1
|
Zhou W, Deng X, Wang B, Yuan Y, Ma J, Meng X. HTF4 modulates the transcription of GID2 to promote the malignant biological behavior of pancreatic cancer. Pancreatology 2024; 24:1073-1083. [PMID: 39216997 DOI: 10.1016/j.pan.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Helix-loop-helix transcription factor 4 (HTF4) as an anti-cancer target has been reported in many human cancers, but limited data exists regarding the effect of HTF4 in pancreatic cancer. In this study, we aimed to investigate the role of HTF4 in pancreatic cancer. METHODS The expression levels of HTF4 in clinical pancreatic cancer samples were measured. HTF4 was knocked down or overexpressed in pancreatic cancer cells and was subsequently tested for bio-function using in vitro assays and in vivo. The regulation of HTF4 on GID2 was assessed via bioinformatic tools and dual-luciferase reporter assay. RESULTS We found that HTF4 was highly expressed in pancreatic cancer tissues and correlated with poor patient prognosis. In addition, knocking down HTF4 expression inhibited cell proliferation, migration, and invasion, whereas HTF4 overexpression exerted the opposite effect. Moreover, HTF4 promoted tumor growth and metastasis in pancreatic cancer. Further, HTF4 bound to the GID2 promoter region and promoted transcriptional activation of GID2 in pancreatic cancer cells. GID2 knockdown suppressed HTF4-induced malignant behaviors of pancreatic cancer cells. CONCLUSIONS Our findings suggest that the HTF4/GID2 axis accelerates the progression of pancreatic cancer, providing a potential therapeutic target and prognostic indicator for the treatment of pancreatic cancer patients.
Collapse
Affiliation(s)
- Wenyang Zhou
- Department of Pathology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xin Deng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Baosheng Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yifeng Yuan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jia Ma
- Department of Gastroenterology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Xiangpeng Meng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
2
|
Yang S, Zhou P, Zhang L, Xie X, Zhang Y, Bo K, Xue J, Zhang W, Liao F, Xu P, Hu Y, Yan R, Liu D, Chang J, Zhou K. VAMP8 suppresses the metastasis via DDX5/β-catenin signal pathway in osteosarcoma. Cancer Biol Ther 2023; 24:2230641. [PMID: 37405957 DOI: 10.1080/15384047.2023.2230641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/07/2023] Open
Abstract
Osteosarcoma is a highly metastatic malignant bone tumor, necessitating the development of new treatments to target its metastasis. Recent studies have revealed the significance of VAMP8 in regulating various signaling pathways in various types of cancer. However, the specific functional role of VAMP8 in osteosarcoma progression remains unclear. In this study, we observed a significant downregulation of VAMP8 in osteosarcoma cells and tissues. Low levels of VAMP8 in osteosarcoma tissues were associated with patients' poor prognosis. VAMP8 inhibited the migration and invasion capability of osteosarcoma cells. Mechanically, we identified DDX5 as a novel interacting partner of VAMP8, and the conjunction of VAMP8 and DDX5 promoted the degradation of DDX5 via the ubiquitin-proteasome system. Moreover, reduced levels of DDX5 led to the downregulation of β-catenin, thereby suppressing the epithelial-mesenchymal transition (EMT). Additionally, VAMP8 promoted autophagy flux, which may contribute to the suppression of osteosarcoma metastasis. In conclusion, our study anticipated that VAMP8 inhibits osteosarcoma metastasis by promoting the proteasomal degradation of DDX5, consequently inhibiting WNT/β-catenin signaling and EMT. Dysregulation of autophagy by VAMP8 is also implicated as a potential mechanism. These findings provide new insights into the biological nature driving osteosarcoma metastasis and highlight the modulation of VAMP8 as a potential therapeutic strategy for targeting osteosarcoma metastasis.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Ping Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Lelei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Xiangpeng Xie
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Yuanyi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Kaida Bo
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Jing Xue
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
- Clinical Pathology Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Faxue Liao
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Pengfei Xu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Yong Hu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruyu Yan
- Cancer Metabolism Laboratory, School of Life Sciences, Anhui Medical University, Hefei, China
| | - Dan Liu
- Cancer Metabolism Laboratory, School of Life Sciences, Anhui Medical University, Hefei, China
| | - Jun Chang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
| | - Kecheng Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Orthopaedics, Anhui Public Health Clinical Center, Hefei, China
- Cancer Metabolism Laboratory, School of Life Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Nguyen YTK, Ha HTT, Nguyen TH, Nguyen LN. The role of SLC transporters for brain health and disease. Cell Mol Life Sci 2021; 79:20. [PMID: 34971415 PMCID: PMC11071821 DOI: 10.1007/s00018-021-04074-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/05/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
The brain exchanges nutrients and small molecules with blood via the blood-brain barrier (BBB). Approximately 20% energy intake for the body is consumed by the brain. Glucose is known for its critical roles for energy production and provides substrates for biogenesis in neurons. The brain takes up glucose via glucose transporters GLUT1 and 3, which are expressed in several neural cell types. The brain is also equipped with various transport systems for acquiring amino acids, lactate, ketone bodies, lipids, and cofactors for neuronal functions. Unraveling the mechanisms by which the brain takes up and metabolizes these nutrients will be key in understanding the nutritional requirements in the brain. This could also offer opportunities for therapeutic interventions in several neurological disorders. For instance, emerging evidence suggests a critical role of lactate as an alternative energy source for neurons. Neuronal cells express monocarboxylic transporters to acquire lactate. As such, treatment of GLUT1-deficient patients with ketogenic diets to provide the brain with alternative sources of energy has been shown to improve the health of the patients. Many transporters are present in the brain, but only a small number has been characterized. In this review, we will discuss about the roles of solute carrier (SLC) transporters at the blood brain barrier (BBB) and neural cells, in transport of nutrients and metabolites in the brain.
Collapse
Affiliation(s)
- Yen T K Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Hoa T T Ha
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Tra H Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Long N Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore.
- SLING/Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- Immunology Translational and Cardiovascular Disease Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.
| |
Collapse
|
4
|
Li F, Fountzilas C, Puzanov I, Attwood KM, Morrison C, Ling X. Multiple functions of the DEAD-box RNA helicase, DDX5 (p68), make DDX5 a superior oncogenic biomarker and target for targeted cancer therapy. Am J Cancer Res 2021; 11:5190-5213. [PMID: 34765320 PMCID: PMC8569338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/22/2021] [Indexed: 06/13/2023] Open
Abstract
DDX5 (p68) is a well-known multifunctional DEAD-box RNA helicase and a transcription cofactor. Since its initial discovery more than three decades ago, DDX5 is gradually recognized as a potential biomarker and target for the treatment of various cancer types. Studies over the years significantly expanded our understanding of the functional diversity of DDX5 in various cancer types and extended our knowledge of its Mechanism of Action (MOA). This provides a rationale for the development of novel cancer therapeutics by using DDX5 as a biomarker and a therapeutic target. However, while most of the published studies have found DDX5 to be an oncogenic target and a cancer treatment-resistant biomarker, a few studies have reported that in certain scenarios, DDX5 may act as a tumor suppressor. After careful review of all the available relevant studies in the literature, we found that the multiple functions of DDX5 make it both a superior independent oncogenic biomarker and target for targeted cancer therapy. In this article, we will summarize the relevant studies on DDX5 in literature with a careful analysis and discussion of any inconsistencies encountered, and then provide our conclusions with respect to understanding the MOA of FL118, a novel small molecule. We hope that such a review will stimulate further discussion on this topic and assist in developing better strategies to treat cancer by using DDX5 as both an oncogenic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
- Department of Developmental Therapeutics Program, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
| | - Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
- Department of Alliance for Clinical Trials in Oncology Pancreatic Ductal Adenocarcinoma Working Group, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
- Department of Developmental Therapeutics Program, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
- Department of Developmental Therapeutics Program, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
| | - Kristopher M Attwood
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
- Department of Developmental Therapeutics Program, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
| | - Carl Morrison
- Department of Pathology & Laboratory Medicine, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
| | - Xiang Ling
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer CenterBuffalo, New York 14263, USA
- Canget BioTekpharma LLCBuffalo, New York 14203, USA
| |
Collapse
|
5
|
Xanthohumol Attenuates Lipopolysaccharide-Induced Depressive Like Behavior in Mice: Involvement of NF-κB/Nrf2 Signaling Pathways. Neurochem Res 2021; 46:3135-3148. [PMID: 34398408 DOI: 10.1007/s11064-021-03396-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 02/08/2023]
Abstract
Depression is the most common psychiatric disorder associated with brain and immune system abnormalities. In recent years, xanthohumol (Xn) a bioactive prenylated flavonoid has received ample attention for its polypharmacological effects, therefore, here we aimed to explore the protective effects of Xn against the LPS-induced depressive-like symptoms mediated by inflammation and oxidative stress. We tested the effect of Xn against LPS-induced behavioural changes in mice by means of forced swimming test (FST), tail suspention test (TST), sucrose preference test (SPT) and open field test (OPT). Examined the neuroinflammation and oxido-nitrosative stress (O&NS) markers and analyze Nrf2 and NF-κB signalling pathways in the hippocampus. Our results indicated that peripheral repeated administration of lipopolysaccharides (LPS) (1 mg/kg, intra peritoneally) induced depressive-like behavior, neuroinflammation and O&NS in mice. Pretreatment with Xn (10 and 20 mg/kg, intra gastrically) reverse the behavioural impairments prophylactically as obvious in the FST and TST without effecting locomotion, however only 20 mg dose improve anhedonic behavior as observed in SPT. Similarly, Xn pretreatment in dose-dependent manner prevented the LPS induced neuro-inflammation and O&NS. Immunofluorescence analysis showed that Xn reduced activated gliosis via attenuation of Iba-1 and GFAP in hippocampus. In addition, Xn considerably reduced the expression of phospho-NF-κB and cleaved caspase-3 while enhanced Nrf2 and HO-1 expression in the hippocampus. To the best of our knowledge, this is the first study to examine the underlying beneficial prophylactic effects of the Xn in neuroinflammation and O&NS mediating depressive-like behaviors.
Collapse
|
6
|
Gupta R, Srivastava D, Sahu M, Tiwari S, Ambasta RK, Kumar P. Artificial intelligence to deep learning: machine intelligence approach for drug discovery. Mol Divers 2021; 25:1315-1360. [PMID: 33844136 PMCID: PMC8040371 DOI: 10.1007/s11030-021-10217-3] [Citation(s) in RCA: 407] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Drug designing and development is an important area of research for pharmaceutical companies and chemical scientists. However, low efficacy, off-target delivery, time consumption, and high cost impose a hurdle and challenges that impact drug design and discovery. Further, complex and big data from genomics, proteomics, microarray data, and clinical trials also impose an obstacle in the drug discovery pipeline. Artificial intelligence and machine learning technology play a crucial role in drug discovery and development. In other words, artificial neural networks and deep learning algorithms have modernized the area. Machine learning and deep learning algorithms have been implemented in several drug discovery processes such as peptide synthesis, structure-based virtual screening, ligand-based virtual screening, toxicity prediction, drug monitoring and release, pharmacophore modeling, quantitative structure-activity relationship, drug repositioning, polypharmacology, and physiochemical activity. Evidence from the past strengthens the implementation of artificial intelligence and deep learning in this field. Moreover, novel data mining, curation, and management techniques provided critical support to recently developed modeling algorithms. In summary, artificial intelligence and deep learning advancements provide an excellent opportunity for rational drug design and discovery process, which will eventually impact mankind. The primary concern associated with drug design and development is time consumption and production cost. Further, inefficiency, inaccurate target delivery, and inappropriate dosage are other hurdles that inhibit the process of drug delivery and development. With advancements in technology, computer-aided drug design integrating artificial intelligence algorithms can eliminate the challenges and hurdles of traditional drug design and development. Artificial intelligence is referred to as superset comprising machine learning, whereas machine learning comprises supervised learning, unsupervised learning, and reinforcement learning. Further, deep learning, a subset of machine learning, has been extensively implemented in drug design and development. The artificial neural network, deep neural network, support vector machines, classification and regression, generative adversarial networks, symbolic learning, and meta-learning are examples of the algorithms applied to the drug design and discovery process. Artificial intelligence has been applied to different areas of drug design and development process, such as from peptide synthesis to molecule design, virtual screening to molecular docking, quantitative structure-activity relationship to drug repositioning, protein misfolding to protein-protein interactions, and molecular pathway identification to polypharmacology. Artificial intelligence principles have been applied to the classification of active and inactive, monitoring drug release, pre-clinical and clinical development, primary and secondary drug screening, biomarker development, pharmaceutical manufacturing, bioactivity identification and physiochemical properties, prediction of toxicity, and identification of mode of action.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Devesh Srivastava
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Swati Tiwari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
7
|
The RNA helicase Dhx15 mediates Wnt-induced antimicrobial protein expression in Paneth cells. Proc Natl Acad Sci U S A 2021; 118:2017432118. [PMID: 33483420 PMCID: PMC7848544 DOI: 10.1073/pnas.2017432118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
RNA helicases play roles in various essential biological processes such as RNA splicing and editing. Recent in vitro studies show that RNA helicases are involved in immune responses toward viruses, serving as viral RNA sensors or immune signaling adaptors. However, there is still a lack of in vivo data to support the tissue- or cell-specific function of RNA helicases owing to the lethality of mice with complete knockout of RNA helicases; further, there is a lack of evidence about the antibacterial role of helicases. Here, we investigated the in vivo role of Dhx15 in intestinal antibacterial responses by generating mice that were intestinal epithelial cell (IEC)-specific deficient for Dhx15 (Dhx15 f/f Villin1-cre, Dhx15ΔIEC). These mice are susceptible to infection with enteric bacteria Citrobacter rodentium (C. rod), owing to impaired α-defensin production by Paneth cells. Moreover, mice with Paneth cell-specific depletion of Dhx15 (Dhx15 f/f Defensinα6-cre, Dhx15ΔPaneth) are more susceptible to DSS (dextran sodium sulfate)-induced colitis, which phenocopy Dhx15ΔIEC mice, due to the dysbiosis of the intestinal microbiota. In humans, reduced protein levels of Dhx15 are found in ulcerative colitis (UC) patients. Taken together, our findings identify a key regulator of Wnt-induced α-defensins in Paneth cells and offer insights into its role in the antimicrobial response as well as intestinal inflammation.
Collapse
|
8
|
Zhu G, Yang S, Wang R, Lei J, Ji P, Wang J, Tao K, Yang C, Ge S, Wang L. P53/miR-154 Pathway Regulates the Epithelial-Mesenchymal Transition in Glioblastoma Multiforme Cells by Targeting TCF12. Neuropsychiatr Dis Treat 2021; 17:681-693. [PMID: 33664574 PMCID: PMC7924251 DOI: 10.2147/ndt.s273578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Glioblastoma multiforme (GBM) is an aggressive brain tumor with a rather short survival time. Mutation of p53 has been observed and reported to play critical roles in the progression of GBM. However, the pathological mechanisms are still unclear. This study was designed to identify the role of miR-154 in mediating the biological functions of p53 in glioblastoma multiforme. METHODS In the current study, the expression of miR-154 in GBM tissue samples and cell lines with wt-p53 or mutant p53 was evaluated. The functions of miR-154 in tumor migration, invasion and epithelial-mesenchymal transition were analyzed in vitro. A luciferase reporter assay was used to identify the target of miR-154. RESULTS We found that expression of miR-154 was much lower in patient tissues with mutant p53. Further study revealed that p53 was a transcription factor of miR-154 and that the R273H mutation led to its inactivation. In addition, overexpression of miR-154 remarkably suppressed cell migration, invasion and EMT in vitro and tumor growth in vivo. Moreover, TCF12 was proven to be a direct target of miR-154, and the tumor suppressive effect of miR-154 was reversed by TCF12. CONCLUSION Overall, miR-154, which was regulated by wt-p53, inhibited migration, invasion and EMT of GBM cells by targeting TCF12, indicating that miR-154 may act as a biomarker and that the p53/miR-154/TCF12 pathway could be a potential therapeutic target for GBM.
Collapse
Affiliation(s)
- Gang Zhu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shirong Yang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Ronglin Wang
- Department of Oncology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jie Lei
- Department of Neurosurgery, Wuhan General Hospital of PLA, Wuhan, Hubei, People's Republic of China
| | - Peigang Ji
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jiancai Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Kai Tao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Chen Yang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
9
|
Zhao X, Bao M, Zhang F, Wang W. Camptothecin induced DDX5 degradation increased the camptothecin resistance of osteosarcoma. Exp Cell Res 2020; 394:112148. [PMID: 32585151 DOI: 10.1016/j.yexcr.2020.112148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/03/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor in children and adolescents. Unfortunately, chemo-resistance is a huge obstacle in the treatment of OS. However, the underlying molecular mechanisms of OS chemo-resistance still remain unknown. Here we reported that the resistance to camptothecin (cpt) therapy was driven by degradation of DDX5. DDX5 knockdown decreased cell death and DNA damage and recovered cell proliferation in cpt treated 143B cells. Furthermore, we found that DDX5 bound to NONO, a kind of DNA repairing protein, and regulated NONO functions. Our data verified that cpt-induced degradation of DDX5 following by breaking down the protein bound of NONO, which participated in the resistance of cpt. In the summary, according to our results, DDX5 might be a potential therapeutic target for improving clinical outcomes of cpt in OS.
Collapse
Affiliation(s)
- Xingkai Zhao
- Department of Orthopedic Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Miao Bao
- The First Affiliated Hospital of Xi'an Medical University, China
| | - Fengmin Zhang
- Department of Microbiology, Harbin Medical University, Harbin, China.
| | - Wenbo Wang
- Department of Orthopedic Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
10
|
Qiu Y, Qu B, Zhen Z, Yuan X, Zhang L, Zhang M. Leucine Promotes Milk Synthesis in Bovine Mammary Epithelial Cells via the PI3K-DDX59 Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8884-8895. [PMID: 31345029 DOI: 10.1021/acs.jafc.9b03574] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Leucine is an essential amino acid in the milk production of bovine mammary glands, but the regulatory roles and molecular mechanisms of leucine are still not known well. This study investigated the roles of leucine on milk synthesis and explored the corresponding mechanism in bovine mammary epithelial cells (BMECs). Leucine (0, 0.25, 0.5, 0.75, 1.0, and 1.25 mM) was added to BMECs that were cultured in FBS-free OPTI-MEM medium. Leucine significantly promoted milk protein and milk fat synthesis and also increased phosphorylation of mTOR signaling protein and the protein expression levels of SREBP-1c, with the most significant effects at 0.75 mM concentration. Leucine increased the expression and nuclear localization of DDX59, and loss and gain of gene function experiments further reveal that DDX59 mediates the stimulation of leucine on the mRNA expression variation of mTOR and SREBP-1c genes. PI3K inhibition experiment further detected that leucine upregulated expression of DDX59 and its downstream signaling via PI3K activation. ChIP-qPCR analysis further proved the binding of DDX59 to the promoter regions of mTOR and SREBP-1c. In summary, these data prove that DDX59 positively regulates the mTOR and SREBP-1c signaling pathways leading to synthesis of milk, and leucine regulates these two signaling pathways through the PI3K-DDX59 signaling.
Collapse
Affiliation(s)
- Youwen Qiu
- The Key Laboratory of Dairy Science of Education Ministry , Northeast Agricultural University , Harbin 150030 , P. R. China
| | - Bo Qu
- The Key Laboratory of Dairy Science of Education Ministry , Northeast Agricultural University , Harbin 150030 , P. R. China
| | - Zhen Zhen
- The Key Laboratory of Dairy Science of Education Ministry , Northeast Agricultural University , Harbin 150030 , P. R. China
| | - Xiaohan Yuan
- The Key Laboratory of Dairy Science of Education Ministry , Northeast Agricultural University , Harbin 150030 , P. R. China
| | - Li Zhang
- The Key Laboratory of Dairy Science of Education Ministry , Northeast Agricultural University , Harbin 150030 , P. R. China
| | - Minghui Zhang
- The Key Laboratory of Dairy Science of Education Ministry , Northeast Agricultural University , Harbin 150030 , P. R. China
| |
Collapse
|
11
|
Chen X, Zhang C, Wang X. Long noncoding RNA DLEU1 aggravates osteosarcoma carcinogenesis via regulating the miR-671-5p/DDX5 axis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3322-3328. [PMID: 31379208 DOI: 10.1080/21691401.2019.1648285] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Xinxin Chen
- Department of Orthopedics, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Chengyong Zhang
- Department of Orthopedics, The Second People’s Hospital of Nanyang City, Nanyang, Henan, China
| | - Xiao Wang
- Department of Orthopedics, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| |
Collapse
|