Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 299(11): 105351, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37838174

ABSTRACT

Breast cancer stem cells are mainly responsible for poor prognosis, especially in triple-negative breast cancer (TNBC). In a previous study, we demonstrated that ε-Sarcoglycan (SGCE), a type Ⅰ single-transmembrane protein, is a potential oncogene that promotes TNBC stemness by stabilizing EGFR. Here, we further found that SGCE depletion reduces breast cancer stem cells, partially through inhibiting the transcription of FGF-BP1, a secreted oncoprotein. Mechanistically, we demonstrate that SGCE could interact with the specific protein 1 transcription factor and translocate into the nucleus, which leads to an increase in the transcription of FGF-BP1, and the secreted FBF-BP1 activates FGF-FGFR signaling to promote cancer cell stemness. The novel SGCE-Sp1-FGF-BP1 axis provides novel potential candidate diagnostic markers and therapeutic targets for TNBC.


Subject(s)
Neoplastic Stem Cells , Sarcoglycans , Sp1 Transcription Factor , Triple Negative Breast Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Neoplastic Stem Cells/metabolism , Sarcoglycans/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Triple Negative Breast Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/metabolism
2.
Mol Cancer ; 23(1): 60, 2024 03 22.
Article in English | MEDLINE | ID: mdl-38520019

ABSTRACT

BACKGROUND: Cancer stem-like cell is a key barrier for therapeutic resistance and metastasis in various cancers, including breast cancer, yet the underlying mechanisms are still elusive. Through a genome-wide lncRNA expression profiling, we identified that LINC00115 is robustly upregulated in chemoresistant breast cancer stem-like cells (BCSCs). METHODS: LncRNA microarray assay was performed to document abundance changes of lncRNAs in paclitaxel (PTX)-resistant MDA-MB-231 BCSC (ALDH+) and non-BCSC (ALDH-). RNA pull-down and RNA immunoprecipitation (RIP) assays were performed to determine the binding proteins of LINC00115. The clinical significance of the LINC00115 pathway was examined in TNBC metastatic lymph node tissues. The biological function of LINC00115 was investigated through gain- and loss-of-function studies. The molecular mechanism was explored through RNA sequencing, mass spectrometry, and the CRISPR/Cas9-knockout system. The therapeutic potential of LINC00115 was examined through xenograft animal models. RESULTS: LINC00115 functions as a scaffold lncRNA to link SETDB1 and PLK3, leading to enhanced SETDB1 methylation of PLK3 at both K106 and K200 in drug-resistant BCSC. PLK3 methylation decreases PLK3 phosphorylation of HIF1α and thereby increases HIF1α stability. HIF1α, in turn, upregulates ALKBH5 to reduce m6A modification of LINC00115, resulting in attenuated degradation of YTHDF2-dependent m6A-modified RNA and enhanced LINC00115 stability. Thus, this positive feedback loop provokes BCSC phenotypes and enhances chemoresistance and metastasis in triple-negative breast cancer. SETDB1 inhibitor TTD-IN with LINC00115 ASO sensitizes PTX-resistant cell response to chemotherapy in a xenograft animal model. Correlative expression of LINC00115, methylation PLK3, SETDB1, and HIF1α are prognostic for clinical triple-negative breast cancers. CONCLUSIONS: Our findings uncover LINC00115 as a critical regulator of BCSC and highlight targeting LINC00115 and SETDB1 as a potential therapeutic strategy for chemotherapeutic resistant breast cancer.


Subject(s)
RNA, Long Noncoding , Triple Negative Breast Neoplasms , Animals , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Breast/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Paclitaxel/pharmacology , Disease Models, Animal , Neoplastic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Polo-like Kinases , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
3.
Cancer Metastasis Rev ; 42(2): 543-574, 2023 06.
Article in English | MEDLINE | ID: mdl-36997828

ABSTRACT

It is well established that innervation is one of the updated hallmarks of cancer and that psychological stress promotes the initiation and progression of cancer. The breast tumor environment includes not only fibroblasts, adipocytes, endothelial cells, and lymphocytes but also neurons, which is increasingly discovered important in breast cancer progression. Peripheral nerves, especially sympathetic, parasympathetic, and sensory nerves, have been reported to play important but different roles in breast cancer. However, their roles in the breast cancer progression and treatment are still controversial. In addition, the brain is one of the favorite sites of breast cancer metastasis. In this review, we first summarize the innervation of breast cancer and its mechanism in regulating cancer growth and metastasis. Next, we summarize the neural-related molecular markers in breast cancer diagnosis and treatment. In addition, we review drugs and emerging technologies used to block the interactions between nerves and breast cancer. Finally, we discuss future research directions in this field. In conclusion, the further research in breast cancer and its interactions with innervated neurons or neurotransmitters is promising in the clinical management of breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Endothelial Cells
4.
Cancer Sci ; 115(7): 2094-2106, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38651282

