33,442 results on '"Nuclear Proteins"'
Search Results
2. [DISTRIBUTION OF NUCLEAR PROTEINS AT THE TIME OF NORMAL MITOSIS].
- Author
-
BASSLEER R
- Subjects
- Chick Embryo, Cell Biology, Cell Division, DNA, Fibroblasts, Microscopy, Microscopy, Interference, Mitosis, Nuclear Proteins, Research, Tissue Culture Techniques
- Published
- 1964
3. The Nesprin-1/-2 ortholog ANC-1 regulates organelle positioning in C. elegans independently from its KASH or actin-binding domains
- Author
-
Hao, Hongyan, Kalra, Shilpi, Jameson, Laura E, Guerrero, Leslie A, Cain, Natalie E, Bolivar, Jessica, and Starr, Daniel A
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Actins ,Animals ,Animals ,Genetically Modified ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Calcium-Binding Proteins ,Cell Cycle Proteins ,Cell Nucleus ,Endoplasmic Reticulum ,Lipid Droplets ,Microfilament Proteins ,Mitochondria ,Movement ,Nuclear Proteins ,Organelles ,Protein Binding ,Protein Interaction Domains and Motifs ,Signal Transduction ,Calponins ,C. elegans ,ER ,LINC complexes ,cell biology ,nesprin ,nuclear envelope ,nuclear positioning ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
KASH proteins in the outer nuclear membrane comprise the cytoplasmic half of linker of nucleoskeleton and cytoskeleton (LINC) complexes that connect nuclei to the cytoskeleton. Caenorhabditis elegans ANC-1, an ortholog of Nesprin-1/2, contains actin-binding and KASH domains at opposite ends of a long spectrin-like region. Deletion of either the KASH or calponin homology (CH) domains does not completely disrupt nuclear positioning, suggesting neither KASH nor CH domains are essential. Deletions in the spectrin-like region of ANC-1 led to significant defects, but only recapitulated the null phenotype in combination with mutations in the transmembrane (TM) span. In anc-1 mutants, the endoplasmic reticulum ER, mitochondria, and lipid droplets were unanchored, moving throughout the cytoplasm. The data presented here support a cytoplasmic integrity model where ANC-1 localizes to the ER membrane and extends into the cytoplasm to position nuclei, ER, mitochondria, and other organelles in place.
- Published
- 2021
4. The autophagy adaptor NDP52 and the FIP200 coiled-coil allosterically activate ULK1 complex membrane recruitment
- Author
-
Shi, Xiaoshan, Chang, Chunmei, Yokom, Adam L, Jensen, Liv E, and Hurley, James H
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Generic health relevance ,Allosteric Regulation ,Autophagy ,Autophagy-Related Protein-1 Homolog ,Autophagy-Related Proteins ,Cell Membrane ,HEK293 Cells ,Humans ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,HDX-MS ,autophagy ,biochemistry ,cell biology ,chemical biology ,coiled-coil ,electron microscopy ,human ,mitophagy ,xenophagy ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The selective autophagy pathways of xenophagy and mitophagy are initiated when the adaptor NDP52 recruits the ULK1 complex to autophagic cargo. Hydrogen-deuterium exchange coupled to mass spectrometry (HDX-MS) was used to map the membrane and NDP52 binding sites of the ULK1 complex to unique regions of the coiled coil of the FIP200 subunit. Electron microscopy of the full-length ULK1 complex shows that the FIP200 coiled coil projects away from the crescent-shaped FIP200 N-terminal domain dimer. NDP52 allosterically stimulates membrane-binding by FIP200 and the ULK1 complex by promoting a more dynamic conformation of the membrane-binding portion of the FIP200 coiled coil. Giant unilamellar vesicle (GUV) reconstitution confirmed that membrane recruitment by the ULK1 complex is triggered by NDP52 engagement. These data reveal how the allosteric linkage between NDP52 and the ULK1 complex could drive the first membrane recruitment event of phagophore biogenesis in xenophagy and mitophagy.
- Published
- 2020
5. The Deubiquitinating Enzyme Otub2 Modulates Pancreatic Beta-Cells Function and Survival.
- Subjects
DNA-binding proteins ,NUCLEAR proteins ,PEPTIDES ,PEPTIDE hormones ,DEUBIQUITINATING enzymes - Abstract
A preprint abstract from biorxiv.org discusses the role of the deubiquitinating enzyme Otub2 in modulating pancreatic beta-cell function and survival. The study found that overexpression of Otub2 inhibited the activity of NF-kappaB, a protein involved in inflammation, and increased the expression of genes related to insulin secretion. Otub2 was also shown to interact with proteins involved in NF-kappaB signaling and beta-cell death. The findings suggest that Otub2 plays a key role in regulating beta-cell function and may have implications for understanding and treating conditions such as impaired glucose tolerance. However, it is important to note that this research has not yet undergone peer review. [Extracted from the article]
- Published
- 2024
6. Human iPSC-derived brain pericytes exhibit differences in inflammatory activation compared to primary human brain pericytes.
- Abstract
A preprint abstract from biorxiv.org discusses the use of induced pluripotent stem cell (iPSC)-derived brain pericytes to model inflammatory diseases. The study aimed to investigate the inflammatory response of these cells to common stimuli and compare it to primary human brain pericytes. The researchers found that iPSC-derived pericytes exhibited differences in their inflammatory activation compared to primary human pericytes, raising concerns about their suitability for studying neuroinflammatory diseases. The study highlights the importance of validating lineage specificity when using iPSC-derived cells in research. However, it should be noted that this preprint has not yet undergone peer review. [Extracted from the article]
- Published
- 2024
7. Study Findings on Cell Biology Described by Researchers at Georgia State University (Crosstalk between PKA and PIAS3 regulates cardiac Kv4 channel SUMOylation).
- Subjects
CYTOLOGY ,ION channels ,UBIQUITIN ligases ,NUCLEAR proteins ,REPORTERS & reporting - Abstract
A study conducted by researchers at Georgia State University explores the relationship between PKA and PIAS3 in regulating the SUMOylation of cardiac Kv4 channels. SUMOylation is a post-translational modification that affects the biophysical properties and trafficking of ion channels. The study identifies PIAS3 as a SUMO E3 ligase for Kv4.2 and HCN2 channels, and demonstrates that PKA phosphorylation blocks Kv4.2-K579 SUMOylation. These findings contribute to our understanding of the signaling cascades that control ion channel surface expression. The research was supported by Georgia State University and the National Institutes of Health. [Extracted from the article]
- Published
- 2024
8. Histone serves as an eat-me signal to induce RAGE-mediated phagocytosis.
- Abstract
According to a preprint abstract, researchers have discovered that histones, which are chromosomal proteins in healthy cells, can serve as "eat-me" signals to induce phagocytosis, the process by which cells engulf and remove dead or dying cells. The study found that histones bind to a receptor called RAGE, and this binding is increased when DNA is attached to the histones. The phagocytosis of apoptotic cells is enhanced when histones are present on the cell surface. The study also found that RAGE is required for the removal of histones from cells, and its absence in knockout mice led to delayed wound healing and removal of apoptotic cells. However, it is important to note that this research has not yet undergone peer review. [Extracted from the article]
- Published
- 2024
9. Chromatin enhancer activity of IkBa mediates the exit from nai ve pluripotency (Updated August 22, 2024).
- Published
- 2024
10. MED26-enriched condensates drive erythropoiesis through modulating transcription pausing.
- Subjects
GENETIC transcription ,ERYTHROPOIESIS ,NUCLEAR proteins ,RNA polymerase II - Abstract
The article reports on how MED26-enriched condensates drive erythropoiesis by modulating transcription pausing. Topics include MED26's unique role in maintaining transcriptional repression, its preferential recruitment of pausing factors, and its condensate-forming capability essential for erythroid development.
- Published
- 2024
11. The nucleolar protein NIFK promotes cancer progression via CK1α/β-catenin in metastasis and Ki-67-dependent cell proliferation.
- Author
-
Lin, Tsung-Chieh, Su, Chia-Yi, Wu, Pei-Yu, Lai, Tsung-Ching, Pan, Wen-An, Jan, Yi-Hua, Chang, Yu-Chang, Yeh, Chi-Tai, Chen, Chi-Long, Ger, Luo-Ping, Chang, Hong-Tai, Yang, Chih-Jen, Huang, Ming-Shyan, Liu, Yu-Peng, Lin, Yuan-Feng, Shyy, John Y-J, Tsai, Ming-Daw, and Hsiao, Michael
- Subjects
Animals ,Humans ,Mice ,Lung Neoplasms ,Neoplasm Metastasis ,Disease Models ,Animal ,Casein Kinase II ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Ki-67 Antigen ,Cell Proliferation ,Korea ,beta Catenin ,NIFK ,casein kinase 1 alpha ,cell biology ,human ,lung cancer ,metastasis ,runx1 ,Disease Models ,Animal ,Biochemistry and Cell Biology - Abstract
Nucleolar protein interacting with the FHA domain of pKi-67 (NIFK) is a Ki-67-interacting protein. However, its precise function in cancer remains largely uninvestigated. Here we show the clinical significance and metastatic mechanism of NIFK in lung cancer. NIFK expression is clinically associated with poor prognosis and metastasis. Furthermore, NIFK enhances Ki-67-dependent proliferation, and promotes migration, invasion in vitro and metastasis in vivo via downregulation of casein kinase 1α (CK1α), a suppressor of pro-metastatic TCF4/β-catenin signaling. Inversely, CK1α is upregulated upon NIFK knockdown. The silencing of CK1α expression in NIFK-silenced cells restores TCF4/β-catenin transcriptional activity, cell migration, and metastasis. Furthermore, RUNX1 is identified as a transcription factor of CSNK1A1 (CK1α) that is negatively regulated by NIFK. Our results demonstrate the prognostic value of NIFK, and suggest that NIFK is required for lung cancer progression via the RUNX1-dependent CK1α repression, which activates TCF4/β-catenin signaling in metastasis and the Ki-67-dependent regulation in cell proliferation.
- Published
- 2016
12. Epigenomic reprogramming via HRP2-MINA dictates response to proteasome inhibitors in multiple myeloma with t(4;14) translocation.
- Author
-
Jingjing Wang, Xu Zhu, Lin Dang, Hongmei Jiang, Ying Xie, Xin Li, Jing Guo, Yixuan Wang, Ziyi Peng, Mengqi Wang, Jingya Wang, Sheng Wang, Qian Li, Yafei Wang, Qiang Wang, Lingqun Ye, Lirong Zhang, Zhiqiang Liu, Wang, Jingjing, and Zhu, Xu
- Subjects
- *
PROTEIN metabolism , *CELL differentiation , *PROTEINS , *CHROMOSOMES , *RESEARCH , *PROTEASE inhibitors , *NUCLEAR proteins , *RESEARCH methodology , *EVALUATION research , *COMPARATIVE studies , *GENES , *CHROMOSOME abnormalities , *OXIDOREDUCTASES , *MULTIPLE myeloma , *CELL lines , *EPIGENOMICS , *PHARMACODYNAMICS - Abstract
The chromosomal t(4;14) (p16;q32) translocation drives high expression of histone methyltransferase nuclear SET domain-containing 2 (NSD2) and plays vital roles in multiple myeloma (MM) evolution and progression. However, the mechanisms of NSD2-driven epigenomic alterations in chemoresistance to proteasome inhibitors (PIs) are not fully understood. Using a CRISPR/Cas9 sgRNA library in a bone marrow-bearing MM model, we found that hepatoma-derived growth factor 2 (HRP2) was a suppressor of chemoresistance to PIs and that its downregulation correlated with a poor response and worse outcomes in the clinic. We observed suppression of HRP2 in bortezomib-resistant MM cells, and knockdown of HRP2 induced a marked tolerance to PIs. Moreover, knockdown of HRP2 augmented H3K27me3 levels, consequentially intensifying transcriptome alterations promoting cell survival and restriction of ER stress. Mechanistically, HRP2 recognized H3K36me2 and recruited the histone demethylase MYC-induced nuclear antigen (MINA) to remove H3K27me3. Tazemetostat, a highly selective epigenetic inhibitor that reduces H3K27me3 levels, synergistically sensitized the anti-MM effects of bortezomib both in vitro and in vivo. Collectively, these results provide a better understanding of the origin of chemoresistance in patients with MM with the t(4;14) translocation and a rationale for managing patients with MM who have different genomic backgrounds. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
13. NOCA-1 functions with γ-tubulin and in parallel to Patronin to assemble non-centrosomal microtubule arrays in C. elegans.
- Author
-
Wang, Shaohe, Wu, Di, Quintin, Sophie, Green, Rebecca A, Cheerambathur, Dhanya K, Ochoa, Stacy D, Desai, Arshad, and Oegema, Karen
- Subjects
Microtubules ,Animals ,Caenorhabditis elegans ,Tubulin ,Cell Cycle Proteins ,Cytoskeletal Proteins ,Microtubule-Associated Proteins ,Caenorhabditis elegans Proteins ,Nuclear Proteins ,Protein Multimerization ,C. elegans ,NOCA-1 ,PTRN-1 ,Patronin ,cell biology ,developmental biology ,ninein ,non-centrosomal microtubule array ,stem cells ,Biochemistry and Cell Biology - Abstract
Non-centrosomal microtubule arrays assemble in differentiated tissues to perform mechanical and transport-based functions. In this study, we identify Caenorhabditis elegans NOCA-1 as a protein with homology to vertebrate ninein. NOCA-1 contributes to the assembly of non-centrosomal microtubule arrays in multiple tissues. In the larval epidermis, NOCA-1 functions redundantly with the minus end protection factor Patronin/PTRN-1 to assemble a circumferential microtubule array essential for worm growth and morphogenesis. Controlled degradation of a γ-tubulin complex subunit in this tissue revealed that γ-tubulin acts with NOCA-1 in parallel to Patronin/PTRN-1. In the germline, NOCA-1 and γ-tubulin co-localize at the cell surface, and inhibiting either leads to a microtubule assembly defect. γ-tubulin targets independently of NOCA-1, but NOCA-1 targeting requires γ-tubulin when a non-essential putatively palmitoylated cysteine is mutated. These results show that NOCA-1 acts with γ-tubulin to assemble non-centrosomal arrays in multiple tissues and highlight functional overlap between the ninein and Patronin protein families.
- Published
- 2015
14. DNA damage induces nuclear actin filament assembly by Formin -2 and Spire-½ that promotes efficient DNA repair. [corrected].
- Author
-
Belin, Brittany J, Lee, Terri, and Mullins, R Dyche
- Subjects
Cell Line ,Cell Nucleus ,Humans ,DNA Damage ,Microfilament Proteins ,Nuclear Proteins ,DNA Repair ,Protein Multimerization ,Actin Cytoskeleton ,DNA damage ,cell biology ,cytoskeleton ,human ,nuclear actin ,nuclear oxidation ,Formins ,Biochemistry and Cell Biology - Abstract
Actin filaments assemble inside the nucleus in response to multiple cellular perturbations, including heat shock, protein misfolding, integrin engagement, and serum stimulation. We find that DNA damage also generates nuclear actin filaments-detectable by phalloidin and live-cell actin probes-with three characteristic morphologies: (i) long, nucleoplasmic filaments; (ii) short, nucleolus-associated filaments; and (iii) dense, nucleoplasmic clusters. This DNA damage-induced nuclear actin assembly requires two biologically and physically linked nucleation factors: Formin-2 and Spire-1/Spire-2. Formin-2 accumulates in the nucleus after DNA damage, and depletion of either Formin-2 or actin's nuclear import factor, importin-9, increases the number of DNA double-strand breaks (DSBs), linking nuclear actin filaments to efficient DSB clearance. Nuclear actin filaments are also required for nuclear oxidation induced by acute genotoxic stress. Our results reveal a previously unknown role for nuclear actin filaments in DNA repair and identify the molecular mechanisms creating these nuclear filaments.
- Published
- 2015
15. Histones are exosome membrane proteins regulated by cell stress (Updated July 29, 2024).
- Abstract
Histones are nuclear proteins that play a role in regulating chromatin structure and gene expression. Recent research suggests that extracellular histones, which are found in biofluids, may contribute to various diseases when elevated. This study analyzed cell culture models and found that histones are secreted via extracellular vesicles (EVs) and the exosome pathway, particularly in response to cellular stress. The histones associated with EVs were found to lack extensive post-translational modifications, suggesting that these modifications may be involved in their trafficking or secretion. This discovery opens up avenues for further investigation into the biological activity of exosome-associated histones and their role in disease. [Extracted from the article]
- Published
- 2024
16. WITHDRAWN: Apelin-13 alleviate inflammatory reaction of ischemia reperfusion in rat kidney transplantation via NF-kappa B signaling pathway (Updated May 24, 2024).
- Abstract
This article, titled "WITHDRAWN: Apelin-13 alleviate inflammatory reaction of ischemia reperfusion in rat kidney transplantation via NF-kappa B signaling pathway," has been withdrawn due to fraudulent submission. The manuscript was falsely attributed to Prof. Gaetano La Manna of the University of Bologna, but there is no record of him being employed there. The department associated with the publication no longer exists, and the listed authors were not involved in the work. Therefore, this article should not be cited as a reference. [Extracted from the article]
- Published
- 2024
17. Targeted degradation via direct 26S proteasome recruitment
- Author
-
Charlene Bashore, Sumit Prakash, Matthew C. Johnson, Ryan J. Conrad, Ivy A. Kekessie, Suzie J. Scales, Noriko Ishisoko, Tracy Kleinheinz, Peter S. Liu, Nataliya Popovych, Aaron T. Wecksler, Lijuan Zhou, Christine Tam, Inna Zilberleyb, Rajini Srinivasan, Robert A. Blake, Aimin Song, Steven T. Staben, Yingnan Zhang, David Arnott, Wayne J. Fairbrother, Scott A. Foster, Ingrid E. Wertz, Claudio Ciferri, and Erin C. Dueber
- Subjects
Ligases ,Proteasome Endopeptidase Complex ,Ubiquitin-Protein Ligases ,Cryoelectron Microscopy ,Proteolysis ,Nuclear Proteins ,Cell Biology ,Molecular Biology ,Transcription Factors - Abstract
Engineered destruction of target proteins by recruitment to the cell’s degradation machinery has emerged as a promising strategy in drug discovery. The majority of molecules that facilitate targeted degradation do so via a select number of ubiquitin ligases, restricting this therapeutic approach to tissue types that express the requisite ligase. Here, we describe a new strategy of targeted protein degradation through direct substrate recruitment to the 26S proteasome. The proteolytic complex is essential and abundantly expressed in all cells; however, proteasomal ligands remain scarce. We identify potent peptidic macrocycles that bind directly to the 26S proteasome subunit PSMD2, with a 2.5-Å-resolution cryo-electron microscopy complex structure revealing a binding site near the 26S pore. Conjugation of this macrocycle to a potent BRD4 ligand enabled generation of chimeric molecules that effectively degrade BRD4 in cells, thus demonstrating that degradation via direct proteasomal recruitment is a viable strategy for targeted protein degradation.
- Published
- 2022
18. RanBP1 plays an essential role in directed migration of neural crest cells during development
- Author
-
Elias H. Barriga, Delan N. Alasaadi, Chiara Mencarelli, Roberto Mayor, and Franck Pichaud
- Subjects
Pregnancy ,Neural Crest ,Cell Movement ,Chemotaxis ,Humans ,Nuclear Proteins ,Female ,Cell Biology ,Molecular Biology ,Developmental Biology - Abstract
Collective cell migration is essential for embryonic development, tissue regeneration and repair, and has been implicated in pathological conditions such as cancer metastasis. It is, in part, directed by external cues that promote front-to-rear polarity in individual cells. However, our understanding of the pathways that underpin the directional movement of cells in response to external cues remains incomplete. To examine this issue we made use of neural crest cells (NC), which migrate as a collective during development to generate vital structures including bones and cartilage. Using a candidate approach, we found an essential role for Ran-binding protein 1 (RanBP1), a key effector of the nucleocytoplasmic transport pathway, in enabling directed migration of these cells. Our results indicate that RanBP1 is required for establishing front-to-rear polarity, so that NCs are able to chemotax. Moreover, our work suggests that RanBP1 function in chemotaxis involves the polarity kinase LKB1/PAR4. We envisage that regulated nuclear export of LKB1 through Ran/RanBP1 is a key regulatory step required for establishing front-to-rear polarity and thus chemotaxis, during NC collective migration.
- Published
- 2022
19. NOLC1 knockdown suppresses prostate cancer progressions by reducing AKT phosphorylation and β-catenin accumulation
- Author
-
Wansoo Kim, Dong-Yeop Yeo, Seong-Kyoon Choi, Hee-Yeon Kim, Seoung-Woo Lee, Janbolat Ashim, Jee Eun Han, Wookyung Yu, Hyohoon Jeong, Jin-Kyu Park, and Song Park
- Subjects
Male ,Prostate ,Biophysics ,Nuclear Proteins ,Androgen Antagonists ,Cell Biology ,Phosphoproteins ,Biochemistry ,Prostatic Neoplasms, Castration-Resistant ,Receptors, Androgen ,Cell Line, Tumor ,Humans ,Phosphorylation ,Proto-Oncogene Proteins c-akt ,Molecular Biology ,beta Catenin - Abstract
Although several studies have focused on cancer diagnosis and therapy, prostate cancer (PC) remains an intractable disease. Androgen deprivation therapy (ADT), which is used to treat early stage PC can lead to the development of castration-resistant prostate cancer (CRPC), which is highly associated with androgen receptor (AR) mutations. Nucleolar and coiled-body phosphoprotein 1 (NOLC1) is a chaperone that shuttles between the nucleus and the cytoplasm. Studies suggest that NOLC1 regulates PC progression; however, the underlying mechanisms remain unclear. Herein, we showed that NOLC1 knockdown suppresses PC cell proliferation by altering the signaling pathways and the expression of various proteins involved in DNA replication, amino acid metabolism, and RNA processing. Mechanistically, NOLC1 knockdown suppressed cell cycle progression by inhibiting AKT phosphorylation and β-catenin accumulation. Finally, we showed that NOLC1 expression is higher in human PC than in human hyperplastic prostate tissues. Altogether, we demonstrated that NOLC1 knockdown suppresses the progression of both AR-positive and AR-negative PC cells by inducing changes in the expression of several genes leading to cell cycle arrest. Thus, NOLC1 might be a novel and promising therapeutic target for PC.
- Published
- 2022
20. MTR4 adaptor PICT1 functions in two distinct steps during pre-rRNA processing
- Author
-
Sotaro Miyao, Kanako Saito, Renta Oshima, Kohichi Kawahara, and Masami Nagahama
- Subjects
Saccharomyces cerevisiae Proteins ,Exosome Multienzyme Ribonuclease Complex ,Tumor Suppressor Proteins ,Oligonucleotides ,Biophysics ,Nuclear Proteins ,Saccharomyces cerevisiae ,Cell Biology ,Biochemistry ,RNA, Ribosomal, 5.8S ,RNA Precursors ,Humans ,RNA Processing, Post-Transcriptional ,RNA, Small Interfering ,Molecular Biology ,RNA Helicases - Abstract
Ribosome biogenesis proceeds with the successive cleavage and trimming of the large 47S rRNA precursor, where the RNA exosome plays major roles in concert with the Ski2-like RNA helicase, MTR4. The recent finding of a consensus amino acid sequence, the arch-interacting motif (AIM), for binding to the arch domain in MTR4 suggests that recruitment of the RNA processing machinery to the maturing pre-rRNA at appropriate places and timings is mediated by several adaptor proteins possessing the AIM sequence. In yeast Saccharomyces cerevisiae, Nop53 plays such a role in the maturation of the 3'-end of 5.8S rRNA. Here, we investigated the functions of PICT1 (also known as GLTSCR2 or NOP53), a mammalian ortholog of Nop53, during ribosome biogenesis in human cells. PICT1 interacted with MTR4 and exosome in an AIM-dependent manner. Overexpression of PICT1 mutants defecting AIM sequence and siRNA-mediated depletion of PICT1 showed that PICT1 is involved in two distinct pre-rRNA processing steps during the generation of 60S ribosomes; first step is the early cleavage of 32S intermediate RNA, while the second step is the late maturation of 12S precursor into 5.8S rRNA. The recruitment of MTR4 and RNA exosome via the AIM sequence was required only during the late processing step. Although, the depletion of MTR4 and PICT1 induced stabilization of the tumor suppressor p53 protein in cancer cell lines, the depletion of the exosome catalytic subunits, RRP6 and DIS3, did not exert such an effect. These results suggest that recruitment of the RNA processing machinery to the 3'-end of pre-5.8S rRNA may be involved in the induction of the nucleolar stress response, but the pre-rRNA processing capabilities themselves were not involved in this process.
- Published
- 2022
21. Apelin-13 alleviate inflammatory reaction of ischemia reperfusion in rat kidney transplantation via NF-kappa B signaling pathway.
- Subjects
NF-kappa B ,REPERFUSION injury ,KIDNEY transplantation ,CELLULAR signal transduction ,REPERFUSION ,ISCHEMIA - Abstract
A preprint abstract from biorxiv.org discusses the use of apelin-13 in a rat kidney transplantation model to investigate its effects on ischemia-reperfusion injury (IRI)-related inflammation. The study found that apelin-13 reduced tissue damage, improved kidney function, and lowered levels of pro-inflammatory cytokines. Additionally, apelin-13 reduced the expression of proteins in the NF-kappa B signaling pathway, which is involved in inflammation. This research suggests that apelin-13 may be effective in reducing the inflammatory response to IRI following kidney transplantation. However, it is important to note that this preprint has not been peer-reviewed. [Extracted from the article]
- Published
- 2024
22. TPR is required for cytoplasmic chromatin fragment formation during senescence.
- Abstract
According to a preprint abstract, researchers have found that a protein called TPR, which is part of the nuclear pore complex basket, plays a role in the early activation of NF-{kappa}B signaling during oncogene-induced senescence. This activation occurs before the onset of the senescence-associated secretory phenotype (SASP) and does not affect NF-{kappa}B nuclear import. The researchers also discovered that TPR is involved in the formation of cytoplasmic chromatin fragments that lack nuclear pore components, suggesting a loss of structural integrity at the nuclear periphery during senescence. This research has not yet undergone peer review. [Extracted from the article]
- Published
- 2024
23. Exosomes are vehicles for the stress-regulated secretion of histones.
- Abstract
A preprint abstract from biorxiv.org discusses the secretion of histones, conserved nuclear proteins that regulate chromatin structure and gene expression. The study found that histones are localized to extracellular vehicles (EVs) and are secreted through the multivesicular body/exosome pathway. The secretion of histones via EVs is upregulated during cellular stress, and the histones lack extensive post-translational modifications compared to nuclear histones. This discovery suggests the need for further investigation into the biological activity of exosome-associated histones and their role in disease. Please note that this preprint has not been peer-reviewed. [Extracted from the article]
- Published
- 2024
24. NCAPH Stabilizes GEN1 in Chromatin to Resolve Ultra-Fine DNA Bridges and Maintain Chromosome Stability
- Author
-
Yuna Youn, Jae Hyeong Kim, and Jin-Hyeok Hwang
- Subjects
Chromosomal Instability ,Humans ,Nuclear Proteins ,Cell Cycle Proteins ,DNA ,Cell Biology ,General Medicine ,Molecular Biology ,Chromatin ,Chromosomes - Abstract
Repairing damaged DNA and removing all physical connections between sister chromosomes is important to ensure proper chromosomal segregation by contributing to chromosomal stability. Here, we show that the depletion of non-SMC condensin I complex subunit H (NCAPH) exacerbates chromosome segregation errors and cytokinesis failure owing to sister-chromatid intertwinement, which is distinct from the ultra-fine DNA bridges induced by DNA inter-strand crosslinks (DNA-ICLs). Importantly, we identified an interaction between NCAPH and GEN1 in the chromatin involving binding at the N-terminus of NCAPH. DNA-ICL activation, using ICL-inducing agents, increased the expression and interaction between NCAPH and GEN1 in the soluble nuclear and chromatin, indicating that the NCAPH-GEN1 interaction participates in repairing DNA damage. Moreover, NCAPH stabilizes GEN1 within chromatin at the G2/M-phase and is associated with DNA-ICL-induced damage repair. Therefore, NCAPH resolves DNA-ICL-induced ultra-fine DNA bridges by stabilizing GEN1 and ensures proper chromosome separation and chromosome structural stability.
- Published
- 2022
25. Prognostic impact of NPM1 and FLT3 mutations in patients with AML in first remission treated with oral azacitidine
- Author
-
Hartmut Döhner, Andrew H. Wei, Gail J. Roboz, Pau Montesinos, Felicitas R. Thol, Farhad Ravandi, Hervé Dombret, Kimmo Porkka, Irwindeep Sandhu, Barry Skikne, Wendy L. See, Manuel Ugidos, Alberto Risueño, Esther T. Chan, Anjan Thakurta, C.L. Beach, and Daniel Lopes de Menezes
- Subjects
Neoplasm, Residual ,Remission Induction ,Immunology ,Nuclear Proteins ,Cell Biology ,Hematology ,Protein-Tyrosine Kinases ,Prognosis ,Biochemistry ,Leukemia, Myeloid, Acute ,fms-Like Tyrosine Kinase 3 ,Recurrence ,Mutation ,Azacitidine ,Humans ,Nucleophosmin - Abstract
The randomized, placebo-controlled, phase 3 QUAZAR AML-001 trial (ClinicalTrials.gov identifier: NCT01757535) evaluated oral azacitidine (Oral-AZA) in patients with acute myeloid leukemia (AML) in first remission after intensive chemotherapy (IC) who were not candidates for hematopoietic stem cell transplantation. Eligible patients were randomized 1:1 to Oral-AZA 300 mg or placebo for 14 days per 28-day cycle. We evaluated relapse-free survival (RFS) and overall survival (OS) in patient subgroups defined by NPM1 and FLT3 mutational status at AML diagnosis and whether survival outcomes in these subgroups were influenced by presence of post-IC measurable residual disease (MRD). Gene mutations at diagnosis were collected from patient case report forms; MRD was determined centrally by multiparameter flow cytometry. Overall, 469 of 472 randomized patients (99.4%) had available mutational data; 137 patients (29.2%) had NPM1 mutations (NPM1mut), 66 patients (14.1%) had FLT3 mutations (FLT3mut; with internal tandem duplications [ITD], tyrosine kinase domain mutations [TKDmut], or both), and 30 patients (6.4%) had NPM1mut and FLT3-ITD at diagnosis. Among patients with NPM1mut, OS and RFS were improved with Oral-AZA by 37% (hazard ratio [HR], 0.63; 95% confidence interval [CI], 0.41-0.98) and 45% (HR, 0.55; 95% CI, 0.35-0.84), respectively, vs placebo. Median OS was improved numerically with Oral-AZA among patients with NPM1mut whether without MRD (48.6 months vs 31.4 months with placebo) or with MRD (46.1 months vs 10.0 months with placebo) post-IC. Among patients with FLT3mut, Oral-AZA improved OS and RFS by 37% (HR, 0.63; 95% CI, 0.35-1.12) and 49% (HR, 0.51; 95% CI, 0.27-0.95), respectively, vs placebo. Median OS with Oral-AZA vs placebo was 28.2 months vs 16.2 months, respectively, for patients with FLT3mut and without MRD and 24.0 months vs 8.0 months for patients with FLT3mut and MRD. In multivariate analyses, Oral-AZA significantly improved survival independent of NPM1 or FLT3 mutational status, cytogenetic risk, or post-IC MRD status.
- Published
- 2022
26. Structural basis for the recognition by 14-3-3 proteins of a conditional binding site within the oligomerization domain of human nucleophosmin
- Author
-
Anna A. Kapitonova, Kristina V. Tugaeva, Larisa A. Varfolomeeva, Konstantin M. Boyko, Richard B. Cooley, and Nikolai N. Sluchanko
- Subjects
Phosphopeptides ,Binding Sites ,14-3-3 Proteins ,Biophysics ,Humans ,Nuclear Proteins ,Cell Biology ,Nucleophosmin ,Molecular Biology ,Biochemistry - Abstract
Nucleophosmin 1 (NPM1) is a multifunctional protein regulating ribosome biogenesis, centrosome duplication and chromatin remodeling. Being a major nucleolar protein, NPM1 can migrate to the nucleus and the cytoplasm, which is controlled by changes of NPM1 oligomerization and interaction with other cell factors. NPM1 forms a stable pentamer with its N-terminal structured domain, where two nuclear export signals and several phosphorylation sites reside. This domain undergoes dissociation and disordering upon Ser48 phosphorylation in the subunit interface. Recent studies indicated that Ser48 is important for NPM1 interaction with other proteins including 14-3-3, the well-known phosphoserine/phosphothreonine binders, but the structural basis for 14-3-3/NPM1 interaction remained unaddressed. By fusing human 14-3-3ζ with an NPM1 segment surrounding Ser48, which was phosphorylated inside Escherichia coli cells by co-expressed protein kinase A, here we obtained the desired protein/phosphopeptide complex and determined its crystal structure. While biochemical data indicated that the interaction is driven by Ser48 phosphorylation, the crystallographic 14-3-3/phosphopeptide interface reveals an NPM1 conformation distinctly different from that in the NPM1 pentamer. Given the canonical phosphopeptide-binding mode observed in our crystal structure, Ser48 emerges as a conditional binding site whose recognition by 14-3-3 proteins is enabled by NPM1 phosphorylation, disassembly and disordering under physiological circumstances.
- Published
- 2022
27. An In Vivo Model for Elucidating the Role of an Erythroid-Specific Isoform of Nuclear Export Protein Exportin 7 (Xpo7) in Murine Erythropoiesis
- Author
-
Susree Modepalli, Sandra Martinez-Morilla, Srividhya Venkatesan, James Fasano, Katerina Paulsen, Dirk Görlich, Shilpa Hattangadi, and Gary M. Kupfer
- Subjects
Mice ,Cancer Research ,ran GTP-Binding Protein ,Active Transport, Cell Nucleus ,Genetics ,Animals ,Nuclear Proteins ,Protein Isoforms ,Erythropoiesis ,Cell Biology ,Hematology ,Karyopherins ,Molecular Biology - Abstract
Erythroid nuclear condensation is a complex process in which compaction to one-tenth its original size occurs in an active nucleus simultaneously undergoing transcription and cell division. We previously found that the nuclear exportin Exportin7 (Xpo7), which is erythroid- specific and highly induced during terminal erythropoiesis, facilitates nuclear condensation. We also identified a previously unannotated, erythroid-specific isoform of Xpo7 (Xpo7B) containing a novel first exon Xpo7-1b expressed only in late Ter119
- Published
- 2022
28. Excessive activation of HOXB13/PIMREG axis promotes hepatocellular carcinoma progression and drug resistance
- Author
-
Cui, Tang, Shixiong, Qiu, Wenying, Mou, Jinming, Xu, and Peijun, Wang
- Subjects
Homeodomain Proteins ,Carcinoma, Hepatocellular ,Liver Neoplasms ,Drug Resistance ,Intracellular Signaling Peptides and Proteins ,Biophysics ,Down-Regulation ,Nuclear Proteins ,Cell Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,Gene Expression Regulation, Neoplastic ,Cell Line, Tumor ,Humans ,Carrier Proteins ,Molecular Biology ,Cell Proliferation ,Transcription Factors - Abstract
The transcription factor HOXB13 is bound up with the occurrence, progression and drug fast of many kinds of cancer. Nevertheless, the specific molecular mechanism of HOXB13 in hepatocellular carcinoma (HCC) is still unknown. This provides an obstacle to the exploration of HCC treatments targeting HOXB13. This study found that HOXB13 was up-regulated in HCC tissues. HOXB13 enhanced the multiplication and metastasis of HCC cells. It enhanced HCC cell drug and anoikis resistance. The analysis of HCC RNA seq data indicated that the expression of HOXB13 and PIMREG were positively correlated. Luciferase report assay showed that HOXB13 could activate PIMREG promoter transcription. The results of RT-qPCR and western blot showed that HOXB13 regulated the transcription of PIMREG. Western blot proved that high expression of PIMREG participated in DNA damage repair and cell cycle regulation by up-regulating RAD51, BRCA1, CDC25A, CDC25B and CDC25C and down-regulating HIPK2. This led to a significant increase in DNA repair capacity, accelerated cell cycle progression, and insensitive to DNA damage. Down-regulation of PIMREG in Hep3B cells overexpressing HOXB13 attenuated the phenotype induced by HOXB13. Therefore, HOXB13 functioned through PIMREG instead of directly regulating the transcription of RAD51, BRCA1, CDC25A, CDC25B and CDC25C. The same results were obtained in vivo. It was concluded that HOXB13 affected the expression of cell cycle and DNA repair related factors by up-regulating the transcription of PIMREG, thereby promoting the progression of HCC and enhancing the resistance of HCC to chemotherapeutics.
