Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Theranostics ; 14(7): 3014-3028, 2024.
Article in English | MEDLINE | ID: mdl-38773979

ABSTRACT

Background: Periostin (POSTN) is a critical extracellular matrix protein in various tumor microenvironments. However, the function of POSTN in thyroid cancer progression remains largely unknown. Methods: Postn and Rag1 knock-out mice and orthotopic mouse models were used to determine the role of POSTN on papillary thyroid tumor progression. Immunofluorescence, cell co-culture, fluorescence in situ hybridization, chromatin immunoprecipitation assay, recombinant protein and inhibitor treatment were performed to explore the underlying mechanisms of POSTN-promoted papillary thyroid tumor growth. Results: POSTN is up-regulated in papillary thyroid tumors and negatively correlates with the overall survival of patients with thyroid cancer. Cancer-associated fibroblast (CAF)-derived POSTN promotes papillary thyroid tumor growth in vivo and in vitro. POSTN deficiency in CAFs significantly impairs CAF-promoted papillary thyroid tumor growth. POSTN promotes papillary thyroid tumor cell proliferation and IL-4 expression through integrin-FAK-STAT3 signaling. In turn, tumor cell-derived IL-4 induces the activation of CAFs and stimulates POSTN expression by activating STAT6. We reveal the crucial role of CAF-derived POSTN and tumor cell-derived IL-4 in driving the development of papillary thyroid tumors through the POSTN-integrin-FAK-STAT3-IL-4 pathway in tumor cells and IL-4-STAT6-POSTN signaling in CAFs. Conclusion: Our findings underscore the significance of POSTN and IL-4 as critical molecular mediators in the dynamic interplay between CAFs and tumor cells, ultimately supporting the growth of papillary thyroid tumors.


Subject(s)
Cancer-Associated Fibroblasts , Cell Adhesion Molecules , Cell Proliferation , Mice, Knockout , STAT3 Transcription Factor , Signal Transduction , Thyroid Cancer, Papillary , Thyroid Neoplasms , Animals , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/genetics , STAT3 Transcription Factor/metabolism , Cancer-Associated Fibroblasts/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/genetics , Mice , Humans , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Cell Line, Tumor , Tumor Microenvironment , Interleukin-4/metabolism , Integrins/metabolism , Focal Adhesion Kinase 1/metabolism , Periostin
2.
Nat Commun ; 15(1): 1929, 2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38431724

ABSTRACT

Single-cell and spatial transcriptome sequencing, two recently optimized transcriptome sequencing methods, are increasingly used to study cancer and related diseases. Cell annotation, particularly for malignant cell annotation, is essential and crucial for in-depth analyses in these studies. However, current algorithms lack accuracy and generalization, making it difficult to consistently and rapidly infer malignant cells from pan-cancer data. To address this issue, we present Cancer-Finder, a domain generalization-based deep-learning algorithm that can rapidly identify malignant cells in single-cell data with an average accuracy of 95.16%. More importantly, by replacing the single-cell training data with spatial transcriptomic datasets, Cancer-Finder can accurately identify malignant spots on spatial slides. Applying Cancer-Finder to 5 clear cell renal cell carcinoma spatial transcriptomic samples, Cancer-Finder demonstrates a good ability to identify malignant spots and identifies a gene signature consisting of 10 genes that are significantly co-localized and enriched at the tumor-normal interface and have a strong correlation with the prognosis of clear cell renal cell carcinoma patients. In conclusion, Cancer-Finder is an efficient and extensible tool for malignant cell annotation.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Gene Expression Profiling , Transcriptome/genetics , Algorithms , Kidney Neoplasms/genetics , Single-Cell Analysis
3.
Br J Cancer ; 130(3): 358-368, 2024 02.
Article in English | MEDLINE | ID: mdl-38097742

