Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
1.
JHEP Rep ; 6(7): 101087, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38882672

ABSTRACT

Background & Aims: Recent studies demonstrated the importance of fibrosis in promoting an immunosuppressive liver microenvironment and thereby aggressive hepatocellular carcinoma (HCC) growth and resistance to immune checkpoint blockade (ICB), particularly via monocyte-to-monocytic myeloid-derived suppressor cell (M-MDSC) differentiation triggered by hepatic stellate cells (HSCs). We thus aimed to identify druggable targets in these immunosuppressive myeloid cells for HCC therapy. Methods: M-MDSC signature genes were identified by integrated transcriptomic analysis of a human HSC-monocyte culture system and tumor-surrounding fibrotic livers of patients with HCC. Mechanistic and functional studies were conducted using in vitro-generated and patient-derived M-MDSCs. The therapeutic efficacy of a M-MDSC targeting approach was determined in fibrosis-associated HCC mouse models. Results: We uncovered over-expression of protein phosphatase 1 regulatory subunit 15A (PPP1R15A), a myeloid cell-enriched endoplasmic reticulum stress modulator, in human M-MDSCs that correlated with poor prognosis and ICB non-responsiveness in patients with HCC. Blocking TGF-ß signaling reduced PPP1R15A expression in HSC-induced M-MDSCs, whereas treatment of monocytes by TGF-ß upregulated PPP1R15A, which in turn promoted ARG1 and S100A8/9 expression in M-MDSCs and reduced T-cell proliferation. Consistently, lentiviral-mediated knockdown of Ppp1r15a in vivo significantly reduced ARG1+S100A8/9+ M-MDSCs in fibrotic liver, leading to elevated intratumoral IFN-γ+GZMB+CD8+ T cells and enhanced anti-tumor efficacy of ICB. Notably, pharmacological inhibition of PPP1R15A by Sephin1 reduced the immunosuppressive potential but increased the maturation status of fibrotic HCC patient-derived M-MDSCs. Conclusions: PPP1R15A+ M-MDSC cells are involved in immunosuppression in HCC development and represent a novel potential target for therapies. Impact and implications: Our cross-species analysis has identified PPP1R15A as a therapeutic target governing the anti-T-cell activities of fibrosis-associated M-MDSCs (monocytic myeloid-derived suppressor cells). The results from the preclinical models show that specific inhibition of PPP1R15A can break the immunosuppressive barrier to restrict hepatocellular carcinoma growth and enhance the efficacy of immune checkpoint blockade. PPP1R15A may also function as a prognostic and/or predictive biomarker in patients with hepatocellular carcinoma.

2.
Cell Signal ; 120: 111197, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38697447

ABSTRACT

OBJECTIVES: The clinical T1 stage solid lung cancer with metastasis is a serious threat to human life and health. In this study, we performed RNA sequencing on T1 advanced-stage lung cancer and adjacent tissues to identify a novel biomarker and explore its roles in lung cancer. METHODS: Quantitative reversed-transcription PCR, reverse transcription PCR and Western blot, MSP and Methtarget were utilized to evaluate FIBIN expression levels at both the transcriptional and protein levels as well as its methylation status. Differential target protein was evaluated for relative and absolute quantitation by isobaric tags. Co-IP was performed to detect the interactions between target protein. Precise location and expression levels of target proteins were revealed by immunofluorescence staining and component protein extraction using specific kits, respectively. RESULTS: We reported that FIBIN was frequently silenced due to promoter hypermethylation in lung cancer. Additionally, both in vitro and in vivo experiments confirmed the significant anti-proliferation and anti-metastasis capabilities of FIBIN. Mechanistically, FIBIN decreased the nuclear accumulation of ß-catenin by reducing the binding activity of GSK3ß with ANXA2 while promoting interaction between GSK3ß and ß-catenin. CONCLUSION: Our findings firstly identify FIBIN is a tumor suppressor, frequently silenced due to promoter hypermethylation. FIBIN may serve as a predictive biomarker for progression or metastasis among early-stage lung cancer patients.


Subject(s)
Annexin A2 , Carcinoma, Non-Small-Cell Lung , DNA Methylation , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Annexin A2/metabolism , Annexin A2/genetics , Animals , Mice , Cell Line, Tumor , Cell Proliferation , beta Catenin/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Mice, Nude , Neoplasm Metastasis , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Male , Promoter Regions, Genetic/genetics , Female , Mice, Inbred BALB C , A549 Cells , Cell Movement
3.
Clin Breast Cancer ; 2024 May 10.
Article in English | MEDLINE | ID: mdl-38821742

ABSTRACT

Although HER2-low breast cancer (BC) constitutes almost 50% of all BC types, its impact on the pathological complete response (pCR) rate and survival in early BC is uncertain. As a result, a systematic review was conducted to compare the pCR rate and survival of HER2-low and HER2-zero BC in the neoadjuvant chemotherapy (NACT) setting. Two reviewers independently performed literature searches using EMBASE, PubMed, and Cochrane Libraries internet databases up to June 2023. Finally, 29 studies with 178,294 patients were included. HER2-low BC had a considerably lower pCR rate compared to HER2-zero BC in the entire population (Risk Ratio [RR] = 0.68, P < .001) and in the hormone receptor (HR)-positive subgroup (RR = 0.73, P = .009), but not in the HR-negative subgroup (RR = 0.99, P = .755). Furthermore, patients with HER2-low BC exhibited prolonged disease-free survival (DFS) and overall survival (OS) compared to those with HER2-zero BC, observed in both the entire cohort (DFS: P = .004; OS: P = .008) and the HR-negative subgroup (DFS: P = .009; OS: P < .001). In the HR-positive population, OS was superior in HER2-low BC patients (P < .001), whereas no significant differences in DFS were observed (P = .064). Our findings imply that the pCR rate and prognosis of HER2-low BC are distinguished from those of HER2-zero BC in early BC treated with NACT, which contributes to a better knowledge of the BC subgroup.

4.
Genes Dis ; 11(4): 101070, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38523673

ABSTRACT

Protein homeostasis is the basis of normal life activities, and the proteasome family plays an extremely important function in this process. The proteasome 20S is a concentric circle structure with two α rings and two ß rings overlapped. The proteasome 20S can perform both ATP-dependent and non-ATP-dependent ubiquitination proteasome degradation by binding to various subunits (such as 19S, 11S, and 200 PA), which is performed by its active subunit ß1, ß2, and ß5. The proteasome can degrade misfolded, excess proteins to maintain homeostasis. At the same time, it can be utilized by tumors to degrade over-proliferate and unwanted proteins to support their growth. Proteasomes can affect the development of tumors from several aspects including tumor signaling pathways such as NF-κB and p53, cell cycle, immune regulation, and drug resistance. Proteasome-encoding genes have been found to be overexpressed in a variety of tumors, providing a potential novel target for cancer therapy. In addition, proteasome inhibitors such as bortezomib, carfilzomib, and ixazomib have been put into clinical application as the first-line treatment of multiple myeloma. More and more studies have shown that it also has different therapeutic effects in other tumors such as hepatocellular carcinoma, non-small cell lung cancer, glioblastoma, and neuroblastoma. However, proteasome inhibitors are not much effective due to their tolerance and singleness in other tumors. Therefore, further studies on their mechanisms of action and drug interactions are needed to investigate their therapeutic potential.

5.
Virus Res ; 341: 199316, 2024 03.
Article in English | MEDLINE | ID: mdl-38215982

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV) infection is a major public health problem. After HBV infection, viral antigens shift the immune balance in favor of viral escape. Sulforaphane (SFN) is a traditional Chinese medicine.It regulates multi-biological activities, including anti-inflammation, anticancer, and antiviral. However, few studies reported that SFN can inhibit HBV infection before. METHODS: An immunocompetent HBV CBA/CaJ mouse model and a co-culture model were used to explore the effect of SFN on HBV and whether SFN altered the immune balance after HBV infection. RESULTS: We found that SFN was able to reduce HBV DNA, cccDNA, HBsAg, HBeAg, and HBcAg levels in serum and liver tissues of HBV-infected mice. In vitro and in vivo experiments showed that SFN could significantly increase the expression of Cd86 and iNOS and inhibit the expression of Arg1 on macrophages after HBV infection. After SFN administration, Th17 markers in liver tissue and serum were significantly increased. There was no significant changes in the proportion of Treg cells in peripheral blood, but a significant increase in the proportion of Th17 cells and decrease of the Treg/Th17 ratio. Using a network pharmacology approach, we predicted macrophage migration inhibitory factor (MIF) as a potential target of SFN and further validated that MIF expression was significantly increased after HBV infection and SFN significantly inhibited MIF expression both in vitro and in vivo. There was an upward trend in HBV markers (p>0.05) after MIF overexpression. Overexpression of MIF combined with the use of SFN resulted in a significant reversion in the expression of HBV markers and polarization of macrophages towards the M1 phenotype. CONCLUSION: Our results indicated that immunocompetent HBV CBA/CaJ mouse model is a good model to evaluate HBV infection. SFN could inhibit the expression of HBV markers, promote polarization of macrophages towards the M1 phenotype after HBV infection, change the proportion of Treg and Th17 cells. Our findings demonstrate that SFN inhibit HBV infection by inhibiting the expression of MIF and promoting the polarization of macrophages towards the M1 phenotype, which illustrates a promising therapeutic approach in HBV infection.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Isothiocyanates , Macrophage Migration-Inhibitory Factors , Sulfoxides , Animals , Mice , DNA, Viral/metabolism , Hepatitis B virus/genetics , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Mice, Inbred CBA , T-Lymphocytes, Regulatory , Th17 Cells/metabolism
6.
Autophagy ; 20(6): 1467-1469, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38124295

ABSTRACT

Macroautophagy/autophagy is a highly conserved metabolic process that degrades intracellular components and recycles bioenergetic substrates. SQSTM1/p62 (sequestosome 1) is a classical autophagy receptor that participates in selective autophagy to eliminate abnormal intracellular components and recycle bioenergetic substrates. In autophagy, SQSTM1 recruits ubiquitinated substrates to form SQSTM1 droplets and delivers these cargoes to phagophores, the precursors to autophagosomes. Recently, we reported a previously unidentified SQSTM1 S-acylation, which is catalyzed by S-acyltransferase ZDHHC19 and reversed by LYPLA1/APT1. S-acylation of SQSTM1 enhances the affinity of SQSTM1 droplets with the phagophore membrane, thereby promoting efficient autophagic degradation of ubiquitinated substrates. Our study uncovers the role of the S-acylation-deacylation cycle in regulating SQSTM1-mediated selective autophagy.


Subject(s)
Autophagy , Sequestosome-1 Protein , Sequestosome-1 Protein/metabolism , Autophagy/physiology , Acylation , Humans , Animals , Autophagosomes/metabolism
8.
Mol Cell ; 83(19): 3485-3501.e11, 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37802024

ABSTRACT

p62 is a well-characterized autophagy receptor that recognizes and sequesters specific cargoes into autophagosomes for degradation. p62 promotes the assembly and removal of ubiquitinated proteins by forming p62-liquid droplets. However, it remains unclear how autophagosomes efficiently sequester p62 droplets. Herein, we report that p62 undergoes reversible S-acylation in multiple human-, rat-, and mouse-derived cell lines, catalyzed by zinc-finger Asp-His-His-Cys S-acyltransferase 19 (ZDHHC19) and deacylated by acyl protein thioesterase 1 (APT1). S-acylation of p62 enhances the affinity of p62 for microtubule-associated protein 1 light chain 3 (LC3)-positive membranes and promotes autophagic membrane localization of p62 droplets, thereby leading to the production of small LC3-positive p62 droplets and efficient autophagic degradation of p62-cargo complexes. Specifically, increasing p62 acylation by upregulating ZDHHC19 or by genetic knockout of APT1 accelerates p62 degradation and p62-mediated autophagic clearance of ubiquitinated proteins. Thus, the protein S-acylation-deacylation cycle regulates p62 droplet recruitment to the autophagic membrane and selective autophagic flux, thereby contributing to the control of selective autophagic clearance of ubiquitinated proteins.


Subject(s)
Autophagosomes , Ubiquitinated Proteins , Mice , Rats , Humans , Animals , Autophagosomes/metabolism , Ubiquitinated Proteins/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Autophagy/genetics , Acylation , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mammals/metabolism
9.
Ann Med ; 55(2): 2236640, 2023.
Article in English | MEDLINE | ID: mdl-37851510

ABSTRACT

BACKGROUND: The prognostic value of the Gustave Roussy immune (GRIm) score in cancer patients has been widely reported but remains inconsistent. The aim of this study is to systematically investigate the relationship between the GRIm score and survival outcomes in cancer patients. METHODS: Relevant literature was identified using electronic databases including Web of Science, PubMed, and Embase from the inception to March 2023. The primary endpoints were long-term oncological outcomes. Subgroup analysis and sensitivity analysis were conducted during the meta-analysis. RESULTS: Fifteen studies (20 cohorts) including 4997 cancer patients were enrolled. The combined results revealed that patients in the high GRIm group had a deteriorated overall survival (HR = 2.07 95%CI: 1.73-2.48; p < 0.0001; I2 = 62%) and progression-free survival (HR = 1.42; 95%CI: 1.22-1.66; p < 0.0001; I2 = 36%). The prognostic values of GRIm on overall survival and progression-free survival were observed across various tumour types and tumour stages. Sensitivity analysis supported the stability and reliability of the above results. CONCLUSION: Our evidence suggested that the GRIm score could be a valuable prognostic marker in cancer patients, which can be used by clinicians to stratify patients and formulate individualized treatment plans.


Subject(s)
Neoplasms , Humans , Prognosis , Reproducibility of Results , Neoplasms/diagnosis , Progression-Free Survival
10.
Epigenomics ; 15(11): 635-650, 2023 06.
Article in English | MEDLINE | ID: mdl-37554122

ABSTRACT

Background: SCARA5 may play an important role in nasopharyngeal carcinoma. Materials & methods: PCR and immunohistochemistry were used to detect the expression and promoter methylation of SCARA5. Cell proliferation assays, spheroid culture, flow cytometry analysis, Transwell assays and xenotransplantation tests were utilized to determine the functional effects of SCARA5. RNA-sequencing, western blotting, immunofluorescence and dual-luciferase reporter assays were used to assess SCARA5-mediated outcomes. Results: SCARA5 was downregulated by promoter methylation. Overexpression of SCARA5 inhibited cell migration, invasion and proliferation. SCARA5 enhanced nasopharyngeal carcinoma cell sensitivity to chemotherapy with cisplatin and 5-fluorouracil. SCARA5 drives tumor apoptosis by downregulating HSPA2. Conclusion: SCARA5 may be a useful clinical marker in nasopharyngeal carcinoma.


Subject(s)
Nasopharyngeal Neoplasms , Humans , Nasopharyngeal Carcinoma/genetics , Cell Line, Tumor , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Genes, Tumor Suppressor , Cell Proliferation/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism
12.
Genes Dis ; 10(2): 568-582, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37223530

ABSTRACT

Our previous studies found that Zinc-finger protein 382 (ZNF382) played as a tumor suppressor gene in esophageal and gastric cancers, and a positive correlation between the high expression of ZNF382 and better outcome in breast cancer patients. However, the biological roles and mechanisms of ZNF382 in breast cancer remains unclear. We detected ZNF382 expression by reverse-transcription PCR (RT-PCR) and real-time quantitative PCR (qRT-PCR) in breast cancer cells and tissues, and explored the impacts and mechanisms of ectopic ZNF382 expression in breast cancer cells in vitro and in vivo, respectively. Our results revealed that ZNF382 was significantly down-regulated in breast cancer tissues compared with adjacent non-cancer tissues. Restoration of ZNF382 expression in silenced breast cancer cells not only inhibited tumor cell colony formation, viability, migration and invasion, and epithelial-mesenchymal-transition (EMT), but also induced apoptosis and G0/G1 arrest. In conclusion, ZNF382 could induce G0/G1 cell cycle arrest through inhibiting CDC25A signaling, and, inhibit cell migration, invasion and EMT by antagonizing ZEB1 signaling in breast cancer cells.

13.
Clin Epigenetics ; 15(1): 57, 2023 04 01.
Article in English | MEDLINE | ID: mdl-37005662

ABSTRACT

BACKGROUND: The incidence of colorectal cancer (CRC) has increased in recent years. Identification of accurate tumor markers has become the focus of CRC research. Early and frequent DNA methylation tends to occur in cancer. Thus, identifying accurate methylation biomarkers would improve the efficacy of CRC treatment. Neuroglobin (NGB) is involved in neurological and oncological diseases. However, there are currently no reports on epigenetic regulation involvement of NGB in CRC. RESULTS: NGB was downregulated or silenced in majority CRC tissues and cell lines. The hypermethylation of NGB was detected in tumor tissue, but no or a very low methylation frequency in normal tissues. Overexpression of NGB induced G2/M phase arrest and apoptosis, suppressed proliferation, migration, invasion in vitro, and inhibited CRC tumor growth and angiogenesis in vivo. Isobaric tag for relative and absolute quantitation (Itraq)-based proteomics identified approximately 40% proteins related to cell-cell adhesion, invasion, and tumor vessel formation in the tumor microenvironment, among which GPR35 was proved critical for NGB-regulated tumor angiogenesis suppression in CRC. CONCLUSIONS: NGB, an epigenetically silenced factor, inhibits metastasis through the GPR35 in CRC. It is expected to grow into a potential cancer risk assessment factor and a valuable biomarker for early diagnosis and prognosis assessment of CRC.


Subject(s)
Colorectal Neoplasms , DNA Methylation , Humans , Neuroglobin/genetics , Neuroglobin/metabolism , Epigenesis, Genetic , Cell Line, Tumor , Colorectal Neoplasms/pathology , Biomarkers/metabolism , Gene Expression Regulation, Neoplastic , Cell Proliferation , Tumor Microenvironment , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
14.
Spectrochim Acta A Mol Biomol Spectrosc ; 296: 122641, 2023 Aug 05.
Article in English | MEDLINE | ID: mdl-36989691

ABSTRACT

Evaluation of human epidermal growth factor receptor 2 (HER2) molecular markers is a very suitable option for early diagnosis of breast cancer. Metal-organic frameworks (MOFs) have large porosity and surface interactions such as π-π stacking, electrostatics, hydrogen bonding, and coordination. Here, we integrated the HER2 aptamer and fluorescent probe coumarin (COU) with zeolite imidazolic acid framework-8 (ZIF-8) to construct a label-free fluorescent aptamer sensor with pH-gated release of COU. In the presence of the target-HER2, the aptamer adsorbed on the surface of ZIF-8@COU specifically recognizes and falls off the HER2 protein, exposing a portion of the pore size of ZIF-8@COU while reducing the negative charge on the sensor surface, under alkaline hydrolysis conditions, a large number of COU fluorescent molecules can be produced and released in the detection system.The aptamer fluorescence sensor has good detection performance, sensitivity and low background interference, the detection linearity range of HER2 protein is 0.05-10 ng/mL, the detection limit is 0.0005 ng/mL, and it has good recovery rate for the serum detection of clinical breast cancer patients. Therefore, this sensor has high potential in detecting and monitoring HER2 levels for the care and clinical diagnosis of breast cancer patients.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Breast Neoplasms , Humans , Female , Fluorescent Dyes , Breast Neoplasms/diagnosis , Coumarins , Hydrogen-Ion Concentration , Limit of Detection
15.
Cell Death Differ ; 30(5): 1166-1183, 2023 05.
Article in English | MEDLINE | ID: mdl-36813924

ABSTRACT

Peptides/small proteins, encoded by noncanonical open reading frames (ORF) of previously claimed non-coding RNAs, have recently been recognized possessing important biological functions, but largely uncharacterized. 1p36 is an important tumor suppressor gene (TSG) locus frequently deleted in multiple cancers, with critical TSGs like TP73, PRDM16, and CHD5 already validated. Our CpG methylome analysis identified a silenced 1p36.3 gene KIAA0495, previously thought coding long non-coding RNA. We found that the open reading frame 2 of KIAA0495 is actually protein-coding and translating, encoding a small protein SP0495. KIAA0495 transcript is broadly expressed in multiple normal tissues, but frequently silenced by promoter CpG methylation in multiple tumor cell lines and primary tumors including colorectal, esophageal and breast cancers. Its downregulation/methylation is associated with poor survival of cancer patients. SP0495 induces tumor cell apoptosis, cell cycle arrest, senescence and autophagy, and inhibits tumor cell growth in vitro and in vivo. Mechanistically, SP0495 binds to phosphoinositides (PtdIns(3)P, PtdIns(3,5)P2) as a lipid-binding protein, inhibits AKT phosphorylation and its downstream signaling, and further represses oncogenic AKT/mTOR, NF-κB, and Wnt/ß-catenin signaling. SP0495 also regulates the stability of autophagy regulators BECN1 and SQSTM1/p62 through modulating phosphoinositides turnover and autophagic/proteasomal degradation. Thus, we discovered and validated a 1p36.3 small protein SP0495, functioning as a novel tumor suppressor regulating AKT signaling activation and autophagy as a phosphoinositide-binding protein, being frequently inactivated by promoter methylation in multiple tumors as a potential biomarker.


Subject(s)
RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphorylation , DNA Methylation/genetics , Carrier Proteins/metabolism , Genes, Tumor Suppressor , Cell Proliferation/genetics , Cell Line, Tumor , Wnt Signaling Pathway , Autophagy/genetics , Gene Expression Regulation, Neoplastic , DNA Helicases/metabolism , Nerve Tissue Proteins/metabolism
16.
Int J Biol Sci ; 19(2): 641-657, 2023.
Article in English | MEDLINE | ID: mdl-36632451

ABSTRACT

Breast cancer metastasis can happen even when the primary tumor is relatively small. But the mechanism for such early metastasis is poorly understood. Herein, we report that neurotrophin 4 (NTF4) plays a dual role in breast cancer proliferation and metastasis. Clinical data showed high levels of NTF4, especially in the early stage, to be associated with poor clinical outcomes, supporting the notion that metastasis, rather than primary cancer, was the major determinant of breast cancer mortality for patients. NTF4 promoted epithelial-mesenchymal transition (EMT), cell motility, and invasiveness of breast cancer cells in vitro and in vivo. Interestingly, NTF4 inhibited cell proliferation while promoting cellular apoptosis in vitro and inhibited xenograft tumorigenicity in vivo. Mechanistically, NTF4 elicited its pro-metastatic effects by activating PRKDC/AKT and ANXA1/NF-κB pathways to stabilize SNAIL protein, therefore decreasing the level of E-cadherin. Conversely, NTF4 increased ANXA1 phosphorylation and sumoylation and the interaction with importin ß, leading to nuclear import and retention of ANXA1, which in turn activates the caspase-3 apoptosis cascade. Our findings identified an unexpected dual role for NTF4 in breast cancer which contributes to early metastasis of the disease. Therefore, NTF4 may serve as a prognostic marker and a potential therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms , Carcinogenesis , Nerve Growth Factors , Female , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition , Neoplasm Metastasis , Nerve Growth Factors/metabolism , NF-kappa B/metabolism , Carcinogenesis/metabolism
17.
Sci Rep ; 12(1): 19026, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36347994

ABSTRACT

Kruppel like factor 15 (KLF15), a transcriptional factor belonging to the Kruppel-like factor (KLF) family of genes, has recently been reported as a tumor suppressor gene in breast cancer. However, the specific mechanisms by which KLF15 inhibits BrCa have not been elucidated. Here we investigated the role and mechanism of KLF15 in triple-negative breast cancer (TNBC). KLF15 expression and methylation were detected by RT-qPCR, RT-PCR and methylation-specific PCR in breast cancer cell lines and tissues. The effects of KLF15 on TNBC cell functions were examined via various cellular function assays. The specific anti-tumor mechanisms of KLF15 were further investigated by RNA sequence, RT-qPCR, Western blotting, luciferase assay, ChIP, and bioinformatics analysis. As the results showed that KLF15 is significantly downregulated in breast cancer cell lines and tissues, which promoter methylation of KLF15 partially contributes to. Exogenous expression of KLF15 induced apoptosis and G2/M phase cell cycle arrest, suppressed cell proliferation, metastasis and in vivo tumorigenesis of TNBC cells. Mechanism studies revealed that KLF15 targeted and downregulated C-C motif chemokine ligand 2 (CCL2) and CCL7. Moreover, transcriptome and metabolome analysis revealed that KLF15 is involved in key anti-tumor regulatory and metabolic pathways in TNBC. In conclusion, KLF15 suppresses cell growth and metastasis in TNBC by downregulating CCL2 and CCL7. KLF15 may be a prognostic biomarker in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/pathology , Ligands , Cell Proliferation/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Chemokines/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Chemokine CCL7/metabolism , Chemokine CCL2/metabolism
18.
Front Oncol ; 12: 937716, 2022.
Article in English | MEDLINE | ID: mdl-36248995

ABSTRACT

Aberrant protocadherins (PCDHs) expression trigger tumor invasion and metastasis. PCDH20 anti-tumor functions in various tumor have been identified. Tumor suppression is due to Wnt/ß-catenin pathway antagonism and may be suppressed caused by PCDH20 downregulation through promotor methylation, whereas PCDH20 effects and regulation mechanism in esophageal squamous cell carcinoma (ESCC) remains elusive. We analyzed PCDH20 effects on ESCC and underlying action mechanisms for PCDH20. We test PCDH20 expression in ESCC tissues and cells by semi-quantitative PCR (RT-PCR) and q-PCR (real-time quantitative polymerase chain reaction). MSP (methylation-specific PCR) was carried out to assess the methylation of PCDH20 in ESCC cells and tissues. Anti-tumor effects of PCDH20 in vitro were assessed by clone formation assay, CCK8 assay, Transwell assay, and flow cytometry. Nude mice tumorigenicity was used to assess PCDH20 anti-tumor effect in vivo. Online database, qPCR, and Western blotting were used to identify the downregulation of MAP3K9 by PCDH20, associated with AKT/ß-catenin signaling inactivation. We found that PCDH20 expression was dramatically attenuated in esophageal cancer tissues and cells, maybe due to promotor methylation, and ectopic PCDH20 expression suppressed ESCC malignant biological phenotypes. PCDH20 exerted anti-tumor effects by MAP3K9 downregulation, which suppressed AKT/ß-catenin signaling in ESCC cells. Conclusion: PCDH20 was a tumor suppressor gene, which antagonized AKT/ß-catenin signaling pathway in ESCC by decreasing MAP3K9.

19.
Bioelectrochemistry ; 148: 108272, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36183562

ABSTRACT

The dual-signal radiometric sensor can effectively reduce the difference between repeated detection and achieve higher accuracy, sensitivity and repeatability detection. In this work, we constructed a simple ratiometric electrochemical aptasensor based on functionalized ZIF-67 and ZIF-90 for sensitive detection of human epidermal growth factor receptor-2 (HER2). ZIF-67@Ferrocene (Fc)/antimonate nano flakes (AMNFs) as the capture probe has a large specific surface area and good conductivity, and have a strong adsorption capacity for aptamer single-stranded deoxyribonucleic acid (ssDNA). When the biomarker - HER2 interacts with aptamer ssDNA, it is easily desorbed from its surface. At the same time, ZIF-90@ methylene blue (MB) as the signal probe realizes one-step encapsulation of MB signal, which can avoid interference from external environment. When the target-HER2 exists, it is recognized by the capture probe, which leads to the decrease of the conductivity of the electrode. Under the action of the signal probe, the conductivity of the signal is recovered and the detection signal is amplified significantly. The designed ratiometric electrochemical aptasensor showed a wide linear range (0.5-1000 pg mL-1) and a low detection limit (155 fg mL-1) for HER2. Subsequently, it was applied to actual serum samples and showed acceptable applicability. It shows great potential for clinical screening and immediate detection of cancer biomarkers.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , DNA , Electrochemical Techniques , Gold , Humans , Limit of Detection , Metallocenes , Methylene Blue
20.
Genes Dis ; 9(4): 1024-1037, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35685472

ABSTRACT

Recent studies suggest that Hypocretin (HCRT, Orexin) are involved in stress regulation of depression through the hypothalamic-pituitary-adrenal (HPA) axis. However, the molecular mechanism by which Hypocretin regulate neurobiological responses is unknown. Herein, the effects of chronic stress on the epigenetic modification of HCRT and its association with depression were explored with regard to a potential role in cancer progression. In the study, Sprague Dawley (SD) rats were used to establish an animal model of cancer with depression by administrating n-nitrosodiethylamine (DEN) and chronic unpredictable mild stress (CUMS). RNA-sequencing was used to detect differentially expressed genes in the hippocampus of rats and quantitative real-time polymerase chain reaction (qRT-PCR) was used to validate the results of RNA-sequencing. The status of HCRT promoter methylation was assessed by methylation specific polymerase chain reaction. Behavioral tests showed that rats exposed to CUMS had significant depressive-like behaviors. The number of liver tumors and tumor load in depressed rats exposed to CUMS was higher than in SD rats without CUMS. RNA-sequencing revealed that HCRT was one of the most siginificantly downregulated gene in the hippocampus of SD rats with CUMS compared to non-stressed group, which was validated by qRT-PCR. HCRT mRNA expression was downregulated and the promoter for HCRT was hyper-methylated in those with depression. These results identified a critical role for chronic psychological stressors in tumorigenesis and cancer progression, via epigenetic HCRT downregulation. Such epigenetic downregulation may be the molecular basis for the association of cancer with depression.

SELECTION OF CITATIONS
SEARCH DETAIL
...