1
|
Zhong BH, Ma YT, Sun J, Tang JT, Dong M. Transcription factor FOXF2 promotes the development and progression of pancreatic cancer by targeting MSI2. Oncol Rep 2024; 52:93. [PMID: 38847273 PMCID: PMC11177171 DOI: 10.3892/or.2024.8752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/10/2024] [Indexed: 06/16/2024] Open
Abstract
Pancreatic cancer (PC) is a malignant tumor possessing high mortality. The role of transcription factor Forkhead Box F2 (FOXF2) in PC remains unverified. The current study investigated the roles of FOXF2 in developing PC in vitro and in vivo. A xenograft tumor model was constructed with nude mice injected using FOXF2‑overexpressing PC cells or FOXF2‑silenced PC cells. High FOXF2 expression significantly enhanced the proliferation ability of PC cells in vitro and pancreatic tumor growth in vivo. The cell cycle analysis indicated that transition of G1‑S phase was promoted by FOXF2. The cell cycle‑associated proteins cyclin D1, CDK2, phosphorylated (p)‑CDK2 and p‑RB were upregulated in the FOXF2‑overexpressing cells and downregulated in the cells with FOXF2 knockdown. Flow cytometric analysis and Hoechst staining showed that the percentage of apoptotic cells was significantly increased after FOXF2 was silenced. FOXF2 knockdown promoted expression of pro‑apoptotic proteins (Bad, Bax and cleaved caspase‑3) while suppressing the anti‑apoptotic proteins (Bcl‑2 and Bcl‑xl) at the protein level. FOXF2 improved the migration and invasion of PC cells in vitro. Moreover, luciferase and chromatin immunoprecipitation assays revealed that FOXF2 binds to the MSI2 promoter, promoting its transcriptional expression. FOXF2 knockdown inhibited the MSI2 protein translation while enhancing the translation of NUMB protein, suppressing PC development in vivo. MSI2 silencing reversed the promotive effect mediated by FOXF2 on cell proliferation. These results demonstrated that FOXF2 is essential in PC progression, and the potential mechanism includes regulating MSI2 transcription.
Collapse
Affiliation(s)
- Bang-Hua Zhong
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yu-Teng Ma
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jian Sun
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jing-Tong Tang
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ming Dong
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
2
|
Bidooki SH, Navarro MA, Fernandes SCM, Osada J. Thioredoxin Domain Containing 5 (TXNDC5): Friend or Foe? Curr Issues Mol Biol 2024; 46:3134-3163. [PMID: 38666927 PMCID: PMC11049379 DOI: 10.3390/cimb46040197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
This review focuses on the thioredoxin domain containing 5 (TXNDC5), also known as endoplasmic reticulum protein 46 (ERp46), a member of the protein disulfide isomerase (PDI) family with a dual role in multiple diseases. TXNDC5 is highly expressed in endothelial cells, fibroblasts, pancreatic β-cells, liver cells, and hypoxic tissues, such as cancer endothelial cells and atherosclerotic plaques. TXNDC5 plays a crucial role in regulating cell proliferation, apoptosis, migration, and antioxidative stress. Its potential significance in cancer warrants further investigation, given the altered and highly adaptable metabolism of tumor cells. It has been reported that both high and low levels of TXNDC5 expression are associated with multiple diseases, such as arthritis, cancer, diabetes, brain diseases, and infections, as well as worse prognoses. TXNDC5 has been attributed to both oncogenic and tumor-suppressive features. It has been concluded that in cancer, TXNDC5 acts as a foe and responds to metabolic and cellular stress signals to promote the survival of tumor cells against apoptosis. Conversely, in normal cells, TXNDC5 acts as a friend to safeguard cells against oxidative and endoplasmic reticulum stress. Therefore, TXNDC5 could serve as a viable biomarker or even a potential pharmacological target.
Collapse
Affiliation(s)
- Seyed Hesamoddin Bidooki
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (S.H.B.); (M.A.N.)
- Centre National de la Recherche Scientifique (CNRS), Institute of Analytical Sciences and Physico-Chemistry for Environment and Materials (IPREM), Universite de Pau et des Pays de l’Adour, E2S UPPA, 64 000 Pau, France;
- MANTA—Marine Materials Research Group, Universite de Pau et des Pays de l’Adour, E2S UPPA, 64 600 Anglet, France
| | - María A. Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (S.H.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Susana C. M. Fernandes
- Centre National de la Recherche Scientifique (CNRS), Institute of Analytical Sciences and Physico-Chemistry for Environment and Materials (IPREM), Universite de Pau et des Pays de l’Adour, E2S UPPA, 64 000 Pau, France;
- MANTA—Marine Materials Research Group, Universite de Pau et des Pays de l’Adour, E2S UPPA, 64 600 Anglet, France
| | - Jesus Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, E-50013 Zaragoza, Spain; (S.H.B.); (M.A.N.)
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, E-50013 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
3
|
Bae J, Kitayama S, Herbert Z, Daheron L, Kurata K, Keskin DB, Livak K, Li S, Tarannum M, Romee R, Samur M, Munshi NC, Kaneko S, Ritz J, Anderson KC. Differentiation of BCMA-specific induced pluripotent stem cells into rejuvenated CD8αβ+ T cells targeting multiple myeloma. Blood 2024; 143:895-911. [PMID: 37890146 PMCID: PMC10940063 DOI: 10.1182/blood.2023020528] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
ABSTRACT A major hurdle in adoptive T-cell therapy is cell exhaustion and failure to maintain antitumor responses. Here, we introduce an induced pluripotent stem cell (iPSC) strategy for reprogramming and revitalizing precursor exhausted B-cell maturation antigen (BCMA)-specific T cells to effectively target multiple myeloma (MM). Heteroclitic BCMA72-80 (YLMFLLRKI)-specific CD8+ memory cytotoxic T lymphocytes (CTL) were epigenetically reprogrammed to a pluripotent state, developed into hematopoietic progenitor cells (CD34+ CD43+/CD14- CD235a-), differentiated into the T-cell lineage and evaluated for their polyfunctional activities against MM. The final T-cell products demonstrated (1) mature CD8αβ+ memory phenotype, (2) high expression of activation or costimulatory molecules (CD38, CD28, and 41BB), (3) no expression of immune checkpoint and senescence markers (CTLA4, PD1, LAG3, and TIM3; CD57), and (4) robust proliferation and polyfunctional immune responses to MM. The BCMA-specific iPSC-T cells possessed a single T-cell receptor clonotype with cognate BCMA peptide recognition and specificity for targeting MM. RNA sequencing analyses revealed distinct genome-wide shifts and a distinctive transcriptional profile in selected iPSC clones, which can develop CD8αβ+ memory T cells. This includes a repertoire of gene regulators promoting T-cell lineage development, memory CTL activation, and immune response regulation (LCK, IL7R, 4-1BB, TRAIL, GZMB, FOXF1, and ITGA1). This study highlights the potential application of iPSC technology to an adaptive T-cell therapy protocol and identifies specific transcriptional patterns that could serve as a biomarker for selection of suitable iPSC clones for the successful development of antigen-specific CD8αβ+ memory T cells to improve the outcome in patients with MM.
Collapse
Affiliation(s)
- Jooeun Bae
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Shuichi Kitayama
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Zach Herbert
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | | | - Keiji Kurata
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Derin B. Keskin
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Kenneth Livak
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Shuqiang Li
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Mubin Tarannum
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Rizwan Romee
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Mehmet Samur
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Nikhil C. Munshi
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Shin Kaneko
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Jerome Ritz
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| | - Kenneth C. Anderson
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
4
|
Lou F, Zhang M. RFC2 promotes aerobic glycolysis and progression of colorectal cancer. BMC Gastroenterol 2023; 23:353. [PMID: 37821801 PMCID: PMC10566032 DOI: 10.1186/s12876-023-02984-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Replication factor C subunit 2 (RFC2) participates in the growth and metastasis of various malignancies. Our study investigated the roles of RFC2 in colorectal cancer (CRC). RESULTS RFC2 expression was upregulated in CRC tissues and cells. High RFC2 expression was associated with poor prognosis. Knockdown RFC2 inhibited proliferation, induced apoptosis, and suppressed migration and invasion of CRC cells. CREB5 was a transcription factor of RFC2, and CREB5 knockdown suppressed RFC2 expression. Furthermore, RFC2 promoted aerobic glycolysis and MET/PI3K/AKT/mTOR pathway. CONCLUSION RFC2 promoted the progression of CRC cells via activating aerobic glycolysis and the MET/PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Fuchen Lou
- Department of Endocrinology, The Second Hospital of Shandong University, Jinan, Shandong, 250033, P.R. China
| | - Mingbao Zhang
- Department of Gastroenterology, The Second Hospital of Shandong University, Beiyuan Street 247,Tianqiao District, Jinan, Shandong, 250033, P.R. China.
| |
Collapse
|
5
|
Wang G, Ni X, Wang J, Dai M. METTL3-mediated m 6A methylation of PYGB facilitates pancreatic ductal adenocarcinoma progression through the activation of NF-κB signaling. Pathol Res Pract 2023; 248:154645. [PMID: 37422970 DOI: 10.1016/j.prp.2023.154645] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 05/23/2023] [Accepted: 06/25/2023] [Indexed: 07/11/2023]
Abstract
Brain Type Glycogen Phosphorylase (PYGB) has been revealed to participate in the progression of multiple human cancers. Nevertheless, the clinical significance and biological function of PYGB in pancreatic ductal adenocarcinoma (PAAD) remains unclarified. This study first analyzed the expression pattern, diagnostic value, and prognostic significance of PYGB in PAAD using the TCGA database. Subsequently, western blot assessed the protein expression of genes in PAAD cells. The viability, apoptosis, migration, and invasion of PAAD cells were assessed by CCK-8, TUNEL, and Transwell assays. Finally, in vivo experiment evaluated the effect of PYGB on PAAD tumor growth and metastasis. Through our investigation, it was revealed that PYGB had extremely high expression in PAAD and predicted a worse prognosis in patients with PAAD. Besides, the aggressiveness of PAAD cells could be suppressed or enhanced by depleting or supplementing PYGB. In addition, we demonstrated that METTL3 enhanced the translation of PYGB mRNA in an m6A-YTHDF1-dependent manner. Moreover, PYGB was revealed to regulate the malignant behaviors of PAAD cells by the mediation of the NF-κB signaling. Finally, PYGB depletion suppressed the growth and distant metastasis of PAAD in vivo. To conclude, our results indicated that METTL3-mediated m6A modification of PYGB exerted the tumor-promotive effect on PAAD through NF-κB signaling, suggesting PYGB is a potential therapeutic target in PAAD.
Collapse
Affiliation(s)
- Guoying Wang
- Department of Gastroenterology, Liyang People's Hospital, Liyang City, Jiangsu, China
| | - Xin Ni
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Jintian Wang
- Department of Gastroenterology, Liyang People's Hospital, Liyang City, Jiangsu, China
| | - Ming Dai
- Department of Gastroenterology, Liyang People's Hospital, Liyang City, Jiangsu, China.
| |
Collapse
|
6
|
Deng L, Xu G, Huang Q. Comprehensive analyses of the microRNA-messenger RNA-transcription factor regulatory network in mouse and human renal fibrosis. Front Genet 2022; 13:925097. [PMID: 36457754 PMCID: PMC9705735 DOI: 10.3389/fgene.2022.925097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/14/2022] [Indexed: 09/19/2023] Open
Abstract
Objective: The aim of this study was to construct a microRNA (miRNA)-messenger RNA (mRNA)-transcription factor (TF) regulatory network and explore underlying molecular mechanisms, effective biomarkers, and drugs in renal fibrosis (RF). Methods: A total of six datasets were downloaded from Gene Expression Omnibus. "Limma" and "DESeq2" packages in R software and GEO2R were applied to identify the differentially expressed miRNAs and mRNAs (DEmiRNAs and DEmRNAs, respectively). The determination and verification of DEmiRNAs and DEmRNAs were performed through the integrated analysis of datasets from five mouse 7 days of unilateral ureteral obstruction datasets and one human chronic kidney disease dataset and the Human Protein Atlas (http://www.proteinatlas.org). Target mRNAs of DEmiRNAs and TFs were predicted by prediction databases and the iRegulon plugin in Cytoscape, respectively. A protein-protein interaction network was constructed using STRING, Cytoscape v3.9.1, and CytoNCA. Functional enrichment analysis was performed by DIANA-miRPath v3.0 and R package "clusterProfiler." A miRNA-mRNA-TF network was established using Cytoscape. Receiver operating characteristic (ROC) curve analysis was used to examine the diagnostic value of the key hub genes. Finally, the Comparative Toxicogenomics Database and Drug-Gene Interaction database were applied to identify potential drugs. Results: Here, 4 DEmiRNAs and 11 hub genes were determined and confirmed in five mouse datasets, of which Bckdha and Vegfa were further verified in one human dataset and HPA, respectively. Moreover, Bckdha and Vegfa were also predicted by miR-125a-3p and miR-199a-5p, respectively, in humans as in mice. The sequences of miR-125a-3p and miR-199a-5p in mice were identical to those in humans. A total of 6 TFs were predicted to regulate Bckdha and Vegfa across mice and humans; then, a miRNA-mRNA-TF regulatory network was built. Subsequently, ROC curve analysis showed that the area under the curve value of Vegfa was 0.825 (p = 0.002). Finally, enalapril was identified to target Vegfa for RF therapy. Conclusion: Pax2, Pax5, Sp1, Sp2, Sp3, and Sp4 together with Bckdha-dependent miR-125a-3p/Vegfa-dependent miR-199a-5p formed a co-regulatory network enabling Bckdha/Vegfa to be tightly controlled in the underlying pathogenesis of RF across mice and humans. Vegfa could act as a potential novel diagnostic marker and might be targeted by enalapril for RF therapy.
Collapse
Affiliation(s)
- Le Deng
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Qipeng Huang
- Department of Nephrology, The Fifth Affiliated Hospital of Jinan University, Heyuan, China
| |
Collapse
|
7
|
Nalla LV, Gondaliya P, Kalia K, Khairnar A. Targeting specificity protein 1 with miR-128-3p overcomes TGF-β1 mediated epithelial-mesenchymal transition in breast cancer: An in vitro study. Mol Biol Rep 2022; 49:6987-6996. [PMID: 35486287 DOI: 10.1007/s11033-022-07466-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/08/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Specificity protein 1 (SP1) was found to play a critical role in the regulation of TGF-β1 driven epithelial-mesenchymal transition (EMT). Recent clinical findings demonstrated a significant drop in the expression of miR-128-3p with the cancer progression in breast cancer patients. However, the impact of miR-128-3p on the SP1 expression in breast cancer remains unknown. Herein, we evaluated the role of miR-128-3p mimics in suppressing EMT of breast cancer cell lines by regulating the TGF-β1/SP1 axis. METHODS miR-128-3p interaction with SP1 was detected by in silico tools and dual-luciferase reporter assay. qPCR, western blot, and immunocytochemistry experiments were conducted for determining the expression levels of miR-128-3p and EMT markers with and without the treatment of miR-128-3p mimics. Further, to understand the effect of miR-128-3p mimics on cancer progression, experiments such as wound healing assay, transwell assay, adhesion assay, and cell cycle analysis were performed. RESULTS A significant inverse relation between SP1 and miR-128-3p levels was found in MCF-7 and MDA-MB-231 cell lines. miR-128-3p overexpression impeded the SP1 mediated EMT markers in TGF-β1 stimulated cells by inhibiting the SP1 nuclear function. Further, treatment with miR-128-3p mimics significantly reduced the migration, invasion and spreading capability of TGF-β1 stimulated cells. Flow cytometry results showed the impeding role of miR-128-3p on the cell cycle progression. CONCLUSIONS Upregulated miR-128-3p inhibited SP1, thereby limiting the TGF-β1 induced EMT in MCF-7 and MDA-MB-231 cell lines for the first time. This study may pave the path to explore novel miRNA therapeutics for eradicating advanced breast cancer cases.
Collapse
Affiliation(s)
- Lakshmi Vineela Nalla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Palaj, Gandhinagar, Gujarat, 382355, India
| | - Piyush Gondaliya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gandhinagar, Gujarat, 382355, India
| | - Kiran Kalia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gandhinagar, Gujarat, 382355, India
| | - Amit Khairnar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|