ABSTRACT

BACKGROUND: This study aimed to investigate the underlying mechanisms of matricellular protein periostin (POSTN) on tumour-stroma crosstalk in the liver metastatic microenvironment of colorectal cancer (CRC). METHODS: Postn-knockout mice and hepatic Postn-overexpressing mice were used to investigate the functions of POSTN on the formation of fibrotic microenvironment and the tumour-stroma crosstalk in the liver metastatic microenvironment of CRC. Clinical samples and database were analyzed to show the correlation between POSTN expression and fibrotic features and TGF-ß signalling in metastatic livers of CRC. RESULTS: POSTN deficiency reduced hepatic stellate cell (HSC) activation and liver metastasis, whereas POSTN overexpression in the liver significantly augmented the formation of a fibrotic microenvironment to support the liver metastatic growth of CRC cells in mice. Moreover, HSC-derived POSTN promoted TGF-ß1 expression in CRC cells through the integrin/FAK/ERK/STAT3 pathway; conversely, tumour cell-derived TGF-ß1 induced POSTN expression in HSCs via the Smad pathway. POSTN levels correlated with fibrotic features and TGF-ß signalling in metastatic liver tissues of CRC patients. CONCLUSIONS: POSTN and TGF-ß1 cooperatively contribute to the tumour-stroma crosstalk by forming a supporting fibrotic microenvironment to promote liver metastasis of CRC cells via the POSTN/integrin/FAK/ERK/STAT3/TGF-ß axis in tumour cells and TGF-ß/Smad/POSTN signalling in activated HSCs.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Animals , Humans , Mice , Colorectal Neoplasms/pathology , Integrins/metabolism , Liver/metabolism , Liver Neoplasms/pathology , Periostin , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment
4.
Matrix Biol ; 121: 22-40, 2023 08.
Article in English | MEDLINE | ID: mdl-37230256

ABSTRACT

Matrix rigidity is a critical contributor to tumor progression; however, whether and how matrix stiffness modulates the collective invasion of tumor cells remain unknown. Here we demonstrate that increased matrix stiffness activates YAP to promote the secretion of periostin (POSTN) in cancer-associated fibroblasts, which in turn augments the matrix rigidity of mammary glands and breast tumor tissues by facilitating collagen crosslinking. Moreover, decreased tissue stiffening resulted from the POSTN deficiency impairs peritoneal metastatic potential of orthotopic breast tumors. Increased matrix stiffness also promotes three-dimensional (3D) collective breast tumor cell invasion via multicellular cytoskeleton remodeling. POSTN triggers the integrin/FAK/ERK/Cdc42/Rac1 mechanotransduction pathway during 3D collective invasion of breast tumor. Clinically, high POSTN expression correlates with high collagen levels in breast tumors and cooperatively determines the metastatic recurrence potential in breast cancer patients. Collectively, these findings indicate that matrix rigidity promotes 3D collective invasion of breast tumor cells via the YAP-POSTN-integrin mechanotransduction signaling.


Subject(s)
Breast Neoplasms , Integrins , Mechanotransduction, Cellular , Female , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Collagen/metabolism , Integrins/genetics , Integrins/metabolism , Mechanotransduction, Cellular/physiology , Neoplasm Invasiveness
5.
Dev Cell ; 58(9): 760-778.e6, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37054705

ABSTRACT

The STING-mediated type I interferon (IFN) signaling pathway has been shown to play critical roles in antitumor immunity. Here, we demonstrate that an endoplasmic reticulum (ER)-localized JmjC domain-containing protein, JMJD8, inhibits STING-induced type I IFN responses to promote immune evasion and breast tumorigenesis. Mechanistically, JMJD8 competes with TBK1 for binding with STING, blocking STING-TBK1 complex formation and restricting type I IFN and IFN-stimulated gene (ISG) expression as well as immune cell infiltration. JMJD8 knockdown improves the efficacy of chemotherapy and immune checkpoint therapy in treating both human and mouse breast cancer cell-derived implanted tumors. The clinical relevance is highlighted in that JMJD8 is highly expressed in human breast tumor samples, and its expression is inversely correlated with that of type I IFN and ISGs as well as immune cell infiltration. Overall, our study found that JMJD8 regulates type I IFN responses, and targeting JMJD8 triggers antitumor immunity.