ABSTRACT

Ubiquitination and deubiquitylation are pivotal posttranslational modifications essential for regulating cellular protein homeostasis and are implicated in the development of human diseases. Ubiquitin-specific protease 3 (USP3), a member of the ubiquitin-specific protease family, serves as a key deubiquitylation enzyme, playing a critical role in diverse cellular processes including the DNA damage response, cell cycle regulation, carcinogenesis, tumor cell proliferation, migration, and invasion. Despite notable research efforts, our current understanding of the intricate and context-dependent regulatory networks governing USP3 remains incomplete. This review aims to comprehensively synthesize existing published works on USP3, elucidating its multifaceted roles, functions, and regulatory mechanisms, while offering insights for future investigations. By delving into the complexities of USP3, this review strives to provide a foundation for a more nuanced understanding of its specific roles in various cellular processes. Furthermore, the exploration of USP3's regulatory networks may uncover novel therapeutic strategies targeting this enzyme in diverse human diseases, thereby holding promising clinical implications. Overall, an in-depth comprehension of USP3's functions and regulatory pathways is crucial for advancing our knowledge and developing targeted therapeutic approaches for human diseases.


Subject(s)
Neoplasms , Ubiquitin-Specific Proteases , Ubiquitination , Humans , Neoplasms/metabolism , Neoplasms/genetics , Ubiquitin-Specific Proteases/metabolism , DNA Damage , Cell Proliferation , Protein Processing, Post-Translational , Carcinogenesis/metabolism , Carcinogenesis/genetics , Animals
5.
Mol Ther ; 30(7): 2603-2617, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35278676

ABSTRACT

Cancer cells respond to various stressful conditions through the dynamic regulation of RNA m6A modification. Doxorubicin is a widely used chemotherapeutic drug that induces DNA damage. It is interesting to know whether cancer cells regulate the DNA damage response and doxorubicin sensitivity through RNA m6A modification. Here, we found that doxorubicin treatment significantly induced RNA m6A methylation in breast cancer cells in both a dose- and a time-dependent manner. However, protein arginine methyltransferase 5 (PRMT5) inhibited RNA m6A modification under doxorubicin treatment by enhancing the nuclear translocation of the RNA demethylase AlkB homolog 5 (ALKBH5), which was previously believed to be exclusively localized in the nucleus. Then, ALKBH5 removed the m6A methylation of BRCA1 for mRNA stabilization and further enhanced DNA repair competency to decrease doxorubicin efficacy in breast cancer cells. Importantly, we identified the approved drug tadalafil as a novel PRMT5 inhibitor that could decrease RNA m6A methylation and increase doxorubicin sensitivity in breast cancer. The strategy of targeting PRMT5 with tadalafil is a promising approach to promote breast cancer sensitivity to doxorubicin through RNA methylation regulation.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Demethylation , Doxorubicin/pharmacology , Female , Humans , Protein-Arginine N-Methyltransferases/genetics , RNA , Tadalafil
6.
Acta Biochim Biophys Sin (Shanghai) ; 55(9): 1487-1495, 2023 May 09.
Article in English | MEDLINE | ID: mdl-37162264

ABSTRACT

Angiopoietin-1 (ANG1) is a pro-angiogenic regulator that contributes to the progression of solid tumors by stimulating the proliferation, migration and tube formation of vascular endothelial cells, as well as the renewal and stability of blood vessels. However, the functions and mechanisms of ANG1 in triple-negative breast cancer (TNBC) are unclear. The clinical sample database shows that a higher level of ANG1 in TNBC is associated with poor prognosis compared to non-TNBC. In addition, knockdown of ANG1 inhibits TNBC cell proliferation and induces cell cycle G1 phase arrest and apoptosis. Overexpression of ANG1 promotes tumor growth in nude mice. Mechanistically, ANG1 promotes TNBC by upregulating carboxypeptidase A4 (CPA4) expression. Overall, the ANG1-CPA4 axis can be a therapeutic target for TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Animals , Mice , Cell Line, Tumor , Triple Negative Breast Neoplasms/metabolism , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Mice, Nude , Endothelial Cells/metabolism , Cell Proliferation/genetics , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Gene Expression Regulation, Neoplastic , Cell Movement/genetics
7.
Acta Biochim Biophys Sin (Shanghai) ; 55(6): 1001-1019, 2023 May 15.
Article in English | MEDLINE | ID: mdl-37184281

