatic capacity, may promote metastasis of CEA-/lo cells via secreting CEA molecule. Our observations suggest that, besides targeting CEA molecule, CEA-/lo cells may represent a critical source of tumor progression and metastasis, and should therefore be the target of future therapies.


Subject(s)
Adenocarcinoma/metabolism , Carcinoembryonic Antigen/metabolism , Cell Movement , Colorectal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Adult , Aged , Animals , Anoikis , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Grading , Neoplastic Stem Cells/pathology , Phenotype , RNA Interference , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Cells, Cultured
14.
Oncotarget ; 7(36): 57866-57877, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27506941

ABSTRACT

Histone methyltransferases and demethylases regulate transcription by altering the epigenetic marks on histones in tumorigenesis. Members of the histone lysine(K)-specific demethylase 4 (KDM4) family are dysregulated in several types of cancer. Here, we report a novel role for KDM4B in mitochondrial apoptosis. In this study, we demonstrate that KDM4B is overexpressed in colorectal cancer (CRC) tissues. Decreased expression of KDM4B significantly promoted apoptosis of CRC cell lines. Moreover, our data indicate that HAX1 is required for KDM4B-mediated mitochondrial apoptosis. The transcription of HAX1 was directly activated by KDM4B. We also show that HAX1 is overexpressed in CRC tissues and is positively correlated with KMD4B expression. Collectively, we demonstrate that KDM4B may play an important role in mitochondrial apoptosis and represent a potential therapeutic cancer target in CRC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases/metabolism , Mitochondria/metabolism , Animals , Cell Line, Tumor , Cell Survival , Colorectal Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , RNA Interference , RNA, Messenger/metabolism , Transcriptional Activation
15.
Sci Rep ; 5: 16578, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26564738

ABSTRACT

Metastasis is a critical factor for the high mortality of colorectal cancer (CRC), but its mechanism is not completely understood. Epithelial-mesenchymal transition (EMT) is thought to play a key role in metastasis and also increases the cancer stem cell (CSC) feature that facilitates metastatic colonization. In this study, we investigated the biological roles of DAB2IP regulating EMT and stem cell-like features in human CRC. We demonstrate that DAB2IP suppresses NF-κB-mediated EMT and CSC features in CRC cells. In DAB2IP knockout mice, we discovered the hyperplasia in colonic epithelium which aberrantly represents the mesenchymal feature and NF-κB pathway activation. In clinic CRC tissue, we also reveal that reduced DAB2IP can enrich the CD133(+) subpopulation. DAB2IP expression was inversely correlated with tumor differentiation and metastasis, and patients with lower DAB2IP expression had shorter overall survival time. Taken together, our study demonstrates that DAB2IP inhibits NF-κB-inducing EMT and CSC to suppress the CRC progression, and also suggests that DAB2IP is a beneficial prediction factor for CRC patient prognosis.


Subject(s)
Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Neoplastic Stem Cells/metabolism , ras GTPase-Activating Proteins/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/drug effects , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Kaplan-Meier Estimate , Male , Mice, Nude , Middle Aged , NF-kappa B/metabolism , Neoplastic Stem Cells/drug effects , Organoplatinum Compounds/pharmacology , Oxaliplatin , Prognosis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays/methods , ras GTPase-Activating Proteins/metabolism
16.
Mol Med Rep ; 12(3): 4071-4078, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26062578

ABSTRACT

Colorectal cancer (CRC) is one of the most common types of cancer worldwide. Hematopoietic cell­specific protein 1­associated protein X­1 (HAX­1) has been found to be involved in several types of cancer. However, the role of HAX­1 in CRC remains to be elucidated. The aim of the present study was to investigate whether the expression of HAX­1 is associated with the progression of CRC, and to determine the effects of HAX­1 on the apoptosis and proliferation of CRC cells. Tumor tissues and adjacent noncancerous tissues were collected from 60 patients with CRC, following the provision of informed consent. The expression levels of HAX­1 and the association with clinical and pathological characteristics were then analyzed. The expression levels of HAX­1 were significantly higher in the cancerous tissues from the patients with CRC, particularly in tissues of an advanced stage of cancer. In addition, HAX­1 expression was associated with malignant progression and poor prognosis. Furthermore, SW480 CRC cells, overexpressing HAX­1, exhibited increased resistance to camptothecin in vitro, and promoted proliferation in vitro and in vivo. By contrast, HAX­1 knockdown significantly decreased the proliferation. In addition, the expression levels of ki­67 and phosphorylated­akt were inhibited following HAX­1 knockdown. In conclusion, the expression levels of HAX­1 were increased in cancerous tissue from patients with CRC, and were associated with progression of the disease. These results suggested that HAX­1 may contribute to chemotherapy resistance and malignant progression in CRC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Aged , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Mice , Mice, Nude , Middle Aged , Prognosis , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transplantation, Heterologous
17.
PLoS One ; 10(5): e0125625, 2015.
Article in English | MEDLINE | ID: mdl-25938772

