6,345 results on '"c-jun"'
Search Results
2. c-JUN interacts with HDAC1 as a potential combinatorial therapeutic target in acute myeloid leukemia
- Author
-
Wu, Ke, Xu, Xiaoyu, Wei, Wei, Wen, Jie, and Hu, Haixi
- Published
- 2025
- Full Text
- View/download PDF
3. Celastrol directly targets LRP1 to inhibit fibroblast-macrophage crosstalk and ameliorates psoriasis progression
- Author
-
Zhu, Yuyu, Zhao, Lixin, Yan, Wei, Ma, Hongyue, Zhao, Wanjun, Qu, Jiao, Zheng, Wei, Zhang, Chenyang, Du, Haojie, Yu, Meng, Wan, Ning, Ye, Hui, Xie, Yicheng, Ke, Bowen, Xu, Qiang, Sun, Haiyan, Sun, Yang, and Ouyang, Zijun
- Published
- 2025
- Full Text
- View/download PDF
4. Identification of chemical inhibitors targeting long noncoding RNA through gene signature-based high throughput screening
- Author
-
An, Jun, Wang, Huili, Wei, Mingming, Yu, Xiankuo, Liao, Yile, Tan, Xue, Hu, Chao, Li, Shengrong, Luo, Yan, Gui, Yu, Lin, Kequan, Wang, Yumei, Huang, Lijun, and Wang, Dong
- Published
- 2025
- Full Text
- View/download PDF
5. A novel biomarker GATM suppresses proliferation and malignancy of cholangiocarcinoma cells by modulating the JNK/c-Jun signalling pathways
- Author
-
Yu, Yi, Gan, Wei, Xiong, Jianping, and Li, Junhe
- Published
- 2024
- Full Text
- View/download PDF
6. Indonesian Newcastle Disease Virus Field Isolate Reduces c-Jun Expression in Rat Mammary Cancer Models.
- Author
-
Sonita Marson, Fransiska Gratia, Sewoyo, Palagan Senopati, Mantik Astawa, I. Nyoman, Ayu Mirah Adi, Anak Agung, Kade Suardana, Ida Bagus, Oka Winaya, Ida Bagus, and Berata, I. Ketut
- Subjects
CANCER treatment ,NEWCASTLE disease virus ,VIROTHERAPY ,CELL death ,IMMUNOHISTOCHEMISTRY - Abstract
c-Jun is often found to be overexpressed in various cancers, so this gene might be a target for cancer therapy. Newcastle disease virus (NDV) is recognized for its oncolytic properties and potential as a cancer virotherapy agent, with various mechanisms reported to trigger cancer cell death. This study aimed to assess the c-Jun expression in rat mammary cancer models. Rat mammary cancer models were categorized into two treatment groups: the control group (C) and the virotherapy group (V). Group C was administered with 0.5 cc of sterile PBS, while group V received 7 log 2 HAU per 0.5 cc of the Indonesian NDV field isolate Tabanan-1/ARP/2017 intratumorally. The treatment was carried out for four days in a row. Two weeks after treatment, all rats were humanely euthanized, and mammary cancer tissues were excised for further examination. Mammary cancer tissues were examined histopathologically and analyzed using immunohistochemistry to determine intranuclear cJun expression, quantified by the H-Score. The results demonstrated that NDV significantly reduced c-Jun expression. It can be inferred that NDV Tabanan-1/ARP/2017 holds potential as a mammary cancer therapy agent by reducing c-Jun expression. This finding is considered novel, as there have been no previous reports of decreased c-Jun expression following virotherapy with NDV. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
7. Astrocytes-Secreted WNT5B Disrupts the Blood–Brain Barrier Via ROR1/JNK/c-JUN Cascade During Meningitic Escherichia Coli Infection.
- Author
-
Qu, Xinyi, Yang, Ruicheng, Tan, Chen, Chen, Huanchun, and Wang, Xiangru
- Abstract
The blood–brain barrier (BBB) is a complex structure that separates the central nervous system (CNS) from the peripheral blood circulation. Effective communication between different cell types within the BBB is crucial for its proper functioning and maintenance of homeostasis. In this study, we demonstrate that meningitic Escherichia coli (E. coli)-induced WNT5B plays a role in facilitating intercellular communication between astrocytes and brain microvascular endothelial cells (BMECs). We discovered that astrocytes-derived WNT5B activates the non-canonical WNT signaling pathway JNK/c-JUN in BMECs through its receptor ROR1, leading to inhibition of ZO-1 expression and impairment of the tight junction integrity in BMECs. Notably, our findings reveal that c-JUN, a transcription factor, directly regulates ZO-1 expression. By employing a dual luciferase reporting system and chromatin immunoprecipitation techniques, we identified specific binding sites of c-JUN on the ZO-1 promoter region. Overall, our study highlights the involvement of WNT5B in mediating intercellular communication between astrocytes and BMECs, provides insights into the role of WNT5B in meningitic E. coli-induced disruption of BBB integrity, and suggests potential therapeutic targeting of WNT5B as a strategy to address BBB dysfunction. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
8. Impact of c-JUN deficiency on thalamus development in mice and human neural models.
- Author
-
Shi, Jiantao, Chen, Qing, Lai, Jianheng, Zhu, Jieying, Zhang, Ran, Mazid, Md. Abdul, Li, Dongwei, Su, Huanxing, and Qin, Dajiang
- Subjects
- *
MEDICAL sciences , *TRANSCRIPTION factors , *EMBRYONIC stem cells , *MEDICAL genetics , *LIFE sciences , *DYSPLASIA - Abstract
Background: c-Jun is a key regulator of gene expression. Through the formation of homo- or heterodimers, c-JUN binds to DNA and regulates gene transcription. While c-Jun plays a crucial role in embryonic development, its impact on nervous system development in higher mammals, especially for some deep structures, for example, thalamus in diencephalon, remains unclear. Methods: To investigate the influence of c-JUN on early nervous system development, c-Jun knockout (KO) mice and c-JUN KO H1 embryonic stem cells (ESCs)-derived neural progenitor cells (NPCs), cerebral organoids (COs), and thalamus organoids (ThOs) models were used. We detected the dysplasia via histological examination and immunofluorescence staining, omics analysis, and loss/gain of function analysis. Results: At embryonic day 14.5, c-Jun knockout (KO) mice exhibited sparseness of fibers in the brain ventricular parenchyma and malformation of the thalamus in the diencephalon. The absence of c-JUN accelerated the induction of NPCs but impaired the extension of fibers in human neuronal cultures. COs lacking c-JUN displayed a robust PAX6+/NESTIN+ exterior layer but lacked a fibers-connected core. Moreover, the subcortex-like areas exhibited defective thalamus characteristics with transcription factor 7 like 2-positive cells. Notably, in guided ThOs, c-JUN KO led to inadequate thalamus patterning with sparse internal nerve fibers. Chromatin accessibility analysis confirmed a less accessible chromatin state in genes related to the thalamus. Overexpression of c-JUN rescued these defects. RNA-seq identified 18 significantly down-regulated genes including RSPO2, WNT8B, MXRA5, HSPG2 and PLAGL1 while 24 genes including MSX1, CYP1B1, LMX1B, NQO1 and COL2A1 were significantly up-regulated. Conclusion: Our findings from in vivo and in vitro experiments indicate that c-JUN depletion impedes the extension of nerve fibers and renders the thalamus susceptible to dysplasia during early mouse embryonic development and human ThO patterning. Our work provides evidence for the first time that c-JUN is a key transcription regulator that play important roles in the thalamus/diencephalon development. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Chemogenetic neuronal silencing decouples c‐Jun activation from cell death in the temporal cortex.
- Author
-
Wood, Caleb A., Somasundaram, Preethi, Dundee, Jacob M., Rudy, Melissa A., Watkins, Trent A., and Jankowsky, Joanna L.
- Subjects
- *
ALZHEIMER'S disease , *TEMPORAL lobe , *TRANSCRIPTION factors , *ENTORHINAL cortex , *NEURODEGENERATION , *ELECTRIC transients - Abstract
Initial symptoms of neurodegenerative diseases are often defined by the loss of the most vulnerable neural populations specific to each disorder. In the early stages of Alzheimer's disease, vulnerable circuits in the temporal lobe exhibit diminished activity prior to overt degeneration. It remains unclear whether these functional changes contribute to regional vulnerability or are simply a consequence of pathology. We previously found that entorhinal neurons in the temporal cortex undergo cell death following transient suppression of electrical activity, suggesting a causal role for activity disruption in neurodegeneration. Here we demonstrate that electrical arrest of this circuit stimulates the injury‐response transcription factor c‐Jun. Entorhinal silencing induces transcriptional changes consistent with c‐Jun activation that share characteristics of gene signatures in other neuronal populations vulnerable to Alzheimer's disease. Despite its established role in the neuronal injury response, inhibiting c‐Jun failed to ameliorate entorhinal degeneration following activity disruption. Finally, we present preliminary evidence of integrated stress response activity that may serve as an alternative hypothesis to what drives entorhinal degeneration after silencing. Our data demonstrate that c‐Jun is activated in response to neuronal silencing in the entorhinal cortex but is decoupled from subsequent neurodegeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Indonesian Newcastle Disease Virus Field Isolate Reduces c-Jun Expression in Rat Mammary Cancer Models
- Author
-
Fransiska Gratia Sonita Marson, Palagan Senopati Sewoyo, I Nyoman Mantik Astawa, Anak Agung Ayu Mirah Adi, Ida Bagus Kade Suardana, Ida Bagus Oka Winaya, and I Ketut Berata
- Subjects
c-jun ,mammary cancer ,newcastle disease virus ,virotherapy ,Veterinary medicine ,SF600-1100 - Abstract
c-Jun is often found to be overexpressed in various cancers, so this gene might be a target for cancer therapy. Newcastle disease virus (NDV) is recognized for its oncolytic properties and potential as a cancer virotherapy agent, with various mechanisms reported to trigger cancer cell death. This study aimed to assess the c-Jun expression in rat mammary cancer models. Rat mammary cancer models were categorized into two treatment groups: the control group (C) and the virotherapy group (V). Group C was administered with 0.5 cc of sterile PBS, while group V received 7 log 2 HAU per 0.5 cc of the Indonesian NDV field isolate Tabanan-1/ARP/2017 intratumorally. The treatment was carried out for four days in a row. Two weeks after treatment, all rats were humanely euthanized, and mammary cancer tissues were excised for further examination. Mammary cancer tissues were examined histopathologically and analyzed using immunohistochemistry to determine intranuclear c-Jun expression, quantified by the H-Score. The results demonstrated that NDV significantly reduced c-Jun expression. It can be inferred that NDV Tabanan-1/ARP/2017 holds potential as a mammary cancer therapy agent by reducing c-Jun expression. This finding is considered novel, as there have been no previous reports of decreased c-Jun expression following virotherapy with NDV.
- Published
- 2025
- Full Text
- View/download PDF
11. Role of the Egr2 Promoter Antisense RNA in Modulating the Schwann Cell Chromatin Landscape.
- Author
-
Martinez Moreno, Margot, Karambizi, David, Hwang, Hyeyeon, Fregoso, Kristen, Michles, Madison J., Fajardo, Eduardo, Fiser, Andras, and Tapinos, Nikos
- Subjects
PERIPHERAL nerve injuries ,SCHWANN cells ,ANTISENSE RNA ,PERIPHERAL nervous system ,NERVOUS system regeneration ,TRANSCRIPTION factors - Abstract
Background: Schwann cells (SCs) and their plasticity contribute to the peripheral nervous system's capacity for nerve regeneration after injury. The Egr2/Krox20 promoter antisense RNA (Egr2-AS) recruits chromatin remodeling complexes to inhibit Egr2 transcription following peripheral nerve injury. Methods: RNA-seq and ATAC-seq were performed on control cells, Lenti-GFP-transduced cells, and cells overexpressing Egr2-AS (Lenti-AS). Egr2 AS-RNA was cloned into the pLVX-DsRed-Express2-N1 lentiviral expression vector (Clontech, Mountain View, CA, USA), and the levels of AS-RNA expression were determined. Ezh2 and Wdr5 were immunoprecipitated from rat SCs and RT-qPCR was performed against AS-Egr2 RNA. ChIP followed by DNA purification columns was used to perform qPCR for relevant promoters. Hi-C, HiC-DC+, R, Bioconductor, and TOBIAS were used for significant and differential loop analysis, identifications of COREs and CORE-promotor loops, comparisons of TF activity at promoter sites, and identification of site-specific TF footprints. OnTAD was used to detect TADs, and Juicer was used to identify A/B compartments. Results: Here we show that a Neuregulin-ErbB2/3 signaling axis mediates binding of the Egr2-AS to YY1
Ser184 and regulates its expression. Egr2-AS modulates the chromatin accessibility of Schwann cells and interacts with two distinct histone modification complexes. It binds to EZH2 and WDR5 and enables targeting of H3K27me3 and H3K4me3 to promoters of Egr2 and C-JUN, respectively. Expression of the Egr2-AS results in reorganization of the global chromatin landscape and quantitative changes in the loop formation and contact frequency at domain boundaries exhibiting enrichment for AP-1 genes. In addition, the Egr2-AS induces changes in the hierarchical TADs and increases transcription factor binding scores on an inter-TAD loop between a super-enhancer regulatory hub and the promoter of mTOR. Conclusions: Our results show that Neuregulin-ErbB2/3-YY1 regulates the expression of Egr2-AS, which mediates remodeling of the chromatin landscape in Schwann cells. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
12. An evidence update to explore molecular targets and protective mechanisms of apigenin against abdominal aortic aneurysms based on network pharmacology and experimental validation.
- Author
-
Li, Dongyu, Wang, Lei, Jiang, Bo, Miao, Yuxi, and Li, Xuan
- Abstract
Abdominal aortic aneurysms (AAA) is a life-threatening disease and the incidence of AAA is still on the rise in recent years. Numerous studies suggest that dietary moderate consumption of polyphenol exerts beneficial effects on cardiovascular disease. Apigenin (API) is a promising dietary polyphenol and possesses potent beneficial effects on our body. Although our previous study revealed protective effects of API on experimental AAA formation, up till now few studies were carried out to further investigate its involved molecular mechanisms. In the present study, network pharmacology combined molecular docking and experimental validation was used to explore API-related therapeutic targets and mechanisms in the treatment of AAA. Firstly, we collected 202 API-related therapeutic targets and 2475 AAA-related pathogenetic targets. After removing duplicates, a total of 68 potential therapeutic targets were obtained. Moreover, 5 targets with high degree including TNF, ACTB, INS, JUN, and MMP9 were identified as core targets of API for treating AAA. In addition, functional enrichment analysis indicated that API exerted pharmacological effects in AAA by affecting versatile mechanisms, including apoptosis, inflammation, blood fluid dynamics, and immune modulation. Molecular docking results further supported that API had strong affinity with the above core targets. Furthermore, protein level of core targets and related pathways were evaluated in a Cacl2-induced AAA model by using western blot and immunohistochemistry. The experimental validation results demonstrated that API significantly attenuated phosphorylation of JUN and protein level of predicted core targets. Taken together, based on network pharmacological and experimental validation, our study systematically explored associated core targets and potential therapeutic pathways of API for AAA treatment, which could supply valuable insights and theoretical basis for AAA treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Saikosaponin A Recovers Impaired Filaggrin Levels in Inflamed Skin by Downregulating the Expression of FRA1 and c-Jun.
- Author
-
Ahn, Sung Shin, Yeo, Hyunjin, Jung, Euitaek, Kim, Tae Yoon, Han, Junekyu, Lee, Young Han, and Shin, Soon Young
- Subjects
- *
MITOGEN-activated protein kinases , *TOPICAL drug administration , *CYTOSKELETAL proteins , *GENETIC transcription , *ATOPIC dermatitis - Abstract
Filaggrin (FLG) is an essential structural protein expressed in differentiated keratinocytes. Insufficient FLG expression contributes to the pathogenesis of chronic inflammatory skin diseases. Saikosaponin A (SSA), a bioactive oleanane-type triterpenoid, exerts anti-inflammatory activity. However, the effects of topically applied SSA on FLG expression in inflamed skin remain unclear. This study aimed to evaluate the biological activity of SSA in restoring reduced FLG expression. The effect of SSA on FLG expression in HaCaT cells was assessed through various biological methods, including reverse transcription PCR, quantitative real-time PCR, immunoblotting, and immunofluorescence staining. TNFα and IFNγ decreased FLG mRNA, cytoplasmic FLG protein levels, and FLG gene promoter–reporter activity compared to the control groups. However, the presence of SSA restored these effects. A series of FLG promoter–reporter constructs were generated to investigate the underlying mechanism of the effect of SSA on FLG expression. Mutation of the AP1-binding site (mtAP1) in the −343/+25 FLG promoter–reporter abrogated the decrease in reporter activities caused by TNFα + IFNγ, suggesting the importance of the AP1-binding site in reducing FLG expression. The SSA treatment restored FLG expression by inhibiting the expression and nuclear localization of FRA1 and c-Jun, components of AP1, triggered by TNFα + IFNγ stimulation. The ERK1/2 mitogen-activated protein kinase signaling pathway upregulates FRA1 and c-Jun expression, thereby reducing FLG levels. The SSA treatment inhibited ERK1/2 activation caused by TNFα + IFNγ stimulation and reduced the levels of FRA1 and c-Jun proteins in the nucleus, leading to a decrease in the binding of FRA1, c-Jun, p-STAT1, and HDAC1 to the AP1-binding site in the FLG promoter. The effect of SSA was evaluated in an animal study using a BALB/c mouse model, which induces human atopic-dermatitis-like skin lesions via the topical application of dinitrochlorobenzene. Topically applied SSA significantly reduced skin thickening, immune cell infiltration, and the expression of FRA1, c-Jun, and p-ERK1/2 compared to the vehicle-treated group. These results suggest that SSA can effectively recover impaired FLG levels in inflamed skin by preventing the formation of the repressor complex consisting of FRA1, c-Jun, HDAC1, and STAT1. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Exploring the globoid cell leukodystrophy protein network and therapeutic interventions
- Author
-
Pukar Khanal, Vishal S. Patil, Kunal Bhattacharya, Amit Kumar Shrivastava, and Vishwambhar V. Bhandare
- Subjects
c-Jun ,Computational pharmacology ,Globoid cell leukodystrophy ,Krabbe disease ,Rare disease ,Medicine ,Science - Abstract
Abstract Globoid cell leukodystrophy is a severe rare disorder characterized by white matter degradation, resulting in a progressive loss of physical and mental abilities and has extremely limited therapeutic interventions. Therefore, this study aimed to delve into the Globoid cell leukodystrophy associated intricate network of differentially expressed genes (p 1) to identify potential druggable targets and possible therapeutic interventions using small molecules. The disease-associated neuronal protein circuit was constructed and analyzed, identifying 53 nodes (minimum edge cutoff 1), among which five (FOS, FOSB, GDNF, GFRA1, and JUN) were discerned as potential core protein nodes. Although our research enumerates the potential small molecules to target various protein nodes in the proposed disease network, we particularly underscore T-5224 to inhibit c-Jun activity as JUN was identified as one of the pivotal elements within the disease-associated neuronal protein circuit. The evaluation of T-5224 binding energy (− 11.0 kcal/mol) from docking study revealed that the compound to exhibit a notable affinity towards Jun/CRE complex. Moreover, the structural integrity of complex was affirmed through comprehensive molecular dynamics simulations, indicating a stable hydrophilic interaction between T-5224 and the Jun/CRE complex, thereby enhancing protein compactness and reducing solvent accessibility. This binding energy was further substantiated by free binding analysis, revealing a substantial thermodynamics complex state (− 448.00 ± 41.73 kJ/mol). Given that this investigation is confined to a computational framework, we additionally propose a hypothetical framework to ascertain the feasibility of inhibiting the Jun/CRE complex with T-5224 against Globoid cell leukodystrophy, employing a combination of in vitro and in vivo methodologies as a prospective avenue of this study.
- Published
- 2024
- Full Text
- View/download PDF
15. TFEB/3 Govern Repair Schwann Cell Generation and Function Following Peripheral Nerve Injury.
- Author
-
Patel, Akash A., Kim, Hyukmin, Ramesh, Raghu, Marquez, Anthony, Faraj, Moler M., Antikainen, Henri, Lee, Andrew S., Torres, Adriana, Khawaja, Imran M., Heffernan, Corey, Bonder, Edward M., Maurel, Patrice, Svaren, John, Son, Young-Jin, Dobrowolski, Radek, and Kim, Haesun A.
- Subjects
- *
SCHWANN cells , *SCIATIC nerve injuries , *PERIPHERAL nerve injuries , *KNOCKOUT mice , *GENETIC regulation , *NERVOUS system regeneration - Abstract
TFEB and TFE3 (TFEB/3), key regulators of lysosomal biogenesis and autophagy, play diverse roles depending on cell type. This study highlights a hitherto unrecognized role of TFEB/3 crucial for peripheral nerve repair. Specifically, they promote the generation of progenitor-like repair Schwann cells after axonal injury. In Schwann cell-specific TFEB/3 double knock-out mice of either sex, the TFEB/3 loss disrupts the transcriptomic reprogramming that is essential for the formation of repair Schwann cells. Consequently, mutant mice fail to populate the injured nerve with repair Schwann cells and exhibit defects in axon regrowth, target reinnervation, and functional recovery. TFEB/3 deficiency inhibits the expression of injury-responsive repair Schwann cell genes, despite the continued expression of c-jun, a previously identified regulator of repair Schwann cell function. TFEB/3 binding motifs are enriched in the enhancer regions of injury-responsive genes, suggesting their role in repair gene activation. Autophagy-dependent myelin breakdown is not impaired despite TFEB/3 deficiency. These findings underscore a unique role of TFEB/3 in adult Schwann cells that is required for proper peripheral nerve regeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. EWS-FLI1 and Activator Protein-1 (AP-1) Reciprocally Regulate Extracellular-Matrix Proteins in Ewing sarcoma Cells.
- Author
-
Croushore, Emma E., Stipp, Christopher S., and Gordon, David J.
- Subjects
- *
TRANSCRIPTION factors , *EWING'S sarcoma , *RIBONUCLEOSIDE diphosphate reductase , *GENE expression , *FIBRONECTINS - Abstract
Ribonucleotide reductase (RNR) is the rate-limiting enzyme in the synthesis of deoxyribonucleotides and the target of multiple chemotherapy drugs, including gemcitabine. We previously identified that inhibition of RNR in Ewing sarcoma tumors upregulates the expression levels of multiple members of the activator protein-1 (AP-1) transcription factor family, including c-Jun and c-Fos, and downregulates the expression of c-Myc. However, the broader functions and downstream targets of AP-1, which are highly context- and cell-dependent, are unknown in Ewing sarcoma tumors. Consequently, in this work, we used genetically defined models, transcriptome profiling, and gene-set -enrichment analysis to identify that AP-1 and EWS-FLI1, the driver oncogene in most Ewing sarcoma tumors, reciprocally regulate the expression of multiple extracellular-matrix proteins, including fibronectins, integrins, and collagens. AP-1 expression in Ewing sarcoma cells also drives, concurrent with these perturbations in gene and protein expression, changes in cell morphology and phenotype. We also identified that EWS-FLI1 dysregulates the expression of multiple AP-1 proteins, aligning with previous reports demonstrating genetic and physical interactions between EWS-FLI1 and AP-1. Overall, these results provide novel insights into the distinct, EWS-FLI1-dependent features of Ewing sarcoma tumors and identify a novel, reciprocal regulation of extracellular-matrix components by EWS-FLI1 and AP-1. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. CRKL Enhances YAP Signaling through Binding and JNK/JUN Pathway Activation in Liver Cancer.
- Author
-
Wesener, Marie C., Weiler, Sofia M. E., Bissinger, Michaela, Klessinger, Tobias F., Rose, Fabian, Merker, Sabine, Luzarowski, Marcin, Ruppert, Thomas, Helm, Barbara, Klingmüller, Ursula, Schirmacher, Peter, and Breuhahn, Kai
- Subjects
- *
HIPPO signaling pathway , *YAP signaling proteins , *ADAPTOR proteins , *GENETIC transcription , *LIVER cancer - Abstract
The Hippo pathway transducers yes-associated protein (YAP) and WW-domain containing transcription regulator 1 (WWTR1/TAZ) are key regulators of liver tumorigenesis, promoting tumor formation and progression. Although the first inhibitors are in clinical trials, targeting the relevant upstream regulators of YAP/TAZ activity could prove equally beneficial. To identify regulators of YAP/TAZ activity in hepatocarcinoma (HCC) cells, we carried out a proximity labelling approach (BioID) coupled with mass spectrometry. We verified CRK-like proto-oncogene adaptor protein (CRKL) as a new YAP-exclusive interaction partner. CRKL is highly expressed in HCC patients, and its expression is associated with YAP activity as well as poor survival prognosis. In vitro experiments demonstrated CRKL-dependent cell survival and the loss of YAP binding induced through actin disruption. Moreover, we delineated the activation of the JNK/JUN pathway by CRKL, which promoted YAP transcription. Our data illustrate that CRKL not only promoted YAP activity through its binding but also through the induction of YAP transcription by JNK/JUN activation. This emphasizes the potential use of targeting the JNK/JUN pathway to suppress YAP expression in HCC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Exploring the globoid cell leukodystrophy protein network and therapeutic interventions.
- Author
-
Khanal, Pukar, Patil, Vishal S., Bhattacharya, Kunal, Shrivastava, Amit Kumar, and Bhandare, Vishwambhar V.
- Subjects
SMALL molecules ,MOLECULAR dynamics ,HYDROPHILIC interactions ,BINDING energy ,WHITE matter (Nerve tissue) - Abstract
Globoid cell leukodystrophy is a severe rare disorder characterized by white matter degradation, resulting in a progressive loss of physical and mental abilities and has extremely limited therapeutic interventions. Therefore, this study aimed to delve into the Globoid cell leukodystrophy associated intricate network of differentially expressed genes (p < 0.05, |Fc|> 1) to identify potential druggable targets and possible therapeutic interventions using small molecules. The disease-associated neuronal protein circuit was constructed and analyzed, identifying 53 nodes (minimum edge cutoff 1), among which five (FOS, FOSB, GDNF, GFRA1, and JUN) were discerned as potential core protein nodes. Although our research enumerates the potential small molecules to target various protein nodes in the proposed disease network, we particularly underscore T-5224 to inhibit c-Jun activity as JUN was identified as one of the pivotal elements within the disease-associated neuronal protein circuit. The evaluation of T-5224 binding energy (− 11.0 kcal/mol) from docking study revealed that the compound to exhibit a notable affinity towards Jun/CRE complex. Moreover, the structural integrity of complex was affirmed through comprehensive molecular dynamics simulations, indicating a stable hydrophilic interaction between T-5224 and the Jun/CRE complex, thereby enhancing protein compactness and reducing solvent accessibility. This binding energy was further substantiated by free binding analysis, revealing a substantial thermodynamics complex state (− 448.00 ± 41.73 kJ/mol). Given that this investigation is confined to a computational framework, we additionally propose a hypothetical framework to ascertain the feasibility of inhibiting the Jun/CRE complex with T-5224 against Globoid cell leukodystrophy, employing a combination of in vitro and in vivo methodologies as a prospective avenue of this study. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Brachial Plexus Root Avulsion Injury-Induced Endothelin-Converting Enzyme-Like 1 Overexpression Is Associated with Injured Motor Neurons Survival.
- Author
-
Huang, Yu, Mai, Yunlin, Ye, Weijian, Lv, Shiqin, Zhou, Yingying, Wu, Pingzhen, Zhou, Lihua, Li, Yingqin, and Zhong, Ke
- Abstract
Brachial plexus root avulsion (BPRA) injury arises from challenging delivery during childbirth, sports-related incidents, or car accidents, leading to extensive loss of motor neurons (MNs) and subsequent paralysis, including both motor and sensory impairment. Surgical nerve re-implantation cannot effectively restore motor function, and the survival of injured MNs is vital for axon regeneration and re-innervating the target muscles. Therefore, identifying novel molecular targets to improve injured MNs survival is of great significance in the treatment of BPRA injuries. Endothelin-converting enzyme-like 1 (ECEL1), a membrane-bound metallopeptidase, was initially identified as a molecule associated with nerve injuries. Damaged neurons exhibit a significant increase in the expression of ECEL1 following various types of nerve injuries, such as optic nerve injury and sciatic nerve injury. This study aimed to investigate the relationship between ECEL1 overexpression and the survival of injured MNs following BPRA injury. Our results observed a significant elevation in ECEL1 expression in injured MNs and positively correlated with MNs survival following BPRA injury. The transcription of ECEL1 is regulated by the transcription factors c-Jun and ATF3 in the context of BPRA injury, which is consistent with previous other nerve injuries study. In addition, the expression of TrkA gradually decreases in ECEL1-positive MNs and ECEL1 possibly preserves the activity of downstream AKT-GSK3β pathway of TrkA in injured MNs. In conclusion, our results introduce a promising therapeutic molecular target to assist re-implantation surgery for the treatment of BPRA injury. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. The upregulation of Annexin A2 by TLR4 pathway facilitates lipid accumulation and liver injury via blocking AMPK/mTOR-mediated autophagy flux during the development of non-alcoholic fatty liver disease.
- Author
-
Wu, Haifeng, Zhou, Meng, Jin, Qin, Wang, Xun, Xu, Yue, Li, Ming, Chen, Shuhui, Tang, Qin, Wang, Qi, Hu, Baoying, Wu, Hongpei, Xiao, Mingbing, Qu, Lishuai, Zhang, Qiong, and Liu, Jinxia
- Abstract
Background and aims: Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease worldwide. In this study, we aimed to investigate the role and regulatory mechanism of Annexin A2 (ANXA2) in the pathogenesis of NAFLD. Methods: Histological analyses and ELISA were used to illuminate the expression of ANXA2 in NAFLD and healthy subjects. The role of ANXA2 was evaluated using high-fat diet (HFD)-fed mice via vein injection of adeno-associated viruses (AAV) knocking down ANXA2 or non-targeting control (NC) shRNAs. Moreover, HepG2 and LO2 cells were employed as in vitro hepatocyte models to investigate the expression and function of ANXA2. Results: ANXA2 was confirmed to be one of three hub genes in liver injury, and its expression was positively correlated with NAFLD activity score (NAS) and macrophage infiltration in NAFLD. Moreover, ANXA2 was significantly upregulated in NAFLD patients and HFD-fed mice. LPS/TLR4 pathway strongly upregulated ANXA2 expression, which is mediated by direct ANXA2 promoter binding by TLR4 downstream NF-κB p65 and c-Jun transcription factors. Increased ANXA2 expression was correlated with decreased autophagy flux and autophagy was activated by the depletion of ANXA2 in the models of NAFLD. Furthermore, ANXA2 interference led to the activation of AMPK/mTOR signaling axis, which may play a causal role in autophagy flux and the amelioration of steatosis. Conclusions: ANXA2 is a pathological predictor and promising therapeutic target for NAFLD. ANXA2 plays a crucial role in linking inflammation to hepatic metabolic disorder and injury, mainly through the blockage of AMPK/mTOR-mediated lipophagy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. Signal Transduction of Transient Receptor Potential TRPM8 Channels: Role of PIP5K, Gq-Proteins, and c-Jun.
- Author
-
Thiel, Gerald and Rössler, Oliver G.
- Subjects
- *
TRP channels , *AP-1 transcription factor , *CELLULAR signal transduction , *TRANSCRIPTION factors , *LEUCINE zippers , *GENETIC regulation - Abstract
Transient receptor potential melastatin-8 (TRPM8) is a cation channel that is activated by cold and "cooling agents" such as menthol and icilin, which induce a cold sensation. The stimulation of TRPM8 activates an intracellular signaling cascade that ultimately leads to a change in the gene expression pattern of the cells. Here, we investigate the TRPM8-induced signaling pathway that links TRPM8 channel activation to gene transcription. Using a pharmacological approach, we show that the inhibition of phosphatidylinositol 4-phosphate 5 kinase α (PIP5K), an enzyme essential for the biosynthesis of phosphatidylinositol 4,5-bisphosphate, attenuates TRPM8-induced gene transcription. Analyzing the link between TRPM8 and Gq proteins, we show that the pharmacological inhibition of the βγ subunits impairs TRPM8 signaling. In addition, genetic studies show that TRPM8 requires an activated Gα subunit for signaling. In the nucleus, the TRPM8-induced signaling cascade triggers the activation of the transcription factor AP-1, a complex consisting of a dimer of basic region leucine zipper (bZIP) transcription factors. Here, we identify the bZIP protein c-Jun as an essential component of AP-1 within the TRPM8-induced signaling cascade. In summary, with PIP5K, Gq subunits, and c-Jun, we identified key molecules in TRPM8-induced signaling from the plasma membrane to the nucleus. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. N-Acetylgalactosamine-4-sulfatase (Arylsulfatase B) Regulates PD-L1 Expression in Melanoma by an HDAC3-Mediated Epigenetic Mechanism.
- Author
-
Bhattacharyya, Sumit, O-Sullivan, InSug, and Tobacman, Joanne K.
- Subjects
- *
PROGRAMMED death-ligand 1 , *ARYLSULFATASES , *IMMUNE checkpoint proteins , *MELANOMA , *EPIGENETICS , *MICROPHTHALMIA-associated transcription factor , *BRAF genes - Abstract
The effects of the enzyme N-acetylgalactosamine-4-sulfatase (Arylsulfatase B, ARSB), which removes the 4-sulfate group at the non-reducing end of chondroitin 4-sulfate, on the expression of PD-L1 were determined, and the underlying mechanism of PD-L1 expression was elucidated. Initial experiments in human melanoma cells (A375) showed that PD-L1 expression increased from 357 ± 31 to 796 ± 50 pg/mg protein (p < 10−11) when ARSB was silenced in A375 cells. In subcutaneous B16F10 murine melanomas, PD-L1 declined from 1227 ± 189 to 583 ± 110 pg/mg protein (p = 1.67 × 10−7), a decline of 52%, following treatment with exogenous, bioactive recombinant ARSB. This decline occurred in association with reduced tumor growth and prolongation of survival, as previously reported. The mechanism of regulation of PD-L1 expression by ARSB is attributed to ARSB-mediated alteration in chondroitin 4-sulfation, leading to changes in free galectin-3, c-Jun nuclear localization, HDAC3 expression, and effects of acetyl-H3 on the PD-L1 promoter. These findings indicate that changes in ARSB contribute to the expression of PD-L1 in melanoma and can thereby affect the immune checkpoint response. Exogenous ARSB acted on melanoma cells and normal melanocytes through the IGF2 receptor. The decline in PD-L1 expression by exogenous ARSB may contribute to the impact of ARSB on melanoma progression. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. SIAH2 suppresses c‐JUN pathway by promoting the polyubiquitination and degradation of HBx in hepatocellular carcinoma.
- Author
-
Hu, Qinghe, Liu, Zhiyi, Liu, Yao, Qiu, Jie, Zhang, Xue, Sun, Jun, Zhang, Bin, and Shi, Hengliang
- Subjects
HEPATOCELLULAR carcinoma ,UBIQUITIN ligases ,HEPATITIS B virus - Abstract
As an important protein encoded by hepatitis B virus (HBV), HBV X protein (HBx) plays an important role in the development of hepatocellular carcinoma (HCC). It has been shown that seven in absentia homologue 1 (SIAH1) could regulates the degradation of HBx through the ubiquitin‐proteasome pathway. However, as a member of SIAH family, the regulatory effects of SIAH2 on HBx remain unclear. In this study, we first confirmed that SIAH2 could reduce the protein levels of HBx depending on its E3 ligase activity. Moreover, SIAH2 interacted with HBx and induced its K48‐linked polyubiquitination and proteasomal degradation. Furthermore, we provided evidence that SIAH2 inhibits HBx‐associated HCC cells proliferation by regulating HBx. In conclusion, our study identified a novel role for SIAH2 in promoting HBx degradation and SIAH2 exerts an inhibitory effect in the proliferation of HBx‐associated HCC through inducing the degradation of HBx. Our study provides a new idea for the targeted degradation of HBx and may have great huge significance into providing novel evidence for the targeted therapy of HBV‐infected HCC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Nur77 Mediates Anaphylaxis by Regulating miR-21a
- Author
-
Hyein Jo, Jaewhoon Jeoung, Kyeonghee Shim, and Dooil Jeoung
- Subjects
allergic inflammation ,C-JUN ,miR-21a ,Nur77 ,Biology (General) ,QH301-705.5 - Abstract
Nur77 belongs to the NR4A subfamily of orphan nuclear hormone receptors. It has been shown to play important roles in metabolism, cancer progression, cellular differentiation, and the regulation of immune process. However, there has yet to be research reporting on the role of Nur77 in allergic inflammations such as anaphylaxis. This study aimed to identify molecules that could mediate allergic inflammations. To this end, we performed RNA sequencing analysis employing bone marrow-derived mast cells (BMMCs). Antigen (DNP-HSA) stimulation increased the expression levels of transcription factors such as Nr4a3 (NOR1), Nr4a1 (Nur77), and Nr4a2 (Nurr1). We focused our study on Nur77. Antigen stimulation increased the expression of Nur77 in a time- and dose-dependent manner in rat basophilic leukemia cells (RBL2H3). The downregulation of Nur77 prevented both antigen-induced increase in β-hexosaminidase activity as well as hallmarks of allergic reactions such as HDAC3, COX2, and MCP1 in RBL2H3 cells. Nur77 was necessary for both passive cutaneous anaphylaxis (PCA) and passive systemic anaphylaxis (PSA). TargetScan analysis predicted that miR-21a would be a negative regulator of Nur77. miR-21a mimic negatively regulated PCA and PSA by inhibiting the hallmarks of allergic reactions. ChIP assays showed that c-JUN could bind to the promoter sequences of Nur77. Antigen stimulation increased the expression of c-JUN in RBL2H3 cells. Altogether, our findings demonstrate the regulatory role played by Nur77-miR-21a loop in allergic inflammations such as anaphylaxis, making this the first report to present the role played by Nur77 in an allergic inflammation. Our results suggest that Nur77 and miR-21 might serve as targets for developing anti-allergy drugs.
- Published
- 2024
- Full Text
- View/download PDF
25. USP5 facilitates bladder cancer progression by stabilizing the c-Jun protein
- Author
-
Zhang, Hui-hui, Zhang, An-qi, Peng, Peng, Huang, Liang, Liu, Cai-ying, Nie, Xin-rui, Hou, De-fu, Zhang, Xia, and Li, Shang-ze
- Published
- 2024
- Full Text
- View/download PDF
26. RETRACTED: Inhibition of HDAC4 Attenuated JNK/c-Jun-Dependent Neuronal Apoptosis and Early Brain Injury Following Subarachnoid Hemorrhage by Transcriptionally Suppressing MKK7.
- Subjects
SUBARACHNOID hemorrhage ,BRAIN injuries ,APOPTOSIS ,SMALL interfering RNA ,PEROXISOME proliferator-activated receptors ,C-Jun N-terminal kinases - Abstract
This article explores the molecular mechanisms underlying early brain injury (EBI) following subarachnoid hemorrhage (SAH). The researchers found that inhibiting histone deacetylase 4 (HDAC4) decreased neuronal apoptosis and brain injury by suppressing the transcription of MKK7, an upstream kinase of the JNK/c-Jun pathway. The study suggests that targeting HDAC4 could be a potential strategy for preventing SAH-induced EBI. The article provides insights into the role of HDAC4 in regulating neuronal apoptosis and the JNK/c-Jun signaling pathway, offering potential therapeutic implications for neurodegenerative diseases and acute brain injuries. [Extracted from the article]
- Published
- 2024
- Full Text
- View/download PDF
27. C-Jun Transcription Factor Oncogenic Activation in Oral Carcinoma.
- Author
-
MASTRONIKOLIS, Nicholas, CHRYSOVERGIS, Aristeidis, PAPANIKOLAOU, Vasileios, DERKA, Spyridoula, ASIMAKOPOULOS, Asimakis D., MASTRONIKOLI, Sofianiki, TSIAMBAS, Evangelos, MANAIOS, Loukas, PAPOULIAKOS, Sotirios, RAGOS, Vasileios, FOTIADES, Panagiotis, PANTOS, Pavlos, STATHOPOULOS, Panagiotis, and KYRODIMOS, Efthymios
- Subjects
- *
TRANSCRIPTION factors , *MOLECULAR biology , *BIOLOGICAL evolution , *HUMAN papillomavirus , *MOLECULAR evolution , *TUMOR suppressor genes - Abstract
Introduction: Oral carcinogenetic is based on a variety of genomic imbalances (gross chromosome or specific gene alterations) that drive the normal oral mucosa to its neoplastic/dysplastic epithelial form and finally to a totally malignant tissue transformation. In this multi-step procedure, down-regulation of suppressor genes combined with overactivation of oncogenes are two crucial and partially early genetic events involved in the onset and progression of neoplastic/malignant epithelia transformation. More specifically, deregulation of strong transcription factors negatively affects the normal expression of a broad spectrum of genes that are involved in cell proliferation and signalling transduction to the nucleus. Objective: The purpose of the current molecular review was to explore the c-Jun (chromosome location: 1p32-p31) transcription factor transformation mechanisms to oncogene in oral squamous cell carcinoma (OSCC). Material and method: A systematic review of the literature was carried out by searching in PubMed international database. The year 2010 was set as a prominent time limit for the publication date of the articles in the majority of them, whereas specific references of great importance and historical value in the field of the c-Jun gene discovery and analysis were also included. The following keywords were used: c-Jun, oncogene, signaling pathway, oral, carcinoma, transcription. A pool of 45 important articles were selected for the present study at the basis of combining molecular knowledge with new targeted therapeutic strategies. Results: C-Jun -- as a part of the c-Jun/c-Fos transcription factors' complex --critically regulates the expression levels in a variety of genes inside the cellular microenvironment. A broad spectrum of malignancies, including OSCC, demonstrate c-Jun alterations driving the gene to its oncogenic phenotype. Interestingly, c-Jun oncogenic activation is mediated by high-risk human papilloma virus (HR-HPV) persistent infection in significant subsets of these malignancies. Conclusions: C-Jun was the first oncogene -- acting as a strong transcription factor -- that was discovered and cloned 35 years ago. C-Jun is the living history of oncogenes and its discovery marks a significant step in the evolution of molecular biology. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Ethyl pyruvate attenuates cellular adhesion and proliferation of diffuse large B-cell lymphoma by targeting c-Jun.
- Author
-
Yan, Zhimin, Zhong, Qiong, Yan, Ling, and Xi Xu, Wenhong Lai
- Subjects
- *
CELL cycle , *CELL death , *DIFFUSE large B-cell lymphomas , *MESENCHYMAL stem cells , *CELL proliferation , *NON-Hodgkin's lymphoma , *PYRUVATES , *B cell lymphoma - Abstract
Diffuse large B-cell lymphoma (DLBCL) stands out as the most common type of malignant cancer, representing the majority of cases of non-Hodgkin's lymphoma. Ethyl pyruvate (EP) is a derivative of pyruvic acid and found to have potent anti-tumor properties. Despite its potential benefits, the impact of EP on DLBCL remains ambiguous. Our objective is to elucidate the role of EP in modulating the development of DLBCL. Analysis of cholecystokinin-8 (CCK-8) revealed that treatment with EP significantly diminished the viability of DLBCL cells. Furthermore, EP administration suppressed colony formation and hindered cell adhesion and invasion in DLBCL cells. Examination of cell cycle progression showed that EP treatment induced arrest at the G1 phase and subsequently reduced the S phase population in DLBCL cells. EP treatment consistently exhibited apoptosis-inducing properties in Annexin-V assays, and notably downregulated the expression of Bcl-2 while increasing levels of proapoptotic cleaved caspase 3 and BAX in DLBCL cells. Additionally, EP treatment decreased the overexpression of c-Jun in c-Jun-transfected DLBCL cells. Further, EP demonstrated DNA-damaging effects in TUNEL assays. In vivo, xenograft animal models revealed that EP treatment significantly mitigated DLBCL tumor growth and suppressed DLBCL cell adhesion to bone marrow stromal cells. In summary, these findings suggest that EP mitigates DLBCL progression by inducing apoptosis, inducing cell cycle arrest, and promoting DNA damage. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Esketamine inhibits the c-Jun N-terminal kinase pathway in the spinal dorsal horn to relieve bone cancer pain in rats.
- Author
-
Duan, Chenxia, Zhu, Yi, Zhang, Zhuoliang, Wu, Tiantian, Shen, Mengwei, Xu, Jinfu, Gao, Wenxin, Pan, Jianhua, Wei, Lei, Su, Huibin, and Shi, Chenghuan
- Abstract
Cancer-induced bone pain (CIBP) is one of the most common and feared symptoms in patients with advanced tumors. The X-C motif chemokine ligand 12 (CXCL12) and the CXCR4 receptor have been associated with glial cell activation in bone cancer pain. Moreover, mitogen-activated protein kinases (MAPKs), as downstream CXCL12/CXCR4 signals, and c-Jun, as activator protein AP-1 components, contribute to the development of various types of pain. However, the specific CIBP mechanisms remain unknown. Esketamine is a non-selective N-methyl- d -aspartic acid receptor (NMDA) inhibitor commonly used as an analgesic in the clinic, but its analgesic mechanism in bone cancer pain remains unclear. We used a tumor cell implantation (TCI) model and explored that CXCL12/CXCR4, p-MAPKs, and p-c-Jun were stably up-regulated in the spinal cord. Immunofluorescence images showed activated microglia in the spinal cord on day 14 after TCI and co-expression of CXCL12/CXCR4, p-MAPKs (p-JNK, p-ERK, p-p38 MAPK), and p-c-Jun in microglia. Intrathecal injection of the CXCR4 inhibitor AMD3100 reduced JNK and c-Jun phosphorylations, and intrathecal injection of the JNK inhibitor SP600125 and esketamine also alleviated TCI-induced pain and reduced the expression of p-JNK and p-c-Jun in microglia. Overall, our data suggest that the CXCL12/CXCR4-JNK-c-Jun signaling pathway of microglia in the spinal cord mediates neuronal sensitization and pain hypersensitivity in cancer-induced bone pain and that esketamine exerts its analgesic effect by inhibiting the JNK-c-Jun pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Annonacin and Squamocin Conjugation with Nanodiamond Alters Metastatic Marker Expression in Breast Cancer Cell Line.
- Author
-
Primula Dewi, Firli Rahmah, Astuti Wahyuningsih, Sri Puji, Milenia Sari, Adelah Putri, Alfiah, Umi Nur, Lim, Vuanghao, Zubaidah, Ummi, and Hayati, Alfiah
- Subjects
- *
GENE expression , *BREAST cancer , *CANCER cells , *CELL survival , *SCANNING electron microscopy , *BREAST - Abstract
Breast cancer can perform metastasis to distant organs and cause more than 90% of malignancy-related deaths. The anti-metastasis potency of nanodiamondconjugated annonacin and squamocin against MCF-7 cells is currently studied. First, IC50 determination of both free annonacin and squamocin to evaluate their potency as cytotoxic agents. Upon getting the IC50 value, both compounds are conjugated into nanodiamonds. Drug loading efficiencies of nanodiamondconjugated annonacin and squamocin are 88.9% and 89.1%, respectively. Meanwhile, the ND-annonacin and ND-squamocin complex size is 150-300 nm based on SEM imaging. Subsequently, cell viability assessment of MCF-7 was performed with six cohort designs, namely, K (control cell), AN (annonacin), SQ (squamocin), NDAN (nanodiamond-conjugated annonacin), and NDSQ (nanodiamond-conjugated squamocin). Both IC50 and cell viability are assessed by MTT assay after 24 h incubation. All cohorts also underwent gene expression analysis subject to the metastasis markers CTNND1 (catenin delta 1), NOTCH4, and C-JUN. Here, the IC50 of both free annonacin (4.52 µg/ml) and squamocin (10.03 µg/ml) are more than IC50 of potent anticancer (< 4 µg/ml) for pure compounds. However, nanodiamond conjugation to both compounds can decrease cell viability better than free compounds. Compared to K, nanodiamond-conjugated annonacin and squamocin significantly decreases cell viability after 24 h incubation. Bioinformatics analysis confirmed significant prometastasis (C-JUN and NOTCH4) upregulation and anti-metastasis (CTNND1) downregulation in tumors compared to normal. Recent findings demonstrated that nanodiamond-conjugated annonacin can significantly upregulate CTNND1 and significantly downregulate C-JUN and NOTCH4. Even so, nanodiamondconjugated squamocin upregulate CTNND1 but not significantly and significantly downregulate C-JUN and NOTCH4. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Activation of Alternate Prosurvival Pathways Accounts for Acquired Sunitinib Resistance in U87MG Glioma Xenografts
- Author
-
Zhou, Qingyu, Lv, Hua, Mazloom, Amin R., Xu, Huilei, Ma'ayan, Avi, and Gallo, James M.
- Published
- 2012
- Full Text
- View/download PDF
32. The role of c-Jun for beating cardiomycyte formation in prepared embryonic body
- Author
-
Lide Su, Guofu Zhang, Lili Jiang, Chao Chi, Bing Bai, and Kai Kang
- Subjects
C-Jun ,Cardiac differentiation ,Mesoderm ,Endoderm ,Embryonic body ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Morbidity and mortality associated with cardiovascular diseases, such as myocardial infarction, stem from the inability of terminally differentiated cardiomyocytes to regenerate, and thus repair the damaged myocardial tissue structure. The molecular biological mechanisms behind the lack of regenerative capacity for those cardiomyocytes remains to be fully elucidated. Recent studies have shown that c-Jun serves as a cell cycle regulator for somatic cell fates, playing a key role in multiple molecular pathways, including the inhibition of cellular reprogramming, promoting angiogenesis, and aggravation of cardiac hypertrophy, but its role in cardiac development is largely unknown. This study aims to delineate the role of c-Jun in promoting early-stage cardiac differentiation. Methods The c-Jun gene in mouse embryonic stem cells (mESCs) was knocked out with CRISPR-Cas9, and the hanging drop method used to prepare the resulting embryoid bodies. Cardiac differentiation was evaluated up to 9 days after c-Jun knockout (ko) via immunofluorescence, flow cytometric, and qPCR analyses. Results Compared to the wild-type control group, obvious beating was observed among the c-Jun-ko mESCs after 6 days, which was also associated with significant increases in myocardial marker expression. Additionally, markers associated with mesoderm and endoderm cell layer development, essential for further differentiation of ESCs into cardiomyocytes, were also up-regulated in the c-Jun-ko cell group. Conclusions Knocking out c-Jun directs ESCs toward a meso-endodermal cell lineage fate, in turn leading to generation of beating myocardial cells. Thus, c-Jun plays an important role in regulating early cardiac cell development.
- Published
- 2023
- Full Text
- View/download PDF
33. Role of the Egr2 Promoter Antisense RNA in Modulating the Schwann Cell Chromatin Landscape
- Author
-
Margot Martinez Moreno, David Karambizi, Hyeyeon Hwang, Kristen Fregoso, Madison J. Michles, Eduardo Fajardo, Andras Fiser, and Nikos Tapinos
- Subjects
promoter antisense-RNA ,EGR2 ,C-JUN ,EZH2 ,WDR5 ,AP-1 ,Biology (General) ,QH301-705.5 - Abstract
Background: Schwann cells (SCs) and their plasticity contribute to the peripheral nervous system’s capacity for nerve regeneration after injury. The Egr2/Krox20 promoter antisense RNA (Egr2-AS) recruits chromatin remodeling complexes to inhibit Egr2 transcription following peripheral nerve injury. Methods: RNA-seq and ATAC-seq were performed on control cells, Lenti-GFP-transduced cells, and cells overexpressing Egr2-AS (Lenti-AS). Egr2 AS-RNA was cloned into the pLVX-DsRed-Express2-N1 lentiviral expression vector (Clontech, Mountain View, CA, USA), and the levels of AS-RNA expression were determined. Ezh2 and Wdr5 were immunoprecipitated from rat SCs and RT-qPCR was performed against AS-Egr2 RNA. ChIP followed by DNA purification columns was used to perform qPCR for relevant promoters. Hi-C, HiC-DC+, R, Bioconductor, and TOBIAS were used for significant and differential loop analysis, identifications of COREs and CORE-promotor loops, comparisons of TF activity at promoter sites, and identification of site-specific TF footprints. OnTAD was used to detect TADs, and Juicer was used to identify A/B compartments. Results: Here we show that a Neuregulin-ErbB2/3 signaling axis mediates binding of the Egr2-AS to YY1Ser184 and regulates its expression. Egr2-AS modulates the chromatin accessibility of Schwann cells and interacts with two distinct histone modification complexes. It binds to EZH2 and WDR5 and enables targeting of H3K27me3 and H3K4me3 to promoters of Egr2 and C-JUN, respectively. Expression of the Egr2-AS results in reorganization of the global chromatin landscape and quantitative changes in the loop formation and contact frequency at domain boundaries exhibiting enrichment for AP-1 genes. In addition, the Egr2-AS induces changes in the hierarchical TADs and increases transcription factor binding scores on an inter-TAD loop between a super-enhancer regulatory hub and the promoter of mTOR. Conclusions: Our results show that Neuregulin-ErbB2/3-YY1 regulates the expression of Egr2-AS, which mediates remodeling of the chromatin landscape in Schwann cells.
- Published
- 2024
- Full Text
- View/download PDF
34. ANKRD49 promotes the metastasis of NSCLC via activating JNK-ATF2/c-Jun-MMP-2/9 axis
- Author
-
Jia Sun, Jin-rui Hu, Chao-feng Liu, Yuan Li, Wei Wang, Rong Fu, Min Guo, Hai-long Wang, and Min Pang
- Subjects
ANKRD49 ,Matrix metalloproteinases ,ATF2 ,c-Jun ,Metastasis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Ankyrin repeat domain 49 (ANKRD49) has been found to be highly expressed in multiple cancer including lung adenocarcinoma (LUAD) and lung squamous carcinoma (LUSC). However, the function of ANKRD49 in the pathogenesis of NSCLC still remains elusive. Previously, ANKRD49 has been demonstrated to promote the invasion and metastasis of A549 cells, a LUAD cell line, via activating the p38-ATF-2-MMP2/MMP9 pathways. Considering the heterogeneity of tumor cells, the function and mechanism of ANKRD49 in NSCLC need more NSCLC-originated cells to clarify. Methods Real-time qPCR was employed to test ANKRD49 expression levels in nine pairs of fresh NSCLC tissues and the corresponding adjacent normal tissues. The function of ANKRD49 was investigated using overexpression and RNA interference assays in lung adenocarcinoma cell line (NCI-H1299) and lung squamous carcinoma cell line (NCI-H1703) through gelatin zymography, cell counting kit-8, colony formation, wound healing, migration and invasion assays mmunoprecipitation was performed to in vitro. Immunoprecipitation was performed to test the interaction of c-Jun and ATF2. Chromatin immunoprecipitation was conducted to assess the transcriptional regulation of ATF2/c-Jun on MMP-2/9. Moreover, the tumorigenicity of ANKRD49 was evaluated in nude mice models and the involved signal molecular was also measured by immunohistochemical method. Results We found that the levels of ANKRD49 in cancerous tissues were higher than those in adjacent normal tissues. in vitro assay showed that ANKRD49 promoted the migration and invasion of NCI-H1299 and NCI-H1703 cells via enhancing the levels of MMP-2 and MMP-9. Furthermore, ANKRD49 elevated phosphorylation of JNK and then activated c-Jun and ATF2 which interact in nucleus to promote the binding of ATF2:c-Jun with the promoter MMP-2 or MMP-9. In vivo assay showed that ANKRD49 promoted lung metastasis of injected-NSCLC cells and the high metastatic rate was positively correlated with the high expression of ANKRD49, MMP-2, MMP-9, p-JNK, p-c-Jun and p-ATF2. Conclusion The present study indicated that ANKRD49 accelerated the invasion and metastasis of NSCLC cells via JNK-mediated transcription activation of c-Jun and ATF2 which regulated the expression of MMP-2/MMP-9. The molecular mechanisms of ANKRD49’s function is different from those found in A549 cells. The current study is a supplement and improvement to the previous research.
- Published
- 2023
- Full Text
- View/download PDF
35. Neurotrophin-3 promotes peripheral nerve regeneration by maintaining a repair state of Schwann cells after chronic denervation via the TrkC/ERK/c-Jun pathway
- Author
-
Xiong Xu, Lili Song, Yueying Li, Jin Guo, Shuo Huang, Shuang Du, Weizhen Li, Rangjuan Cao, and Shusen Cui
- Subjects
Neurotrophin-3 ,c-Jun ,Chronic denervation ,Peripheral nerve regeneration ,Repair Schwann cell ,ERK pathway ,Medicine - Abstract
Abstract Background Maintaining the repair phenotype of denervated Schwann cells in the injured distal nerve is crucial for promoting peripheral nerve regeneration. However, when chronically denervated, the capacity of Schwann cells to support repair and regeneration deteriorates, leading to peripheral nerve regeneration and poor functional recovery. Herein, we investigated whether neurotrophin-3 (NT-3) could sustain the reparative phenotype of Schwann cells and promote peripheral nerve regeneration after chronic denervation and aimed to uncover its potential molecular mechanisms. Methods Western blot was employed to investigate the relationship between the expression of c-Jun and the reparative phenotype of Schwann cells. The inducible expression of c-Jun by NT-3 was examined both in vitro and in vivo with western blot and immunofluorescence staining. A chronic denervation model was established to study the role of NT-3 in peripheral nerve regeneration. The number of regenerated distal axons, myelination of regenerated axons, reinnervation of neuromuscular junctions, and muscle fiber diameters of target muscles were used to evaluate peripheral nerve regeneration by immunofluorescence staining, transmission electron microscopy (TEM), and hematoxylin and eosin (H&E) staining. Adeno-associated virus (AAV) 2/9 carrying shRNA, small molecule inhibitors, and siRNA were employed to investigate whether NT-3 could signal through the TrkC/ERK pathway to maintain c-Jun expression and promote peripheral nerve regeneration after chronic denervation. Results After peripheral nerve injury, c-Jun expression progressively increased until week 5 and then began to decrease in the distal nerve following denervation. NT-3 upregulated the expression of c-Jun in denervated Schwann cells, both in vitro and in vivo. NT-3 promoted peripheral nerve regeneration after chronic denervation, mainly by upregulating or maintaining a high level of c-Jun rather than NT-3 itself. The TrkC receptor was consistently presented on denervated Schwann cells and served as NT-3 receptors following chronic denervation. NT-3 mainly upregulated c-Jun through the TrkC/ERK pathway. Conclusion NT-3 promotes peripheral nerve regeneration by maintaining the repair phenotype of Schwann cells after chronic denervation via the TrkC/ERK/c-Jun pathway. It provides a potential target for the clinical treatment of peripheral nerve injury after chronic denervation.
- Published
- 2023
- Full Text
- View/download PDF
36. c-Jun as a one-way valve at the naive to primed interface
- Author
-
Dongwei Li, Ling Luo, Lin Guo, Chuman Wu, Ran Zhang, Yuling Peng, Menghua Wu, Junqi Kuang, Yan Li, Yudan Zhang, Jun Xie, Wenxiu Xie, Rui Mao, Gang Ma, Xiuling Fu, Jiekai Chen, Andrew P. Hutchins, and Duanqing Pei
- Subjects
EpiSCs ,c-Jun ,Naïve to primed transition ,Primed to naïve transition ,Biotechnology ,TP248.13-248.65 ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
Abstract Background c-Jun is a proto-oncogene functioning as a transcription factor to activate gene expression under many physiological and pathological conditions, particularly in somatic cells. However, its role in early embryonic development remains unknown. Results Here, we show that c-Jun acts as a one-way valve to preserve the primed state and impair reversion to the naïve state. c-Jun is induced during the naive to primed transition, and it works to stabilize the chromatin structure and inhibit the reverse transition. Loss of c-Jun has surprisingly little effect on the naïve to primed transition, and no phenotypic effect on primed cells, however, in primed cells the loss of c-Jun leads to a failure to correctly close naïve-specific enhancers. When the primed cells are induced to reprogram to a naïve state, these enhancers are more rapidly activated when c-Jun is lost or impaired, and the conversion is more efficient. Conclusions The results of this study indicate that c-Jun can function as a chromatin stabilizer in primed EpiSCs, to maintain the epigenetic cell type state and act as a one-way valve for cell fate conversions.
- Published
- 2023
- Full Text
- View/download PDF
37. c-Jun signaling during initial HSV-1 infection modulates latency to enhance later reactivation in addition to directly promoting the progression to full reactivation.
- Author
-
Dochnal, Sara A., Whitford, Abigail L., Francois, Alison K., Krakowiak, Patryk A., Cuddy, Sean, and Cliffe, Anna R.
- Subjects
- *
LATENT infection , *HUMAN herpesvirus 1 , *SYMPTOMS , *PHASE transitions , *INFECTION - Abstract
Herpes simplex virus-1 (HSV-1) establishes a latent infection in peripheral neurons and periodically reactivates to permit transmission, which can result in clinical manifestations. Viral transactivators required for lytic infection are largely absent during latent infection, and therefore, HSV-1 relies on the co-option of neuronal host signaling pathways to initiate its gene expression. The activation of the neuronal c-Jun N-terminal kinase (JNK) cell stress pathway is central to initiating biphasic reactivation in response to multiple stimuli. However, how host factors work with JNK to stimulate the initial wave of gene expression (known as Phase I) or the progression to full Phase II reactivation remains unclear. Here, we found that c-Jun, the primary target downstream of neuronal JNK cell stress signaling, functions during reactivation but not during the JNK-mediated initiation of Phase I gene expression. Instead, c-Jun was required to transition from Phase I to full HSV-1 reactivation and was detected in viral replication compartments of reactivating neurons. Interestingly, we also identified a role for both c-Jun and enhanced neuronal stress during initial neuronal infection in promoting a more reactivation-competent form of HSV-1 latency. Therefore, c-Jun functions at multiple stages during the HSV latent infection of neurons to promote reactivation but not during the initial JNK-dependent Phase I. Importantly, by demonstrating that initial infection conditions can contribute to later reactivation abilities, this study highlights the potential for latently infected neurons to maintain a molecular scar of previous exposure to neuronal stressors. IMPORTANCE The molecular mechanisms that regulate the reactivation of herpes simplex virus-1 (HSV-1) from latent infection are unknown. The host transcription and pioneer factor c-Jun is the main target of the JNK cell stress pathway that is known to be important in exit of HSV from latency. Surprisingly, we found that c-Jun does not act with JNK during exit from latency but instead promotes the transition to full reactivation. Moreover, c-Jun and enhanced neuronal stress during initial neuronal infection promoted a more reactivation-competent form of HSV-1 latency. c-Jun, therefore, functions at multiple stages during HSV-1 latent infection of neurons to promote reactivation. Importantly, this study contributes to a growing body of evidence that de novo HSV-1 infection conditions can modulate latent infection and impact future reactivation events, raising important questions on the clinical impact of stress during initial HSV-1 acquisition on future reactivation events and consequences. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Oleanolic acid attenuates hydrogen peroxide‐induced apoptosis of IPEC‐J2 cells through suppressing c‐Jun and MAPK pathway.
- Author
-
Hu, Mingyang, Zhang, Lei, Jia, Hongpeng, Xue, Chenyu, Zhao, Lu, Dong, Na, and Shan, Anshan
- Subjects
MITOGEN-activated protein kinases ,GENE expression ,GENE expression profiling ,WESTERN immunoblotting ,APOPTOSIS - Abstract
Oleanolic acid (OA) is a natural triterpenoid with therapeutic potential for a multitude of diseases. However, the precise mechanism by which OA influences stress‐induced apoptosis of intestinal epithelial cells remains elusive. Therefore, the effect of OA on intestinal diseases under stressful conditions and its possible mechanisms have been investigated. In a hydrogen peroxide (H2O2)‐induced oxidative stress model, OA attenuated H2O2‐induced apoptosis in a concentration‐dependent manner. To investigate the underlying mechanisms, the gene expression profile of OA on IPEC‐J2 cells was analyzed using an RNA sequencing system. Results from gene ontology and Kyoto encyclopedia of genes and genomes analysis confirmed that OA may mitigate the cytotoxic effects of H2O2 by downregulating gene expression through the MAPK signaling pathway. Furthermore, Quantitative real‐time polymerase chain reaction results validated the differentially expressed genes data. Western blot analysis further demonstrated that OA effectively suppressed the expression level of c‐Jun protein induced by H2O2 in IPEC‐J2 cells. Collectively, our results indicate that OA pretreatment significantly attenuated H2O2‐induced apoptosis in intestinal epithelial cells through suppressing c‐Jun and MAPK pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. The role of c-Jun for beating cardiomycyte formation in prepared embryonic body.
- Author
-
Su, Lide, Zhang, Guofu, Jiang, Lili, Chi, Chao, Bai, Bing, and Kang, Kai
- Subjects
MESODERM ,EMBRYONIC stem cells ,CARDIAC hypertrophy ,SOMATIC cells ,HEART cells ,CARDIOVASCULAR disease related mortality - Abstract
Background: Morbidity and mortality associated with cardiovascular diseases, such as myocardial infarction, stem from the inability of terminally differentiated cardiomyocytes to regenerate, and thus repair the damaged myocardial tissue structure. The molecular biological mechanisms behind the lack of regenerative capacity for those cardiomyocytes remains to be fully elucidated. Recent studies have shown that c-Jun serves as a cell cycle regulator for somatic cell fates, playing a key role in multiple molecular pathways, including the inhibition of cellular reprogramming, promoting angiogenesis, and aggravation of cardiac hypertrophy, but its role in cardiac development is largely unknown. This study aims to delineate the role of c-Jun in promoting early-stage cardiac differentiation. Methods: The c-Jun gene in mouse embryonic stem cells (mESCs) was knocked out with CRISPR-Cas9, and the hanging drop method used to prepare the resulting embryoid bodies. Cardiac differentiation was evaluated up to 9 days after c-Jun knockout (ko) via immunofluorescence, flow cytometric, and qPCR analyses. Results: Compared to the wild-type control group, obvious beating was observed among the c-Jun-ko mESCs after 6 days, which was also associated with significant increases in myocardial marker expression. Additionally, markers associated with mesoderm and endoderm cell layer development, essential for further differentiation of ESCs into cardiomyocytes, were also up-regulated in the c-Jun-ko cell group. Conclusions: Knocking out c-Jun directs ESCs toward a meso-endodermal cell lineage fate, in turn leading to generation of beating myocardial cells. Thus, c-Jun plays an important role in regulating early cardiac cell development. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Aspartoacylase suppresses prostate cancer progression by blocking LYN activation
- Author
-
Hong Weng, Kang-Ping Xiong, Wang Wang, Kai-Yu Qian, Shuai Yuan, Gang Wang, Fang Yu, Jun Luo, Meng-Xin Lu, Zhong-Hua Yang, Tao Liu, Xing Huang, Hang Zheng, and Xing-Huan Wang
- Subjects
Prostate cancer ,Aspartoacylase ,LYN ,JNK ,AP-1 ,C-Jun ,Medicine (General) ,R5-920 ,Military Science - Abstract
Abstract Background Globally, despite prostate cancer (PCa) representing second most prevalent malignancy in male, the precise molecular mechanisms implicated in its pathogenesis remain unclear. Consequently, elucidating the key molecular regulators that govern disease progression could substantially contribute to the establishment of novel therapeutic strategies, ultimately advancing the management of PCa. Methods A total of 49 PCa tissues and 43 adjacent normal tissues were collected from January 2017 to December 2021 at Zhongnan Hospital of Wuhan University. The advanced transcriptomic methodologies were employed to identify differentially expressed mRNAs in PCa. The expression of aspartoacylase (ASPA) in PCa was thoroughly evaluated using quantitative real-time PCR and Western blotting techniques. To elucidate the inhibitory role of ASPA in PCa cell proliferation and metastasis, a comprehensive set of in vitro and in vivo assays were conducted, including orthotopic and tumor-bearing mouse models (n = 8 for each group). A combination of experimental approaches, such as Western blotting, luciferase assays, immunoprecipitation assays, mass spectrometry, glutathione S-transferase pull-down experiments, and rescue studies, were employed to investigate the underlying molecular mechanisms of ASPA’s action in PCa. The Student’s t-test was employed to assess the statistical significance between two distinct groups, while one-way analysis of variance was utilized for comparisons involving more than two groups. A two-sided P value of less than 0.05 was deemed to indicate statistical significance. Results ASPA was identified as a novel inhibitor of PCa progression. The expression of ASPA was found to be significantly down-regulated in PCa tissue samples, and its decreased expression was independently associated with patients’ prognosis (HR = 0.60, 95% CI 0.40–0.92, P = 0.018). Our experiments demonstrated that modulation of ASPA activity, either through gain- or loss-of-function, led to the suppression or enhancement of PCa cell proliferation, migration, and invasion, respectively. The inhibitory role of ASPA in PCa was further confirmed using orthotopic and tumor-bearing mouse models. Mechanistically, ASPA was shown to directly interact with the LYN and inhibit the phosphorylation of LYN as well as its downstream targets, JNK1/2 and C-Jun, in both PCa cells and mouse models, in an enzyme-independent manner. Importantly, the inhibition of LYN activation by bafetinib abrogated the promoting effect of ASPA knockdown on PCa progression in both in vitro and in vivo models. Moreover, we observed an inverse relationship between ASPA expression and LYN activity in clinical PCa samples, suggesting a potential regulatory role of ASPA in modulating LYN signaling. Conclusion Our findings provide novel insights into the tumor-suppressive function of ASPA in PCa and highlight its potential as a prognostic biomarker and therapeutic target for the management of this malignancy.
- Published
- 2023
- Full Text
- View/download PDF
41. Detrimental alteration of mesenchymal stem cells by an articular inflammatory microenvironment results in deterioration of osteoarthritis
- Author
-
Mengqiang Fan, Peijian Tong, Li Yan, Ting Li, Jiadan Ren, Jiefeng Huang, Wenxi Du, Li Zhou, and Letian Shan
- Subjects
Knee osteoarthritis ,Synovial fluid ,Human umbilical cord mesenchymal stem cells ,Articular inflammatory microenvironment ,MMP13 ,c-Jun ,Medicine - Abstract
Abstract Background Articular injection of mesenchymal stem cells (MSCs) has been applied to treat knee osteoarthritis (kOA), but its clinical outcomes are controversial. This study investigated whether an articular inflammatory microenvironment (AIM) impacts MSC-based therapy in a rat model of kOA. Methods The biological change of MSCs and the functional change of MSCs on chondrocytes were evaluated under AIM. The key mediator and mechanism for the AIM impact on MSC therapy were explored via gain- and loss-of-function approaches. Results The results showed that MSCs exerted potent anti-kOA effects in vivo and in vitro, but that this therapy become chondrodestructive if a chronic AIM was present. Mechanistically, the overexpression of MMP13 in the injected MSCs via a MAPKs-AP1 signaling axis was revealed as the underlying mechanism for the detriment outcome. Conclusions This study thus clarifies recent clinical findings while also suggesting a means to overcome any detrimental effects of MSC-based therapy while improving its efficacy.
- Published
- 2023
- Full Text
- View/download PDF
42. HO-1 attenuates testicular ischaemia/reperfusion injury by activating the phosphorylated C-jun-miR-221/222-TOX pathway
- Author
-
Bo Xie, Bing Cheng, Lugeng He, Yunfu Liu, and Ning He
- Subjects
Heme oxygenase (HO-1) ,Knockout rats ,miR-221/222 ,c-Jun ,Testicular ischaemia/reperfusion ,Gene sequencing ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Aims: Heme oxygenase (HO-1) affords protection against ischaemia/reperfusion (I/R) injury; however, its effects on testicular I/R injury remain poorly explored. Herein, we aimed to examine the effects of HO-1 on testicular I/R injury and elucidate the underlying mechanism. Methods: Using the TALEN technique, we knocked out the HO-1 gene from rats. In vivo: Thirty hmox+/+ and 30 hmox−/− rats were randomly assigned to six groups: sham-operated (sham), I/R (the left testicle torsion/detorsion) 0 d,I/R 1d, I/R 3d, I/R 7d and I/R 28d. In vitro: GC-1 were suffered from: control,H/R (oxygen-deprivation/reoxygenation),H/R + HO-1 siRNA,H/R + c-Jun siRNA or H/R + HO-1 siRNA + c-jun.We performed immunofluorescence and immunohistochemistry experiments to detect HO-1 nuclear translocation. Flow cytometry was used to detect cell apoptosis and analyse the cell cycle. High-resolution miRNA, mRNA sequencing, reverse transcription-quantitative PCR, and western blotting were performed to identify testicular I/R injury-related genes strongly conserved in HO-1 knockout rats. A double luciferase reporter assay was performed to verify the relationship between C-jun and miR-221/222. Main findings: In vivo, HO-1 improved the pathological damage induced by testicular I/R. In GC-1 cells, we confirmed the nuclear translocation of HO-1 and its protective effect against hypoxia/reoxygenation (H/R) damage. Accordingly, HO-1 protein itself, rather than heme metabolites, might play a key role in testicular I/R. Gene sequencing was performed to screen for miR221/222 and its downstream gene, thymocyte selection-associated high mobility group box (TOX). HO-1 increased c-Jun phosphorylation in the H/R group, knocked down c-Jun in GC-1 cells, and decreased miR-221/222 expression. Inhibition of HO-1 expression decreased the expression of c-Jun and miR-221/222, which was rescued by adding c-Jun. Dual-luciferase reporter assay confirmed the interaction between c-Jun and miR-221/222. Conclusions: HO-1 could exert a protective effect against testicular I/R via the phosphorylated c-Jun-miR-221/222-TOX pathway.
- Published
- 2024
- Full Text
- View/download PDF
43. The m 7 G Reader NCBP2 Promotes Pancreatic Cancer Progression by Upregulating MAPK/ERK Signaling.
- Author
-
Xie, Jiancong, Mo, Taiwei, Li, Ruibing, Zhang, Hao, Liang, Guanzhan, Ma, Tao, Chen, Jing, Xie, Hanlin, Wen, Xiaofeng, Hu, Tuo, Xian, Zhenyu, and Pan, Weidong
- Subjects
- *
PANCREATIC tumors , *ONCOGENES , *ANIMAL experimentation , *CELLULAR signal transduction , *GENE expression , *GENES , *CELL proliferation , *RESEARCH funding , *MITOGEN-activated protein kinases , *CELL lines , *CARRIER proteins , *MICE - Abstract
Simple Summary: N7-methylguanosine (m7G) methylation plays an important role in the development of pancreatic adenocarcinoma (PDAC). Among the 29 regulatory genes of m7G methylation, they can be roughly divided into three categories: methyltransferase (writer), demethylase (eraser), and binding protein (reader) according to their functions. In this study, we explore the role of m7G reader NCBP2 in PDAC progression: NCBP2 activates MEK/ERK signaling by upregulating c-JUN, thereby promoting PDAC cell proliferation. In summary, we identify a novel potential therapeutic target for PDAC, which sheds new light on the treatment of PDAC patients. PDAC is one of the most common malignant tumors worldwide. The difficulty of early diagnosis and lack of effective treatment are the main reasons for its poor prognosis. Therefore, it is urgent to identify novel diagnostic and therapeutic targets for PDAC patients. The m7G methylation is a common type of RNA modification that plays a pivotal role in regulating tumor development. However, the correlation between m7G regulatory genes and PDAC progression remains unclear. By integrating gene expression and related clinical information of PDAC patients from TCGA and GEO cohorts, m7G binding protein NCBP2 was found to be highly expressed in PDAC patients. More importantly, PDAC patients with high NCBP2 expression had a worse prognosis. Stable NCBP2-knockdown and overexpression PDAC cell lines were constructed to further perform in-vitro and in-vivo experiments. NCBP2-knockdown significantly inhibited PDAC cell proliferation, while overexpression of NCBP2 dramatically promoted PDAC cell growth. Mechanistically, NCBP2 enhanced the translation of c-JUN, which in turn activated MEK/ERK signaling to promote PDAC progression. In conclusion, our study reveals that m7G reader NCBP2 promotes PDAC progression by activating MEK/ERK pathway, which could serve as a novel therapeutic target for PDAC patients. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
44. ANKRD49 promotes the metastasis of NSCLC via activating JNK-ATF2/c-Jun-MMP-2/9 axis.
- Author
-
Sun, Jia, Hu, Jin-rui, Liu, Chao-feng, Li, Yuan, Wang, Wei, Fu, Rong, Guo, Min, Wang, Hai-long, and Pang, Min
- Subjects
RNA interference ,NON-small-cell lung carcinoma ,SMALL interfering RNA ,SQUAMOUS cell carcinoma ,METASTASIS - Abstract
Background: Ankyrin repeat domain 49 (ANKRD49) has been found to be highly expressed in multiple cancer including lung adenocarcinoma (LUAD) and lung squamous carcinoma (LUSC). However, the function of ANKRD49 in the pathogenesis of NSCLC still remains elusive. Previously, ANKRD49 has been demonstrated to promote the invasion and metastasis of A549 cells, a LUAD cell line, via activating the p38-ATF-2-MMP2/MMP9 pathways. Considering the heterogeneity of tumor cells, the function and mechanism of ANKRD49 in NSCLC need more NSCLC-originated cells to clarify. Methods: Real-time qPCR was employed to test ANKRD49 expression levels in nine pairs of fresh NSCLC tissues and the corresponding adjacent normal tissues. The function of ANKRD49 was investigated using overexpression and RNA interference assays in lung adenocarcinoma cell line (NCI-H1299) and lung squamous carcinoma cell line (NCI-H1703) through gelatin zymography, cell counting kit-8, colony formation, wound healing, migration and invasion assays mmunoprecipitation was performed to in vitro. Immunoprecipitation was performed to test the interaction of c-Jun and ATF2. Chromatin immunoprecipitation was conducted to assess the transcriptional regulation of ATF2/c-Jun on MMP-2/9. Moreover, the tumorigenicity of ANKRD49 was evaluated in nude mice models and the involved signal molecular was also measured by immunohistochemical method. Results: We found that the levels of ANKRD49 in cancerous tissues were higher than those in adjacent normal tissues. in vitro assay showed that ANKRD49 promoted the migration and invasion of NCI-H1299 and NCI-H1703 cells via enhancing the levels of MMP-2 and MMP-9. Furthermore, ANKRD49 elevated phosphorylation of JNK and then activated c-Jun and ATF2 which interact in nucleus to promote the binding of ATF2:c-Jun with the promoter MMP-2 or MMP-9. In vivo assay showed that ANKRD49 promoted lung metastasis of injected-NSCLC cells and the high metastatic rate was positively correlated with the high expression of ANKRD49, MMP-2, MMP-9, p-JNK, p-c-Jun and p-ATF2. Conclusion: The present study indicated that ANKRD49 accelerated the invasion and metastasis of NSCLC cells via JNK-mediated transcription activation of c-Jun and ATF2 which regulated the expression of MMP-2/MMP-9. The molecular mechanisms of ANKRD49's function is different from those found in A549 cells. The current study is a supplement and improvement to the previous research. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
45. c‐JUN‐induced upregulation of LINC00174 contributes to colorectal cancer proliferation and invasion through accelerating USP21 expression.
- Author
-
Liang, Weijie, Liu, Dongdong, and Wu, Jie
- Subjects
- *
LINCRNA , *DEUBIQUITINATING enzymes , *COLORECTAL cancer , *OVERALL survival , *PROGRESSION-free survival , *PROGNOSIS - Abstract
Colorectal cancer (CRC) is one of the most common human malignancies due to its invasiveness and metastasis. Recent studies revealed the pivotal roles of long noncoding RNAs (lncRNAs) in tumorigenesis and progressions of various tumors. However, the biological roles and molecular mechanisms of long intergenic noncoding RNA 00174 (LINC00174) in human CRC remain unclear. Here, we report that LINC00174 expression was higher in human CRC tissues and cell lines than in adjacent normal tissues and a colon epithelial cell line (FHC). High expression of LINC00174 was positively correlated with poor overall and disease‐free survival in patients with CRC. Loss‐ and gain‐of‐function of LINC00174 demonstrated its critical roles in promoting cell proliferation, apoptosis resistance, migration, and invasion of CRC cells in vitro. Moreover, overexpression of LINC00174 enhanced tumor growth in vivo. Mechanistic experiments revealed that LINC00174 could bind to microRNA (miR)−2467‐3p and augment the expression and function of ubiquitin‐specific peptidase 21 (USP21). Rescue assays found that miR‐2467‐3p inhibition can offset the actions of LINC00174 or USP21 knockdown in CRC cells. Additionally, transcriptional factor c‐JUN transcriptionally activated LINC00174 expression and mediated LINC00174‐induced malignant phenotypes of CRC cell lines. Totally, our findings shed light on a new therapeutic strategy in modulating LINC00174/miR‐2467‐3p, which may interfere with the expression of USP21, and revealed that LINC00174 could be a new therapeutic target or prognostic marker in CRC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. c-Jun as a one-way valve at the naive to primed interface.
- Author
-
Li, Dongwei, Luo, Ling, Guo, Lin, Wu, Chuman, Zhang, Ran, Peng, Yuling, Wu, Menghua, Kuang, Junqi, Li, Yan, Zhang, Yudan, Xie, Jun, Xie, Wenxiu, Mao, Rui, Ma, Gang, Fu, Xiuling, Chen, Jiekai, Hutchins, Andrew P., and Pei, Duanqing
- Subjects
CHECK valves ,SOMATIC cells ,GENE expression ,TRANSCRIPTION factors ,EMBRYOLOGY - Abstract
Background: c-Jun is a proto-oncogene functioning as a transcription factor to activate gene expression under many physiological and pathological conditions, particularly in somatic cells. However, its role in early embryonic development remains unknown. Results: Here, we show that c-Jun acts as a one-way valve to preserve the primed state and impair reversion to the naïve state. c-Jun is induced during the naive to primed transition, and it works to stabilize the chromatin structure and inhibit the reverse transition. Loss of c-Jun has surprisingly little effect on the naïve to primed transition, and no phenotypic effect on primed cells, however, in primed cells the loss of c-Jun leads to a failure to correctly close naïve-specific enhancers. When the primed cells are induced to reprogram to a naïve state, these enhancers are more rapidly activated when c-Jun is lost or impaired, and the conversion is more efficient. Conclusions: The results of this study indicate that c-Jun can function as a chromatin stabilizer in primed EpiSCs, to maintain the epigenetic cell type state and act as a one-way valve for cell fate conversions. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
47. Oleanolic Acid Complexation with Cyclodextrins Improves Its Cell Bio-Availability and Biological Activities for Cell Migration.
- Author
-
Stelling-Férez, Javier, López-Miranda, Santiago, Gabaldón, José Antonio, and Nicolás, Francisco José
- Subjects
- *
CELL migration , *CYCLODEXTRINS , *CHEMICAL stability , *HYDROPHOBIC compounds , *POLYMERSOMES , *CELL preservation , *WOUND healing - Abstract
Wound healing is a complex process to restore skin. Plant-derived bioactive compounds might be a source of substances for the treatment of wounds stalled in a non-resolving stage of wound healing. Oleanolic acid (OA), a pentacyclic triterpene, has shown favorable wound healing properties both in vitro and in vivo. Unfortunately, OA cannot be solubilized in aqueous media, and it needs to be helped by the use of dimethyl sulfoxide (DMSO). In this paper, we have shown that cyclodextrins (CDs) are a good alternative to DMSO as agents to deliver OA to cells, providing better features than DMSO. Cyclodextrins are natural macromolecules that show a unique tridimensional structure that can encapsulate a wide variety of hydrophobic compounds. We have studied the cyclodextrin-encapsulated form of OA with OA/DMSO, comparing their stability, biological properties for cell migration, and cell viability. In addition, detailed parameters related to cell migration and cytoskeletal reorganization have been measured and compared. Our results show that OA-encapsulateds compound exhibit several advantages when compared to non-encapsulated OA in terms of chemical stability, migration enhancement, and preservation of cell viability. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
48. Neurotrophin-3 promotes peripheral nerve regeneration by maintaining a repair state of Schwann cells after chronic denervation via the TrkC/ERK/c-Jun pathway.
- Author
-
Xu, Xiong, Song, Lili, Li, Yueying, Guo, Jin, Huang, Shuo, Du, Shuang, Li, Weizhen, Cao, Rangjuan, and Cui, Shusen
- Abstract
Background: Maintaining the repair phenotype of denervated Schwann cells in the injured distal nerve is crucial for promoting peripheral nerve regeneration. However, when chronically denervated, the capacity of Schwann cells to support repair and regeneration deteriorates, leading to peripheral nerve regeneration and poor functional recovery. Herein, we investigated whether neurotrophin-3 (NT-3) could sustain the reparative phenotype of Schwann cells and promote peripheral nerve regeneration after chronic denervation and aimed to uncover its potential molecular mechanisms. Methods: Western blot was employed to investigate the relationship between the expression of c-Jun and the reparative phenotype of Schwann cells. The inducible expression of c-Jun by NT-3 was examined both in vitro and in vivo with western blot and immunofluorescence staining. A chronic denervation model was established to study the role of NT-3 in peripheral nerve regeneration. The number of regenerated distal axons, myelination of regenerated axons, reinnervation of neuromuscular junctions, and muscle fiber diameters of target muscles were used to evaluate peripheral nerve regeneration by immunofluorescence staining, transmission electron microscopy (TEM), and hematoxylin and eosin (H&E) staining. Adeno-associated virus (AAV) 2/9 carrying shRNA, small molecule inhibitors, and siRNA were employed to investigate whether NT-3 could signal through the TrkC/ERK pathway to maintain c-Jun expression and promote peripheral nerve regeneration after chronic denervation. Results: After peripheral nerve injury, c-Jun expression progressively increased until week 5 and then began to decrease in the distal nerve following denervation. NT-3 upregulated the expression of c-Jun in denervated Schwann cells, both in vitro and in vivo. NT-3 promoted peripheral nerve regeneration after chronic denervation, mainly by upregulating or maintaining a high level of c-Jun rather than NT-3 itself. The TrkC receptor was consistently presented on denervated Schwann cells and served as NT-3 receptors following chronic denervation. NT-3 mainly upregulated c-Jun through the TrkC/ERK pathway. Conclusion: NT-3 promotes peripheral nerve regeneration by maintaining the repair phenotype of Schwann cells after chronic denervation via the TrkC/ERK/c-Jun pathway. It provides a potential target for the clinical treatment of peripheral nerve injury after chronic denervation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
49. Similarity of oncogenic protein expression in KRASG12D gene delivery-based rat pancreatic cancer model to that of human pancreatic cancer.
- Author
-
Tanaka, Yuto, Kamimura, Kenya, Shibata, Osamu, Ogawa, Kohei, Oda, Chiyumi, Abe, Hiroyuki, Ikarashi, Satoshi, Hayashi, Kazunao, Yokoo, Takeshi, Wakai, Toshifumi, and Terai, Shuji
- Subjects
- *
ONCOGENIC proteins , *PANCREATIC cancer , *PROTEIN expression , *PANCREATIC tumors , *GENE expression - Abstract
The development of effective therapies and biomarkers for pancreatic cancer is an unmet clinical need. To address this, we have developed an easy-to-use pancreatic cancer rat animal model via pancreas-targeted hydrodynamic gene delivery of human pancreatic cancer-related genes. Our study aimed to determine the molecular similarity between the pancreatic tumor in the rat model and human pancreatic cancer. KRAS G12D gene-expressing plasmid was delivered to the pancreas of wild type rats via pancreas-targeted hydrodynamic gene delivery as previously reported. Tissue samples were collected at 5 weeks after the first gene delivery. The tumors developed in the rats were assessed for the expression of oncogenic proteins that are involved in human pancreatic cancer development. The development of a tumor mimicking pancreatic ductal adenocarcinoma was confirmed. The expression levels of Cyclin D1, c-Jun, IL-33, and Zip4 proteins in the tumor were immunohistochemically assessed and the correlation of the proteins was confirmed. The expression pattern showed similarity to that of surgically resected human pancreatic cancer tissues. Our study findings showing a similar pattern of oncogenic protein expression in novel KRAS G12D gene-induced rat pancreatic cancer model and human pancreatic cancer will be useful for establishing novel tumor markers and therapeutic options for pancreatic cancer. [Display omitted] • Pancreas-targeted KRAS G12D gene delivery developed pancreatic cancer in rats. • The pancreatic tumors in the rats showed similar histological character to that of human pancreatic cancer. • Various oncogenic pathways are activated in the rats' pancreatic cancer similar to that of human pancreatic cancer. • Expressions of Cyclin D1, c-Jun, IL-33, and Zip4 protein showed significant correlation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
50. Knockdown of AK142426 suppresses M2 macrophage polarization and inflammation in peritoneal fibrosis via binding to c‐Jun.
- Author
-
Shao, Qiuyuan, Jiang, Chunming, Zhang, Qingyan, Liu, Jing, Jin, Bo, Zhao, Min, and Xia, Yangyang
- Abstract
Background: Peritoneal fibrosis is a common complication of peritoneal dialysis, which may lead to ultrafiltration failure and ultimately treatment discontinuation. LncRNAs participate in many biological processes during tumorigenesis. We investigated the role of AK142426 in peritoneal fibrosis. Methods: The AK142426 level in peritoneal dialysis (PD) fluid was detected by quantitative real‐time‐PCR assay. The M2 macrophage distribution was determined by flow cytometry. The inflammatory cytokines of TNF‐α and TGF‐β1 were measured by ELISA assay. The direct interaction between AK142426 and c‐Jun was evaluated by RNA pull‐down assay. In addition, the c‐Jun and fibrosis related proteins were assessed by western blot analysis. Results: The PD‐induced peritoneal fibrosis mouse model was successfully established. More importantly, PD treatment induced M2 macrophage polarization and the inflammation in PD fluid, which might be associated with exosome transmission. Fortunately, AK142426 was observed to be upregulated in PD fluid. Mechanically, knockdown of AK142426 suppressed M2 macrophage polarization and inflammation. Furthermore, AK142426 could upregulate c‐Jun through binding c‐Jun protein. In rescue experiments, overexpression of c‐Jun could partially abolish the inhibitory effect of sh‐AK142426 on the activation of M2 macrophages and inflammation. Consistently, knockdown of AK142426 alleviated peritoneal fibrosis in vivo. Conclusions: This study demonstrated that knockdown of AK142426 suppressed M2 macrophage polarization and inflammation in peritoneal fibrosis via binding to c‐Jun, suggesting that AK142426 might be a promising therapeutic target for patients of peritoneal fibrosis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.