ABSTRACT

The functional capacity of organisms declines in the process of aging. In the case of breast tissue, abnormal mammary gland development can lead to dysfunction in milk secretion, a primary function, as well as the onset of various diseases, such as breast cancer. In the process of aging, the terminal duct lobular units (TDLUs) within the breast undergo gradual degeneration, while the proportion of adipose tissue in the breast continues to increase and hormonal levels in the breast change accordingly. Here, we review changes in morphology, internal structure, and cellular composition that occur in the mammary gland during aging. We also explore the emerging mechanisms of breast aging and the relationship between changes during aging and breast-related diseases, as well as potential interventions for delaying mammary gland aging and preventing breast disease.


Subject(s)
Breast Neoplasms , Mammary Glands, Human , Humans , Female , Breast , Aging
8.
Int J Mol Sci ; 24(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298108

ABSTRACT

Although DNA damage repair plays a critical role in cancer chemotherapy, the function of lncRNAs in this process remains largely unclear. In this study, in silico screening identified H19 as an lncRNA that potentially plays a role in DNA damage response and sensitivity to PARP inhibitors. Increased expression of H19 is correlated with disease progression and with a poor prognosis in breast cancer. In breast cancer cells, forced expression of H19 promotes DNA damage repair and resistance to PARP inhibition, whereas H19 depletion diminishes DNA damage repair and increases sensitivity to PARP inhibitors. H19 exerted its functional roles via direct interaction with ILF2 in the cell nucleus. H19 and ILF2 increased BRCA1 stability via the ubiquitin-proteasome proteolytic pathway via the H19- and ILF2-regulated BRCA1 ubiquitin ligases HUWE1 and UBE2T. In summary, this study has identified a novel mechanism to promote BRCA1-deficiency in breast cancer cells. Therefore, targeting the H19/ILF2/BRCA1 axis might modulate therapeutic approaches in breast cancer.


Subject(s)
Breast Neoplasms , RNA, Long Noncoding , Humans , Female , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/therapeutic use , Ubiquitin/metabolism , DNA Damage , Nuclear Factor 45 Protein/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
9.
Hepatology ; 74(3): 1461-1479, 2021 09.
Article in English | MEDLINE | ID: mdl-33813748

ABSTRACT

BACKGROUND AND AIMS: Hypoxia is a common feature of the tumor microenvironment (TME), which promotes tumor progression, metastasis, and therapeutic drug resistance through a myriad of cell activities in tumor and stroma cells. While targeting hypoxic TME is emerging as a promising strategy for treating solid tumors, preclinical development of this approach is lacking in the study of HCC. APPROACH AND RESULTS: From a genome-wide CRISPR/CRISPR-associated 9 gene knockout screening, we identified aldolase A (ALDOA), a key enzyme in glycolysis and gluconeogenesis, as an essential driver for HCC cell growth under hypoxia. Knockdown of ALDOA in HCC cells leads to lactate depletion and consequently inhibits tumor growth. Supplementation with lactate partly rescues the inhibitory effects mediated by ALDOA knockdown. Upon hypoxia, ALDOA is induced by hypoxia-inducible factor-1α and fat mass and obesity-associated protein-mediated N6 -methyladenosine modification through transcriptional and posttranscriptional regulation, respectively. Analysis of The Cancer Genome Atlas shows that elevated levels of ALDOA are significantly correlated with poor prognosis of patients with HCC. In a screen of Food and Drug Administration-approved drugs based on structured hierarchical virtual platforms, we identified the sulfamonomethoxine derivative compound 5 (cpd-5) as a potential inhibitor to target ALDOA, evidenced by the antitumor activity of cpd-5 in preclinical patient-derived xenograft models of HCC. CONCLUSIONS: Our work identifies ALDOA as an essential driver for HCC cell growth under hypoxia, and we demonstrate that inhibition of ALDOA in the hypoxic TME is a promising therapeutic strategy for treating HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Fructose-Bisphosphate Aldolase/genetics , Liver Neoplasms/genetics , Tumor Hypoxia/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Fructose-Bisphosphate Aldolase/metabolism , Gene Knockdown Techniques , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactic Acid/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Loss of Function Mutation , Mice , Neoplasm Transplantation , Sulfamonomethoxine/analogs & derivatives , Tumor Microenvironment , Xenograft Model Antitumor Assays
10.
Cancer Sci ; 112(6): 2097-2117, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33811715

ABSTRACT

Krüppel-like factor 5 (KLF5) is a member of the KLF family. Recent studies have suggested that KLF5 regulates the expression of a large number of new target genes and participates in diverse cellular functions, such as stemness, proliferation, apoptosis, autophagy, and migration. In response to multiple signaling pathways, various transcriptional modulation and posttranslational modifications affect the expression level and activity of KLF5. Several transgenic mouse models have revealed the physiological and pathological functions of KLF5 in different cancers. Studies of KLF5 will provide prognostic biomarkers, therapeutic targets, and potential drugs for cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Neoplasms/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Physiological Phenomena , Humans , Kruppel-Like Transcription Factors/chemistry , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Signal Transduction
11.
Cell Mol Life Sci ; 77(8): 1483-1495, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31637449

ABSTRACT

Ubiquitin modification plays significant roles in protein fate determination, signaling transduction, and cellular processes. Over the past 2 decades, the number of studies on ubiquitination has demonstrated explosive growth. E3 ubiquitin ligases are the key enzymes that determine the substrate specificity and are involved in cancer. Several recent studies shed light on the functions and mechanisms of HECTD3 E3 ubiquitin ligase. This review describes the progress in the recent studies of HECTD3 in cancer and other diseases. We propose that HECTD3 is a potential biomarker and a therapeutic target, and discuss the future directions for HECTD3 investigations.


Subject(s)
Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Enzyme Inhibitors/pharmacology , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitination/drug effects
12.
J Biol Chem ; 294(46): 17471-17486, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31594861

ABSTRACT

Constitutive activation of signal transducer and activator of transcription 3 (STAT3) occurs in ∼70% of human cancers, and STAT3 is regarded as one of the most promising targets for cancer therapy. However, specific direct STAT3 inhibitors remain to be developed. Oridonin is an ent-kaurane plant-derived diterpenoid with anti-cancer and anti-inflammatory activities. Here, using an array of cell-based and biochemical approaches, including cell proliferation and apoptosis assays, pulldown and reporter gene assays, site-directed mutagenesis, and molecular dynamics analyses, we report that a thiazole-derived oridonin analogue, CYD0618, potently and directly inhibits STAT3. We found that CYD0618 covalently binds to Cys-542 in STAT3 and suppresses its activity through an allosteric effect, effectively reducing STAT3 dimerization and nuclear translocation, as well as decreasing expression of STAT3-targeted oncogenes. Remarkably, CYD0618 not only strongly inhibited growth of multiple cancer cell lines that harbor constitutive STAT3 activation, but it also suppressed in vivo tumor growth via STAT3 inhibition. Taken together, our findings suggest Cys-542 as a druggable site for selectively inhibiting STAT3 and indicate that CYD0618 represents a promising lead compound for developing therapeutic agents against STAT3-driven diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Diterpenes, Kaurane/pharmacology , Neoplasms/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Diterpenes, Kaurane/chemistry , Diterpenes, Kaurane/therapeutic use , Female , Humans , Mice, Inbred BALB C , Models, Molecular , Neoplasms/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Thiazoles/chemistry , Thiazoles/pharmacology , Thiazoles/therapeutic use
13.
J Biol Chem ; 294(47): 17837-17847, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31624151

ABSTRACT

The Krüppel-like factor 5 (KLF5) transcription factor is highly expressed in basal type breast cancer and promotes breast cancer cell proliferation, survival, migration, and tumorigenesis. KLF5 protein stability is regulated by ubiquitination. In this study, ubiquitin-specific protease 3 (USP3) was identified as a new KLF5 deubiquitinase by genome-wide siRNA library screening. We demonstrated that USP3 interacts with KLF5 and stabilizes KLF5 via deubiquitination. USP3 knockdown inhibits breast cancer cell proliferation in vitro and tumorigenesis in vivo, which can be partially rescued by ectopic expression of KLF5. Furthermore, we observed a positive correlation between USP3 and KLF5 protein expression levels in human breast cancer samples. These findings suggest that USP3 is a new KLF5 deubiquitinase and that USP3 may represent a potential therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Kruppel-Like Transcription Factors/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Animals , Cell Line, Tumor , Cell Proliferation , Female , HEK293 Cells , Humans , Mice, Nude , Protein Binding , Protein Stability , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
14.
Pharmacol Res ; 155: 104748, 2020 05.
Article in English | MEDLINE | ID: mdl-32147403

ABSTRACT

The dysfunction of E3 ubiquitin ligases is important in the pathogenesis of many human diseases, as they play important roles in multiple cellular processes. In this review, we evaluated the structures, functions and clinical significance of two RING-type E3 ubiquitin ligases from the same subfamily, ring-finger protein 126 (RNF126) and breast cancer associated gene 2 (BCA2). Interestingly, the expression of RNF126 and BCA2 are regulated by multiple signaling pathways, including EGFR, ERK, AKT, and NF-κB. RNF126 and BCA2 appear to be functional mediators for not only DNA damage repair but also cancer development. Due to their significant functions in cell proliferation and DNA damage repair, RNF126 and BCA2 may be two potential diagnostic biomarkers and therapeutic targets for cancers.


Subject(s)
Neoplasms/enzymology , Ubiquitin-Protein Ligases/metabolism , Animals , DNA Damage , DNA Repair , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Signal Transduction
15.
Int J Cancer ; 145(5): 1371-1381, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30807646

ABSTRACT

The Cullin 7 (CUL7) gene encodes a member of the cullin family of E3 ubiquitin ligases. Accumulated evidence suggests that CUL7 is oncogenic. However, the mechanism by which CUL7 improves cancer cell survival has not been fully elucidated. Here, we reported that CUL7 confers anti-apoptotic functions by interacting with Caspase-8. CUL7 prevents Caspase-8 activation by promoting Caspase-8 modification with non-degradative polyubiquitin chains at K215. CUL7 knockdown sensitized cancer cells to TRAIL-induced apoptosis in vitro and in nude mice. These results suggest that CUL7 limits extrinsic apoptotic signaling by promoting Caspase-8 ubiquitination.


Subject(s)
Breast Neoplasms/enzymology , Caspase 8/metabolism , Cullin Proteins/metabolism , Uterine Cervical Neoplasms/enzymology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/immunology , Cullin Proteins/genetics , Female , HEK293 Cells , HeLa Cells , Heterografts , Humans , MCF-7 Cells , Mice , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Ubiquitination , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
16.
J Pathol ; 246(4): 497-507, 2018 12.
Article in English | MEDLINE | ID: mdl-30101462

ABSTRACT

Krüpple-like factor 5 (KLF5) is required for the development of the embryo and multiple organs, such as the lung and intestine. KLF5 plays a pro-proliferative and oncogenic role in several carcinomas, including breast cancer. However, its role in normal mammary gland development and oncogenesis has not been elucidated in vivo. In this study, we used mammary gland-specific Klf5 conditional knockout mice derived by mating Klf5-LoxP and MMTV-Cre mice. The genetic ablation of Klf5 suppresses mammary gland ductal elongation and lobuloalveolar formation. Klf5 deficiency inhibits mammary epithelial cell proliferation, survival, and stem cell maintenance. Klf5 promotes mammary stemness, at least partially, by directly promoting the transcription of Slug. Finally, Klf5 depletion suppressed PyMT-induced mammary gland tumor cell stemness, tumor initiation, and growth in vivo. Slug also mediated these functions of Klf5 in vivo. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/metabolism , Animals , Cell Line , Cell Proliferation , Female , Humans , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Tumor Burden
17.
Acta Biochim Biophys Sin (Shanghai) ; 51(10): 1064-1070, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31559416

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with poor clinical outcomes and without effective targeted therapies. Numerous studies have suggested that HDAC inhibitors (TSA/SAHA) may be effective in TNBCs. Proline oxidase, also known as proline dehydrogenase (POX/PRODH), is a key enzyme in the proline metabolism pathway and plays a vital role in tumorigenesis. In this study, we found that HDAC inhibitors (TSA/SAHA) significantly increased POX expression and autophagy through activating AMPK. Depletion of POX decreased autophagy and increased apoptosis induced by HDAC inhibitors in TNBC cells. These results suggest that POX contributes to cell survival under chemotherapeutic stresses and might serve as a potential target for treatment of TNBC.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Proline Oxidase/genetics , Transcriptional Activation/drug effects , Triple Negative Breast Neoplasms/drug therapy , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Female , Humans , Triple Negative Breast Neoplasms/genetics
18.
Acta Biochim Biophys Sin (Shanghai) ; 51(8): 778-790, 2019 Aug 05.
Article in English | MEDLINE | ID: mdl-31314051