ABSTRACT

Chemotherapy resistance observed in patients with colorectal cancer (CRC) may be related to the presence of cancer stem cells (CSCs), but the underlying mechanism(s) remain unclear. Carcinoma-associated fibroblasts (CAFs) are intimately involved in tumor recurrence, and targeting them increases chemo-sensitivity. We investigated whether fibroblasts might increase CSCs thus mediating chemotherapy resistance. CSCs were isolated from either patient-derived xenografts or CRC cell lines based on expression of CD133. First, CSCs were found to be inherently resistant to cell death induced by chemotherapy. In addition, fibroblast-derived conditioned medium (CM) promoted percentage, clonogenicity and tumor growth of CSCs (i.e., CD133+ and TOP-GFP+) upon treatment with 5-fluorouracil (5-Fu) or oxaliplatin (OXA). Further investigations exhibited that exosomes, isolated from CM, similarly took the above effects. Inhibition of exosome secretion decreased the percentage, clonogenicity and tumor growth of CSCs. Altogether, our findings suggest that, besides targeting CSCs, new therapeutic strategies blocking CAFs secretion even before chemotherapy shall be developed to gain better clinical benefits in advanced CRCs.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Exosomes/metabolism , Fibroblasts/metabolism , Neoplastic Stem Cells/pathology , AC133 Antigen , Animals , Antigens, CD/metabolism , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Drug Resistance, Neoplasm/drug effects , Exosomes/drug effects , Exosomes/ultrastructure , Female , Fibroblasts/drug effects , Glycoproteins/metabolism , Humans , Mice, Nude , Neoplastic Stem Cells/drug effects , Paracrine Communication/drug effects , Peptides/metabolism , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays
18.
Cytometry A ; 83(10): 944-51, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23926097

ABSTRACT

Although the restriction point (R-point) was proposed in animal cells several decades ago, its existence in normal cells is still controversial, because, in most studies, long-term cultured cell lines rather than primary normal cells were used. Furthermore, cell synchronization was generally applied, resulting in growth imbalance between DNA synthesis and protein expression in cells. Finally, R-point was originally proposed as a unique arrest point that may be in G0 phase; however, generally believed R-point locates within G1 phase. Thus, up to now, there is no solid experimental evidence that supports the existence of R-point in asynchronous primary normal cells. In this study, we used freshly purified peripheral human blood lymphocytes, as asynchronous primary normal cells, to confirm the existence of restriction point in G1 not G0 phase. Our findings may help uncover the mystery of the deregulation of cell cycle progression in malignant tumors. © 2013 International Society for Advancement of Cytometry.


Subject(s)
Flow Cytometry/methods , G1 Phase Cell Cycle Checkpoints/physiology , Lymphocytes/cytology , Humans
19.
J Huazhong Univ Sci Technolog Med Sci ; 33(2): 205-211, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23592131

ABSTRACT

The forkhead family members of transcription factors (FoxOs) are expected to be potential cancer-related drug targets and thus are being extremely studied recently. In the present study, FoxO3a, one major member of this family, was identified to be down-regulated in colorectal cancer through micro-array analysis, which was confirmed by RT-PCR and Western blot in 28 patients. Moreover, immunohistochemistry (IHC) showed that the expression levels of FoxO3a were remarkably reduced in 99 cases of primary colorectal cancer, liver metastasis, and even in metaplastic colorectal tissue. IHC also revealed an exclusion of FoxO3a from the nucleus of most cells of tumor-associated tissues. Silencing FoxO3a by siRNA led to elevation of G2-M phase cells. We conclude that the downregulation of FoxO3a may greatly contribute to tumor development, and thus FoxO3a may represent a novel therapeutic target in colorectal cancer.


Subject(s)
Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Rectum/pathology , Cell Cycle Checkpoints , Colon/metabolism , Down-Regulation , Female , Forkhead Box Protein O3 , Humans , Liver Neoplasms/pathology , Male , Metaplasia/metabolism , Metaplasia/pathology , Rectum/metabolism , Tumor Cells, Cultured
20.
J Huazhong Univ Sci Technolog Med Sci ; 32(5): 680-685, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23073796

ABSTRACT

The normal range of oral mucosal cell apoptosis and proliferation rate through a larger sample of non-malnourished crowd was investigated, and the nutritional status of clinical patients was assessed. Of 194 clinical patients selected according to "NRS2002" guidance, there were 167 non-malnourished patients and 27 malnourished cases, respectively. Twelve patients with toxic reactions of grade III after postoperative chemotherapy (POC) were chosen. The oral mucosal epithelial apoptosis and proliferation rate were measured by using flow cytometry. The statistical significance was processed by using unpaired t-test. The results showed that there was no significant difference in gender, age and body weight between malnourished and non-malnourished groups. The normal range of oral mucosal epithelial apoptosis and the proliferation rate was (27.50±1.50)% and (15.12±1.68)% in non-malnourished patients, and that was (19.90±4.14)% and (6.66±5.83)% in the malnourished patients, respectively. It is concluded that the normal range of oral mucosa cell apoptosis and proliferation rate is achieved, which can not be influenced by gender, age, weight and other factors, and could be used as a sensitive and accurate index to assess the nutritional status of clinical patients.


Subject(s)
Apoptosis/physiology , Mouth Mucosa/pathology , Mouth Mucosa/physiology , Nutritional Status/physiology , Cell Proliferation , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL