Min Wang’s research while affiliated with Northeastern University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (5)


Identification of DEGs and their functional annotation in serous ovarian cancer. a GO and KEGG enrichment analysis of up-regulated DEGs in GSE14407 serous ovarian cancer database. Raw data between serous ovarian cancer and normal ovarian tissue were obtained from the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/gds/) dataset and the gene expression profiling (GSE14407) was selected. Differentially expressed genes (DEGs) were selected by |log2FC|> 1 and adj. P < 0.05 in the GSE14407 dataset. Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis software were R packages ggplot2. b Enrichment fraction diagram of GO-GSEA in the GSE14407 dataset. c Enrichment fraction diagram of KEGG-GSEA in the GSE14407 dataset. Gene Set Enrichment Analysis (GSEA) analysis software was R package clusterProfiler + enrichplot. GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Gene and Genome; DEG, differentially expressed genes; GSEA, Gene Set Enrichment Analysis
ZNF384 is highly expressed in serous ovarian cancer. a Circos plot showed the location of up-regulated genes in chromosomes of the GSE14407 dataset. The outermost track showed the chromosome information of these genes. The box plot in the middle of the Circos plot showed the expression of ZNF384 in normal and cancer tissue in the GSE14407 database. b Relative expression of ZNF384 in 30 serous ovarian cancer tissues and 30 normal ovarian tissues by qPCR. c The genomic profile of ZNF384 was obtained from the serous ovarian cancer data set (TCGA) using cBioportal database (https://www.cbioportal.org/). d The association between ZNF384 expression and OS, PFS, DFI, and DSS in serous ovarian cancer from the GSCA database (https://guolab.wchscu.cn/GSCA/). e Immunohistochemistry staining images of high and low expression of ZNF384. The scale bar indicates 50 µm. TCGA, The Cancer Genome Atlas; GSCA, Gene Set Cancer Analysis; OS, overall survival; PFS, progression-free survival; DFI, disease-free interval; DSS, disease-specific survival. ****p < 0.0001 as compared to the Normal group. Data are presented as mean ± SD. qPCR, quantitative real-time PCR
ZNF384 promotes the growth of serous ovarian cancer cells in vitro. a Western blot analysis of ZNF384 expression in normal ovarian epithelial cells (IOSE-80) and 5 lines of serous ovarian cancer cells (OVCAR-8, HEY A8, OVCAR-3, HEY and SKOV3). b Western blot and qPCR analysis of ZNF384 expression in OVCAR-8 and OVCAR-3 cells with ZNF384 knockdown. c Analysis of ZNF384 expression in SKOV3 cells with ZNF384 overexpression. ****p < 0.0001 as compared to the shCon or Vector group. d Cell proliferation of OVCAR-8, OVCAR-3, and SKOV3 cells after lentivirus infection was detected by CCK8 assay. *p < 0.05, **p < 0.01 as compared to the shCon or Vector group. e Colony formation assay of OVCAR-8, OVCAR-3, and SKOV3 cells was performed. The experiments were repeated three times. **p < 0.01, ***p < 0.001, ****p < 0.0001 as compared to the shCon or Vector group. Data are presented as mean ± SD. qPCR, quantitative real-time PCR
ZNF384 promotes the invasion and migration of serous ovarian cancer cells. a The effects of ZNF384 on the migration of serous ovarian cancer cells by wound healing assay. Photographs showed cell migration before and after injury under the microscope at 100 × magnification field. Quantification of cell migration by measuring wound closure areas before and after injury. b Effect of ZNF384 on the invasion of serous ovarian cancer cells by transwell assay (crystal violet staining × 200). c MMP-2 levels in the supernatants of OVCAR-8, OVCAR-3, and SKOV3 cells were detected by ELISA assay. *p < 0.05, **p < 0.01, ****p < 0.0001 as compared to the shCon or Vector group. d Representative immunofluorescence images of vimentin expression (red) in OVCAR-8, OVCAR-3, and SHOV3 cells. Cell nuclei were stained by DAPI (blue). The scale bar indicates 50 µm. Data are presented as mean ± SD. DAPI, 4′6’-diamidino-2-phenylindole dihydrochloride
ZNF384 promotes tumor growth and metastasis in vivo. a-c Xenograft tumors were formed by subcutaneous injection of OVCAR-8 or SKOV3 cells into BALB/c nude mice. The tumors were stripped after 25 days, photographed, measured, and weighed. *p < 0.05, **p < 0.01, ****p < 0.0001 as compared to the shCon or Vector group. d Immunohistochemical staining images of ZNF384, Ki67 expression in tumor tissues. The scale bar indicates 50 µm. e Bioluminescence images of BALB/c nude mice after intraperitoneal injection of OVCAR-8 or SKOV3 cells. f Representative images of abdominal cavity metastasis of tumors in different groups of nude mice (left panel) and quantification of the number of tumor metastatic nodules (right panel). ***p < 0.001, ****p < 0.0001 as compared to the shCon or Vector group. Data are presented as mean ± SD