ABSTRACT

Chondrogenic differentiation is a coordinated biological process orchestrated by various cell signaling pathways, involving complex pathways regulated at both transcriptional and post-transcriptional levels. Long noncoding RNAs (lncRNAs) are emerging as important regulators in the modulation of multiple cell processes. However, the potential roles of lncRNAs and their regulatory mechanisms in chondrogenic differentiation remain largely unclear. In this study, microarray was performed to detect the expression profiles of lncRNAs and messenger RNAs (mRNAs) during chondrogenic differentiation of murine chondrogenic cell line ATDC5. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to explore their functions. Coding-noncoding co-expression (CNC) and competing endogenous RNA (ceRNA) networks were also constructed with bioinformatics methods. The results revealed that 1009 lncRNAs and 1206 mRNAs were differentially regulated during chondrogenic differentiation. GO and KEGG pathway analysis indicated that the principal functions of the transcripts were associated with system development and extracellular matrix-receptor interaction, TGF-ß signaling, and PI3K-Akt signaling pathways. The CNC network showed that lncRNA AK136902 was positively correlated with prostaglandin F receptor (FP). The ceRNA network covered 3 lncRNAs, 121 miRNAs and 241 edges. The upregulated lncRNA AK136902, AK016344, and ENSMUST00000180767 might promote chondrogenic differentiation by acting as ceRNAs. Knockdown of lncRNA AK136902 could inhibit the mRNA expression of FP and other chondrogenic related genes, including Aggrecan and Col2a1 during chondrogenic differentiation. Our results provide a new perspective on the modulation of lncRNAs during chondrogenic differentiation.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis , Oligonucleotides, Antisense , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Aggrecans/metabolism , Animals , Cell Differentiation , Cell Line , Chondrocytes/cytology , Collagen Type II/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Mice , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Prostaglandin/metabolism , Transforming Growth Factor beta/metabolism
19.
J Cell Mol Med ; 22(11): 5188-5195, 2018 11.
Article in English | MEDLINE | ID: mdl-30145807

ABSTRACT

TNFα-induced protein 2 (TNFAIP2) is a primary response gene of TNFα. TNFAIP2 is highly expressed in immune cells and the urinary bladder. The expression of TNFAIP2 is regulated by multiple transcription factors and signalling pathways, including NF-κB, KLF5 and retinoic acid. Physiologically, TNFAIP2 appears to be a multiple functional mediator not only for inflammation, angiogenesis and tunneling nanotube (TNT) formation but also as a regulator of cell proliferation and migration. The expression of TNFAIP2 is frequently abnormal in human cancers and in infectious diseases. Due to its significant functions in cell proliferation, angiogenesis, migration and invasion, TNFAIP2 could be a potential diagnostic biomarker and therapeutic target for cancer.


Subject(s)
Communicable Diseases/genetics , Cytokines/genetics , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Cell Proliferation/genetics , Communicable Diseases/pathology , Humans , NF-kappa B/genetics , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Signal Transduction , Transcription Factor RelA/genetics , Tumor Necrosis Factor-alpha/genetics
20.
Angiogenesis ; 21(4): 849-860, 2018 11.
Article in English | MEDLINE | ID: mdl-29959560

ABSTRACT

The sprouting of endothelial cells is the first step of tumor angiogenesis. Our previous study suggests that miR-153 suppresses breast tumor angiogenesis partially through targeting hypoxia-induced factor (HIF1α). In this study, we demonstrated that miR-153 also suppresses the migration and the tube formation of endothelial cells through directly targeting angiopoietin 1 (ANG1) in breast cancer cells. There was a negative correlation between miR-153 and ANG1 levels in breast cancer. miR-153 blocked the expression and secretion of ANG1 in breast cancer cells through binding to ANG1 mRNA. Conditioned medium from the breast cancer cell, MCF7, treated with miR-153 had no effect on the proliferation of HUVECs, but significantly inhibited the migration and tube formation of HUVECs, which could be rescued by overexpression of ANG1. In addition, miR-153 also directly inhibited the proliferation and migration of MCF7 through downregulation of ANG1. These findings suggest that miR-153 suppresses the activity of tumor cells and the migration and tube formation of endothelial cells by silencing ANG1.


Subject(s)
Angiopoietin-1/metabolism , Breast Neoplasms , Cell Movement , Endothelial Cells/metabolism , Genes, Tumor Suppressor , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Paracrine Communication , RNA, Neoplasm/metabolism , Angiopoietin-1/genetics , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endothelial Cells/pathology , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , RNA, Neoplasm/genetics
SELECTION OF CITATIONS
SEARCH DETAIL