Subject(s)
Breast Neoplasms , Immune Evasion , Animals , Female , Humans , Mice , Endoplasmic Reticulum/metabolism , Immunity, Innate , Membrane Proteins/genetics , Membrane Proteins/metabolism , Signal Transduction/genetics
6.
Cell Rep ; 42(2): 112090, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36773295

ABSTRACT

Periostin, a multifunctional extracellular protein, plays an important role in inflammatory disorders and tumorigenesis. Our previous work has demonstrated that periostin deficiency inhibits colorectal cancer (CRC) progression. Here, we aim to clarify the role of periostin in the immune microenvironment of CRC. We find that periostin deficiency significantly decreases the infiltration of programmed death receptor 1 (PD-1)+ tumor-associated macrophages (TAMs) in CRC tissues. Periostin promotes the expression of PD-1 on TAMs by integrin-ILK-nuclear factor κB (NF-κB) signaling, and PD-1+ TAMs produce interleukin-6 (IL-6) and interferon γ (IFN-γ) to induce the expression of PD-L1 on colorectal tumor cells. Moreover, combined inhibition of periostin and PD-1 significantly suppresses CRC progression compared with the inhibition of periostin or PD-1 alone. In summary, our results suggest that periostin deficiency reduces the infiltration of PD-1+ TAMs and enhances the efficacy of anti-PD-1 treatment in CRC.


Subject(s)
Colorectal Neoplasms , Tumor-Associated Macrophages , Humans , B7-H1 Antigen/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Macrophages/metabolism , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism
7.
Cell Mol Gastroenterol Hepatol ; 15(6): 1475-1504, 2023.
Article in English | MEDLINE | ID: mdl-36801449

ABSTRACT

BACKGROUND & AIMS: The matricellular protein periostin plays a critical role in liver inflammation, fibrosis, and even carcinoma. Here, the biological function of periostin in alcohol-related liver disease (ALD) was investigated. METHODS: We used wild-type (WT), Postn-null (Postn-/-) mice and Postn-/- mice with periostin recovery to investigate the biological function of periostin in ALD. Proximity-dependent biotin identification analysis identified the protein that interacted with periostin, and coimmunoprecipitation analysis validated the interaction between protein disulfide isomerase (PDI) and periostin. Pharmacological intervention and genetic knockdown of PDI were used to investigate the functional correlation between periostin and PDI in ALD development. RESULTS: Periostin was markedly upregulated in the livers of mice that were fed ethanol. Interestingly, periostin deficiency severely aggravated ALD in mice, whereas the recovery of periostin in the livers of Postn-/- mice significantly ameliorated ALD. Mechanistic studies showed that the upregulation of periostin alleviated ALD by activating autophagy through inhibition of the mechanistic target of rapamycin complex 1 (mTORC1) pathway, which was verified in murine models treated with the mTOR inhibitor rapamycin and the autophagy inhibitor MHY1485. Furthermore, a protein interaction map of periostin was generated by proximity-dependent biotin identification analysis. Interaction profile analysis identified PDI as a key protein that interacted with periostin. Intriguingly, periostin-mediated enhancement of autophagy by inhibiting the mTORC1 pathway in ALD depended on its interaction with PDI. Moreover, alcohol-induced periostin overexpression was regulated by transcription factor EB. CONCLUSIONS: Collectively, these findings clarify a novel biological function and mechanism of periostin in ALD and the periostin-PDI-mTORC1 axis is a critical determinant of ALD.


Subject(s)
Hepatocytes , Liver Diseases, Alcoholic , Mice , Animals , Hepatocytes/metabolism , Protein Disulfide-Isomerases/genetics , Protein Disulfide-Isomerases/metabolism , Biotin/metabolism , Liver Diseases, Alcoholic/pathology , Ethanol/toxicity , Mechanistic Target of Rapamycin Complex 1/metabolism , Autophagy
8.
J Pathol ; 255(2): 212-223, 2021 10.
Article in English | MEDLINE | ID: mdl-34228359

ABSTRACT

Periostin is a critical extracellular regulator in the pathogenesis of liver disorders such as hepatosteatosis, non-alcoholic steatohepatitis, inflammation, and fibrosis. Periostin is also involved in the progression of hepatocellular carcinoma (HCC). However, the molecular mechanisms of periostin in hepatic stellate cell (HSC) activation and tumor cell proliferation in the pathogenesis of HCC remain largely unknown. We demonstrate that periostin is markedly upregulated in diethylnitrosamine (DEN)-induced mouse HCC tissues and that periostin knockout impairs DEN-induced HCC development. Periostin is predominantly derived from activated HSCs and periostin deficiency in HSCs impairs HSC activation and inhibits HSC-promoted HCC cell proliferation in vitro and tumor growth in vivo. Mechanistically, periostin promotes HSC activation through the integrin-FAK-STAT3-periostin pathway and augments HCC cell proliferation by activating ERK. There are positive correlations between periostin and HSC activation and cell proliferation in HCC clinical samples. Collectively, our findings demonstrate that HSC-derived periostin promotes HCC development by enhancing HSC activation through an autocrine periostin-integrin-FAK-STAT3-periostin circuit and by augmenting HCC cell proliferation via the ERK pathway in a paracrine manner. Thus, periostin is a multifaceted extracellular regulator in the development of HCC. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/metabolism , Hepatic Stellate Cells/metabolism , Liver Neoplasms/pathology , Animals , Carcinogens/toxicity , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Diethylnitrosamine/toxicity , Humans , Liver Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology
9.
FEBS Lett ; 595(16): 2099-2112, 2021 08.
Article in English | MEDLINE | ID: mdl-34165806

ABSTRACT

Periostin (POSTN) is a type of matricellular protein, but its functions in adipose fibrosis remain unclear. Here, we found that POSTN expression is significantly increased in mouse adipose tissue after treatment with lipopolysaccharide (LPS) or a high-fat diet (HFD) and that adipose progenitor cells are the main source of POSTN. In our mouse model of fibrosis, POSTN deletion protected mice from adipose fibrosis, probably through reducing the accumulation of macrophages and promoting adipocyte differentiation of progenitor cells. Taken together, our study demonstrates that POSTN deficiency attenuates adipose tissue fibrosis and improves insulin resistance, providing new insights into the diagnosis and treatment of type II diabetes by targeting adipose tissue fibrosis.


Subject(s)
Adipose Tissue/pathology , Cell Adhesion Molecules/deficiency , Lipopolysaccharides/pharmacology , Obesity/pathology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Cell Adhesion Molecules/genetics , Diet, High-Fat/adverse effects , Fibrosis , Insulin Resistance , Mice , Mice, Inbred C57BL
10.
Cell Rep ; 30(3): 793-806.e6, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31968254

ABSTRACT

Periostin is a multifunctional extracellular matrix protein involved in various inflammatory diseases and tumor metastasis; however, evidence regarding whether and how periostin actively contributes to inflammation-associated tumorigenesis remains elusive. Here, we demonstrate that periostin deficiency significantly inhibits the occurrence of colorectal cancer in azoxymethane/dextran sulfate sodium-treated mice and in ApcMin/+ mice. Moreover, periostin deficiency attenuates the severity of colitis and reduces the proliferation of tumor cells. Mechanistically, stromal fibroblast-derived periostin activates FAK-Src kinases through integrin-mediated outside-in signaling, which results in the activation of YAP/TAZ and, subsequently, IL-6 expression in tumor cells. Conversely, IL-6 induces periostin expression in fibroblasts by activating STAT3, which ultimately facilitates colorectal tumor development. These findings provide the evidence that periostin promotes colorectal tumorigenesis, and identify periostin- and IL-6-mediated tumor-stroma interaction as a promising target for treating colitis-associated colorectal cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/pathology , Cell Adhesion Molecules/metabolism , Colorectal Neoplasms/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , src-Family Kinases/metabolism , Adenomatous Polyposis Coli/metabolism , Animals , Azoxymethane , Cell Adhesion Molecules/deficiency , Cell Proliferation , Colitis/complications , Dextran Sulfate , Humans , Inflammation/pathology , Interleukin-6/metabolism , Intestines/pathology , Mice, Inbred C57BL , Myofibroblasts/pathology , Precancerous Conditions/pathology , STAT3 Transcription Factor , Signal Transduction , Stromal Cells/pathology , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
11.
Adv Exp Med Biol ; 1132: 125-136, 2019.
Article in English | MEDLINE | ID: mdl-31037631

ABSTRACT

Extracellular matrix protein periostin is highly expressed in various tumors and plays a critical role in tumor development and progression. Periostin is mainly secreted by stromal cells such as cancer-associated fibroblasts, myofibroblasts, osteoblasts and bone marrow-derived mesenchymal stromal cells. But in some cases, tumor cells, especially cancer stem cells, can also produce periostin. Periostin has been shown to regulate multiple biological behaviors of tumor cells, including proliferation, survival, invasion, angiogenesis, metastasis and chemoresistance. Moreover, an excessive periostin deposition exerts a pivotal role in remodeling various tumor microenvironments, such as cancer stem cell niche, perivascular niche, premetastatic niche, immunosuppressive microenvironment, bone marrow microenvironment and other tumor growth-supportive microenvironments. In this review, we provide an update understanding of the multifaceted functions and mechanisms of periostin in tumor development and progression.