+3

The pivotal role of ZNF384: driving the malignant behavior of serous ovarian cancer cells via the LIN28B/UBD axis
  • Article
  • Full-text available

November 2024

·

2 Reads

Cell Biology and Toxicology

Ye Yang

·

Runze He

·

Dongxiao Li

·

[...]

·

Min Wang

Zinc finger protein 384 (ZNF384) is a highly conserved transcribed gene associated with the development of multiple tumors, however, its role and mechanism in serous ovarian cancer (SOC) are unknown. We first confirmed that ZNF384 was abnormally highly expressed in SOC tissues by bioinformatics analysis and immunohistochemistry. We further used lentivirus packaging and transfection techniques to construct ZNF384 overexpression or knockdown cell lines, and through a series of cell function experiments, gradually verified that ZNF384 promoted a series of malignant behaviors of SOC cell proliferation, migration, and invasion. By establishing a xenotransplantation model in nude mice, it was confirmed that ZNF384 promoted the progress of SOC in vivo. Mechanistically, Overexpression of ZNF384 enhanced the transcriptional activity of Lin-28 homolog B (LIN28B), which promoted the malignant behavior of SOC cells. In addition, LIN28B could regulate the expression of the downstream factor ubiquitin D (UBD) in SOC cells, further promoting the development of SOC. This study shows that ZNF384 aggravates the malignant behavior of SOC cells through the LIN28B/UBD axis, which may be used as a diagnostic biomarker for patients with SOC. Graphical abstract

Download

Advances in natural products modulating autophagy influenced by cellular stress conditions and their anticancer roles in the treatment of ovarian cancer

October 2024

·

56 Reads

·

2 Citations

Autophagy is a conservative catabolic process that typically serves a cell‐protective function. Under stress conditions, when the cellular environment becomes unstable, autophagy is activated as an adaptive response for self‐protection. Autophagy delivers damaged cellular components to lysosomes for degradation and recycling, thereby providing essential nutrients for cell survival. However, this function of promoting cell survival under stress conditions often leads to malignant progression and chemotherapy resistance in cancer. Consequently, autophagy is considered a potential target for cancer therapy. Herein, we aim to review how natural products act as key modulators of autophagy by regulating cellular stress conditions. We revisit various stressors, including starvation, hypoxia, endoplasmic reticulum stress, and oxidative stress, and their regulatory relationship with autophagy, focusing on recent advances in ovarian cancer research. Additionally, we explore how polyphenolic compounds, flavonoids, alkaloids, terpenoids, and other natural products modulate autophagy mediated by stress responses, affecting the malignant biological behavior of cancer. Furthermore, we discuss their roles in ovarian cancer therapy. This review emphasizes the importance of natural products as valuable resources in cancer therapeutics, highlighting the need for further exploration of their potential in regulating autophagy. Moreover, it provides novel insights and potential therapeutic strategies in ovarian cancer by utilizing natural products to modulate autophagy.


STAU1-mediated CNBP mRNA degradation by LINC00665 alters stem cell characteristics in ovarian cancer

July 2024

·

21 Reads

·

1 Citation

Biology Direct

Background To investigate the role of lncRNA LINC00665 in modulating ovarian cancer stemness and its influence on treatment resistance and cancer development. Methods We isolated ovarian cancer stem cells (OCSCs) from the COC1 cell line using a combination of chemotherapeutic agents and growth factors, and verified their stemness through western blotting and immunofluorescence for stem cell markers. Employing bioinformatics, we identified lncRNAs associated with ovarian cancer, with a focus on LINC00665 and its interaction with the CNBP mRNA. In situ hybridization, immunohistochemistry, and qPCR were utilized to examine their expression and localization, alongside functional assays to determine the effects of LINC00665 on CNBP. Results LINC00665 employs its Alu elements to interact with the 3’-UTR of CNBP mRNA, targeting it for degradation. This molecular crosstalk enhances stemness by promoting the STAU1-mediated decay of CNBP mRNA, thereby modulating the Wnt and Notch signaling cascades that are pivotal for maintaining CSC characteristics and driving tumor progression. These mechanistic insights were corroborated by a series of in vitro assays and validated in vivo using tumor xenograft models. Furthermore, we established a positive correlation between elevated CNBP levels and increased disease-free survival in patients with ovarian cancer, underscoring the prognostic value of CNBP in this context. Conclusions lncRNA LINC00665 enhances stemness in ovarian cancer by mediating the degradation of CNBP mRNA, thereby identifying LINC00665 as a potential therapeutic target to counteract drug resistance and tumor recurrence associated with CSCs.


An LRPPRC-HAPSTR1-PSMD14 interaction regulates tumor progression in ovarian cancer

April 2024

·

20 Reads

·

2 Citations

Aging

Ovarian cancer is the second most common cause of gynecologic cancer death. Chemoresistance and metastasis remain major challenges for current treatment. Previously, HAPSTR1 was shown to be a target gene of a paclitaxel resistance-associated miRNA. However, the biological function and underlying molecular mechanisms of HAPSTR1 in ovarian cancer progression remain unclear. Herein, we aimed to measure HAPSTR1 expression in ovarian cancer specimens and examine its correlations with clinical features and key functional interactions with other genes and proteins. An immunohistochemistry assay showed that HAPSTR1 was overexpressed in ovarian cancer tissues and was significantly associated with the FIGO stage and clinical outcome. HAPSTR1 overexpression promoted proliferation, invasion and migration in cellular and mouse models, whereas inhibition induced the opposite effects. In addition, HAPSTR1 stimulated the EMT pathway and affected the expression of autophagy biomarkers. Mechanistically, we demonstrated that HAPSTR1 is bound to LRPPRC and PSMD14 via immunoprecipitation. HAPSTR1 suppressed LRPPRC ubiquitination and recruited PSMD14 to interact with LRPPRC. Moreover, LRPPRC knockdown reversed HAPSTR1-mediated improvement in cellular proliferation, invasion, and migration. Our study is the first detailed and comprehensive analysis of HAPSTR1 in cancer progression and offers an experimental basis for the clinical treatment of ovarian carcinoma.


Figure 2. Mechanisms of action for the presence of GPX3 in cancer. (A) Hypermethylation of the CpG island of the GPX3 promoter results in decreased GPX3 expression, leading to the proliferation of tumor cells. (B) Modifications in GPX3 play a crucial role in the regulation of various signaling pathways in tumors, including NF-κB, Wnt-β connexin, and JNK signaling. (C) SNPs are predominantly located in the non-coding region of the GPX3 gene and have been shown to have a positive association with cancer risk. GPX3 rs3805435 and rs3828599 have been linked to the development of gastric cancer, and GPX3 rs736775 may serve as a prognostic marker. (D) GPX3 can actively scavenge ROS within the TME, thereby enhancing the removal of soluble lipid hydroperoxides present in the extracellular tumor environment. This process protects the tumor cells. GPX3, glutathione peroxidase 3; CpG, cytosine, and guanine; mRNA, messenger RNA; Erk, extracellular regulated protein kinases; NF-κB, nuclear factor-κB; Wnt, Wingless; SIP1, Smad interacting protein 1; JNK, c-Jun N-terminal kinase; MMP2, matrix metalloproteinase 2; SNP, single nucleotide polymorphism; ROS, reactive oxygen species; CSC, cancer stem cell; TME, tumor microenvironment.
Advances in the role of GPX3 in ovarian cancer (Review)

January 2024

·

29 Reads

·

4 Citations

International Journal of Oncology

Ovarian cancer (OC) is the 5th most common malignancy in women, and the leading cause of death from gynecologic malignancies. Owing to tumor heterogeneity, lack of reliable early diagnostic methods and high incidence of chemotherapy resistance, the 5-year survival rate of patients with advanced OC remains low despite considerable advances in detection and therapeutic approaches. Therefore, identifying novel therapeutic targets to improve the prognosis of patients with OC is crucial. The expression of glutathione peroxidase 3 (GPX3) plays a crucial role in the growth, proliferation and differentiation of various malignant tumors. In OC, GPX3 is the only antioxidant enzyme the high expression of which is negatively correlated with the overall survival of patients. GPX3 may affect lipid metabolism in tumor stem cells by influencing redox homeostasis in the tumor microenvironment. The maintenance of stemness in OC stem cells (OCSCs) is strongly associated with poor prognosis and recurrence in patients. The aim of the present study was to review the role of GPX3 in OC and investigate the potential factors and effects of GPX3 on OCSCs. The findings of the current study offer novel potential targets for drug therapy in OC, enhance the theoretical foundation of OC drug therapy and provide valuable references for clinical treatment.

Citations (4)


... This inhibition results in mTORC1 activation, which inhibits autophagy by phosphorylating ULK1, hence obstructing autophagosome formation [16]. In contrast, under stress circumstances, such as food restriction or hypoxia, PI3K/AKT signaling is suppressed, resulting in mTORC1 inhibition, which activates ULK1 and initiates autophagy [17]. This transition allows cells to decompose and reutilize intracellular constituents to sustain energy equilibrium and viability. ...

Reference:

A Comprehensive Review of Nanoparticle-Based Drug Delivery for Modulating PI3K/AKT/mTOR-Mediated Autophagy in Cancer
Advances in natural products modulating autophagy influenced by cellular stress conditions and their anticancer roles in the treatment of ovarian cancer

... The staufen double-stranded RNA binding protein 1 (STAU1), a multifunctional protein primarily involved in mRNA transport and localization, has recently emerged as a potential biomarker in COPD. 4 It has been implicated in various cellular processes, including autophagy and stress responses. Previous studies linked STAU1 to neurodegenerative diseases, where its overexpression was shown to disrupt autophagy and regulate the mechanistic target of rapamycin (mTOR), which plays a critical role in cellular growth and survival. ...

STAU1-mediated CNBP mRNA degradation by LINC00665 alters stem cell characteristics in ovarian cancer

Biology Direct

... Such observations are in keeping with the correlation between RPN11/PSMD14 and LRPPRC expression in clinical specimens [76]. The C16orf72/HUWE1associated protein modifying stress responses (HAPSTR1) has also been implicated in sustaining tumor progression in ovarian cancer through stimulation of epithelial mesenchymal transition (EMT) and inhibition of autophagy [77]. Indeed, HAPSTR1 colocalizes with LRPPRC and inhibits its ubiquitination, with HAPSTR1 overexpression stabilizing LRPPRC through RPN11/PSMD14. ...

An LRPPRC-HAPSTR1-PSMD14 interaction regulates tumor progression in ovarian cancer

Aging

... Glutathione peroxidase 3 (GPX3), as a member of the GPX family, can scavenge free radicals such as hydrogen peroxide for achieving the goal of protecting cells. Some studies have shown that a high expression of GPX3 can inhibit the development of malignant tumours such as gastric cancer and endometrial cancer, but for ovarian cancer, on the contrary, a high expression of GPX3 promotes the progression and recurrence of ovarian cancer and reduces the survival rate of patients [126,127]. ...

Advances in the role of GPX3 in ovarian cancer (Review)

International Journal of Oncology