Subject(s)
Cell Adhesion Molecules/physiology , Neoplasms/pathology , Neoplastic Stem Cells , Stromal Cells , Tumor Microenvironment , Humans , Stem Cell Niche
12.
Cell Rep ; 26(6): 1533-1543.e4, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30726736

ABSTRACT

Periostin (POSTN) is a multifunctional extracellular component that regulates cell-matrix interactions and cell-cell crosstalk. POSTN deletion significantly decreases leukemia burden in mice; however, the underlying mechanisms by which POSTN promotes B cell acute lymphoblastic leukemia (B-ALL) progression remain largely unknown. Here, we demonstrate that bone marrow (BM)-derived mesenchymal stromal cells (BM-MSCs) express higher levels of POSTN when co-cultured with B-ALL cells in vitro and in vivo. POSTN deficiency in BM-MSCs significantly decreases CCL2 expression in co-cultured B-ALL cells in vitro and in vivo. Moreover, POSTN treatment increases expression of CCL2 in B-ALL cells by activating the integrin-ILK-NF-κB pathway. Conversely, CCL2 treatment upregulates expression of POSTN in BM-MSCs via STAT3 activation. Furthermore, there is a positive correlation between POSTN expression and CCL2 level in the BM of mice and patients with B-ALL. These findings suggest that B-ALL cell-derived CCL2 contributes to the increased leukemia burden promoted by BM-MSC-derived POSTN.


Subject(s)
Bone Marrow Cells/metabolism , Cell Adhesion Molecules/metabolism , Chemokine CCL2/metabolism , Mesenchymal Stem Cells/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Line , Cells, Cultured , Humans , Integrins/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Protein Serine-Threonine Kinases , STAT3 Transcription Factor/metabolism
13.
Matrix Biol ; 66: 81-92, 2018 03.
Article in English | MEDLINE | ID: mdl-28965986

ABSTRACT

Periostin (Postn) is a crucial extracellular remodeling factor that has been implicated in the pathogenesis of hepatic inflammation, fibrosis, non-alcoholic fatty liver disease and liver cancer. However, the role of Postn in liver regeneration remains unclear. Here, we demonstrate that Postn mRNA and protein levels are significantly upregulated in the mice after 2/3 partial hepatectomy (PHx). Compared with wild-type mice, Postn-deficient mice exhibit lower liver/body weight ratio and less Ki67-positive cells at days 2, 8 and 14 after PHx. Macrophage infiltration and the levels of TNF-α, IL-6 and HGF in the livers of Postn-deficient mice are significantly decreased compared with wild-type mice one day after PHx. In addition, overexpression of Postn leads to higher liver/body weight ratio and more Ki67-positive cells in the livers of mice and promotes hepatocyte proliferation in vitro. Moreover, liver sinusoidal endothelial cells, biliary epithelial cells and hepatocytes can express Postn after PHx, and Postn deficiency impairs angiogenesis during liver regeneration. Our findings indicate that Postn deficiency impairs liver regeneration in mice after PHx and Postn might be a novel promoter for liver regeneration.


Subject(s)
Cell Adhesion Molecules/deficiency , Hepatectomy/methods , Liver Regeneration , Liver/cytology , Animals , Body Weight , Cell Proliferation , Cytokines/metabolism , Gene Expression Regulation , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/immunology , Macrophages/immunology , Mice , Organ Size
14.
Cell Mol Life Sci ; 74(23): 4287-4291, 2017 12.
Article in English | MEDLINE | ID: mdl-28884337

ABSTRACT

Tumor microenvironment consists of tumor cells, stromal cells, extracellular matrix and a plethora of soluble components. The complex array of interactions between tumor cells and their surrounding tumor microenvironments contribute to the determination of the fate of tumor cells during tumorigenesis and metastasis. Matricellular protein periostin is generally absent in most adult tissues but is highly expressed in tumor microenvironments. Current evidence reveals that periostin plays a critical role in establishing and remodeling tumor microenvironments such as the metastatic niche, cancer stem cell niche, perivascular niche, pre-metastatic niche, fibrotic microenvironment and bone marrow microenvironment. Here, we summarize the current knowledge of the multifaceted role of periostin in the tumor microenvironments.


Subject(s)
Cell Adhesion Molecules/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Neoplasms/genetics , Tumor Microenvironment/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Adhesion Molecules/metabolism , Cell Communication , Disease Progression , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Stem Cell Niche/genetics , Stromal Cells/metabolism , Stromal Cells/pathology
15.
J Exp Med ; 214(1): 245-267, 2017 01.
Article in English | MEDLINE | ID: mdl-27923907

ABSTRACT

Glioblastoma is the most lethal brain tumor and harbors glioma stem cells (GSCs) with potent tumorigenic capacity. The function of GSCs in tumor propagation is maintained by several core transcriptional regulators including c-Myc. c-Myc protein is tightly regulated by posttranslational modification. However, the posttranslational regulatory mechanisms for c-Myc in GSCs have not been defined. In this study, we demonstrate that the deubiquitinase USP13 stabilizes c-Myc by antagonizing FBXL14-mediated ubiquitination to maintain GSC self-renewal and tumorigenic potential. USP13 was preferentially expressed in GSCs, and its depletion potently inhibited GSC proliferation and tumor growth by promoting c-Myc ubiquitination and degradation. In contrast, overexpression of the ubiquitin E3 ligase FBXL14 induced c-Myc degradation, promoted GSC differentiation, and inhibited tumor growth. Ectopic expression of the ubiquitin-insensitive mutant T58A-c-Myc rescued the effects caused by FBXL14 overexpression or USP13 disruption. These data suggest that USP13 and FBXL14 play opposing roles in the regulation of GSCs through reversible ubiquitination of c-Myc.


Subject(s)
Brain Neoplasms/pathology , Endopeptidases/physiology , F-Box Proteins/antagonists & inhibitors , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-myc/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitination , Cell Line, Tumor , Cell Proliferation , F-Box Proteins/physiology , Humans , Ubiquitin-Protein Ligases/physiology , Ubiquitin-Specific Proteases
16.
J Pathol ; 239(4): 484-95, 2016 08.
Article in English | MEDLINE | ID: mdl-27193093

ABSTRACT

Periostin (POSTN) is a limiting factor in the metastatic colonization of disseminated tumour cells. However, the role of POSTN in regulating the immunosuppressive function of immature myeloid cells in tumour metastasis has not been documented. Here, we demonstrate that POSTN promotes the pulmonary accumulation of myeloid-derived suppressor cells (MDSCs) during the early stage of breast tumour metastasis. Postn deletion decreases neutrophil and monocytic cell populations in the bone marrow of mice and suppresses the accumulation of MDSCs to premetastatic sites. We also found that POSTN-deficient MDSCs display reduced activation of ERK, AKT and STAT3 and that POSTN deficiency decreases the immunosuppressive functions of MDSCs during tumour progression. Moreover, the pro-metastatic role of POSTN is largely limited to ER-negative breast cancer patients. Lysyl oxidase contributes to POSTN-promoted premetastatic niche formation and tumour metastasis. Our findings indicate that POSTN is essential for immunosuppressive premetastatic niche formation in the lungs during breast tumour metastasis and is a potential target for the prevention and treatment of breast tumour metastasis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Breast Neoplasms/pathology , Cell Adhesion Molecules/metabolism , Immune Tolerance/genetics , Myeloid-Derived Suppressor Cells/pathology , Neoplasm Metastasis/pathology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Adhesion Molecules/genetics , Mice , Mice, Knockout , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Metastasis/genetics , Signal Transduction/physiology
17.
Oncotarget ; 7(16): 21825-39, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26968810

ABSTRACT

miR-543 has been implicated as having a critical role in the development of breast cancer, endometrial cancer and hepatocellular carcinoma. However, the exact clinical significance and biological functions of miR-543 in colorectal cancer (CRC) remain unclear. Here, we found that miR-543 expression significantly downregulated in tumors from patients with CRC, APCMin mice and a mouse model of colitis-associated colon cancer. miR-543 level was inversely correlated with the metastatic status of patients with CRC and the metastatic potential of CRC cell lines. Moreover, ectopic expression of miR-543 inhibited the proliferation and metastasis of CRC cells in vitro and in vivo by targeting KRAS, MTA1 and HMGA2. Conversely, miR-543 knockdown promoted the proliferation, migration and invasion of CRC cells in vitro and augmented tumor growth and metastasis in vivo. Furthermore, we found that miR-543 expression was negatively correlated with the levels of KRAS, MTA1 and HMGA2 in clinical samples. Collectively, these data show that miR-543 inhibits the proliferation and metastasis of CRC cells by targeting KRAS, MTA1 and HMGA2. Our study highlights a pivotal role for miR-543 as a suppressor in the regulation of CRC growth and metastasis and suggests that miR-543 may serve as a novel diagnostic and prognostic biomarker for CRC metastasis.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , HMGA2 Protein/genetics , Histone Deacetylases/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Repressor Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , HCT116 Cells , HEK293 Cells , HMGA2 Protein/metabolism , HT29 Cells , Histone Deacetylases/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Neoplasm Metastasis , Proto-Oncogene Proteins p21(ras)/metabolism , Repressor Proteins/metabolism , Trans-Activators , Transplantation, Heterologous , Tumor Burden/drug effects , Tumor Burden/genetics
18.
Oncotarget ; 6(37): 39550-63, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26474460

ABSTRACT

Tumor cells actively contribute to constructing their own microenvironment during tumorigenesis and tumor progression. The tumor microenvironment contains multiple types of stromal cells that work together with the extracellular matrix and local and systemic factors to coordinately contribute to tumor initiation and progression. Tumor cells and their stromal compartments acquire many genetic and/or epigenetic alternations to facilitate tumor growth and metastasis. The cancer stem cell (CSC) concept has been widely applied to interpreting tumor initiation, growth, metastasis, dormancy and relapse. CSCs have differentiation abilities to generate the original lineage cells that are similar to their normal stem cell counterparts. Interestingly, recent evidence demonstrates that CSCs also have the potential to transdifferentiate into vascular endothelial cells and pericytes, indicating that CSCs can transdifferentiate into other lineage cells for promoting tumor growth and metastasis in some tissue contexts instead of only recruiting stromal cells from local or distant tissues. Although the transdifferentiation of CSCs into tumor stromal cells provides a new dimension that explains tumor heterogeneity, many aspects of CSC transdifferentiation remain elusive. In this review, we summarize the multi-lineage differentiation and transdifferentiation potentials of CSCs as well as discuss their potential contributions to tumor heterogeneity and tumor microenvironment in tumor progression.


Subject(s)
Cell Differentiation , Cell Transdifferentiation , Neoplastic Stem Cells/pathology , Tumor Microenvironment , Cell Lineage , Endothelial Cells/pathology , Fibroblasts/pathology , Humans , Mesenchymal Stem Cells/pathology , Models, Biological
19.
Cell Biosci ; 5: 32, 2015.
Article in English | MEDLINE | ID: mdl-26146543

ABSTRACT

MicroRNAs (miRNAs) are small noncoding regulatory RNAs that regulate gene expression post-transcriptionally by either inhibiting protein translation or degrading target mRNAs. The differential expression profiles of miRNAs in different types of cancers and in the multi-step process of tumor progression indicate that miRNAs are involved in tumor onset, growth and progression. Metastasis is the most common cause of cancer-related mortality. Current evidence demonstrates that aberrant miRNA expression promotes or inhibits tumor metastasis by modulating the expression of numerous target genes. Therefore, the identification of metastasis-related miRNAs and a better understanding of the complex functions of miRNAs in tumor metastasis will provide potential diagnostic and prognostic biomarkers, as well as therapeutic targets for clinical application. Here, we review the functions of miRNAs in the control of multiple steps of tumor metastasis.

20.
Cell Mol Life Sci ; 72(18): 3411-24, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25967289

ABSTRACT

Current evidence indicates that a subpopulation of cancer cells, named cancer stem cells (CSCs) or tumor-initiating cells, are responsible for the initiation, growth, metastasis, therapy resistance and recurrence of cancers. CSCs share core regulatory pathways with normal stem cells; however, CSCs rely on distinct reprogrammed pathways to maintain stemness and to contribute to the progression of cancers. The specific targeting of CSCs, together with conventional chemotherapy or radiotherapy, may achieve stable remission or cure cancer. Therefore, the identification of CSCs and a better understanding of the complex characteristics of CSCs will provide invaluable diagnostic, therapeutic and prognostic targets for clinical application. In this review, we will introduce the dysregulated properties of CSCs in cancers and discuss the possible challenges in targeting CSCs for cancer treatment.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Disease Progression , Drug Resistance, Neoplasm/drug effects , Humans , Neoplasms/therapy , Neoplastic Stem Cells/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...