- Published
- 2022
29. DSC2 Suppresses the Metastasis of Gastric Cancer through Inhibiting the BRD4/Snail Signaling Pathway and the Transcriptional Activity of β-Catenin
- Author
-
Chao Sun, Lei Wang, Dan-dan Du, Jian-bo Ji, Xiao-xia Yang, Bing-fang Yu, Peng-fei Shang, and Xiu-Li Guo
- Subjects
Desmocollins ,Aging ,Article Subject ,Nuclear Proteins ,Cell Cycle Proteins ,Cell Biology ,General Medicine ,Biochemistry ,Gene Expression Regulation, Neoplastic ,Mice ,Stomach Neoplasms ,Cell Line, Tumor ,Animals ,Humans ,beta Catenin ,Signal Transduction ,Transcription Factors - Abstract
Downregulated DSC2 involved in the metastasis of cancers. Unfortunately, its role on the development of gastric cancer (GC) and the potential mechanisms remain unclear. Bioinformatics analysis, Western blot, qRT-PCR, and immunohistochemistry were performed to detect the DSC2 levels of human GC and normal stomach tissues. The role of DSC2 and the downstream signaling in gastric carcinogenesis were explored by using GC specimens, GC cells with different DSC2 expression, inhibitors, and mouse metastasis models. We found that the level of DSC2 decreased significantly in GC tissues and cells. Recovered DSC2 inhibited the invasion and migration of GC cells both in culture and in xenografts. Mechanistically, DSC2 could not only decrease Snail level and nuclear BRD4 level by forming DSC2/BRD4, but also inhibit nuclear translocation of β-catenin. We concluded that DSC2 inhibited the metastasis of GC, and the underlying mechanisms were closely related to the regulation on nuclear translocation of BRD4 and β-catenin. Our results suggest that DSC2 may serve as a novel therapeutic target for GC.
- Published
- 2022
30. Understanding the interaction of 14-3-3 proteins with hDMX and hDM2
- Author
-
Srdanović, S, Wolter, M, Trinh, CH, Ottmann, C, Warriner, SL, Wilson, AJ, Chemical Biology, and ICMS Core
- Subjects
Nuclear Proteins ,Cell Cycle Proteins ,Proto-Oncogene Proteins c-mdm2 ,14-3-3 proteins ,Cell Biology ,Biochemistry ,Proto-Oncogene Proteins ,hDM2 and hDMX ,Humans ,structural biology ,Tumor Suppressor Protein p53 ,p53 pathway ,Molecular Biology ,Protein Binding - Abstract
p53 plays a critical role in regulating diverse biological processes: DNA repair, cell cycle arrest, apoptosis and senescence. The p53 pathway has therefore served as the focus of multiple drug-discovery efforts. p53 is negatively regulated by hDMX and hDM2; prior studies have identified 14-3-3 proteins as hDMX and hDM2 client proteins. 14-3-3 proteins are adaptor proteins that modulate localization, degradation and interactions of their targets in response to phosphorylation. Thus, 14-3-3 proteins may indirectly modulate the interaction between hDMX or hDM2 and p53 and represent potential targets for modulation of the p53 pathway. In this manuscript, we report on the biophysical and structural characterization of peptide/protein interactions that are representative of the interaction between 14-3-3 and hDMX or hDM2. The data establish that proximal phosphosites spaced ~20–25 residues apart in both hDMX and hDM2 co-operate to facilitate high-affinity 14-3-3 binding and provide structural insight that can be utilized in future stabilizer/inhibitor discovery efforts.
- Published
- 2022
31. Autophagy regulates rRNA synthesis
- Author
-
Yinfeng, Xu and Wei, Wan
- Subjects
Sirolimus ,RNA Polymerase I ,RNA, Ribosomal ,Sequestosome-1 Protein ,Autophagy ,Humans ,Nuclear Proteins ,Cell Biology ,Mechanistic Target of Rapamycin Complex 1 ,DNA, Ribosomal ,Transcription Factors - Abstract
Autophagy has emerged as a key regulator of cell metabolism. Recently, we have demonstrated that autophagy is involved in RNA metabolism by regulating ribosomal RNA (rRNA) synthesis. We found that autophagy-deficient cells display much higher 47S precursor rRNA level, which is caused by the accumulation of SQSTM1/p62 (sequestosome 1) but not other autophagy receptors. Mechanistically, SQSTM1 accumulation potentiates the activation of MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) signaling, which facilitates the assembly of RNA polymerase I pre-initiation complex at ribosomal DNA (rDNA) promoter regions and leads to the activation of rDNA transcription. Finally, we showed that SQSTM1 accumulation is responsible for the increase in protein synthesis, cell growth and cell proliferation in autophagy-deficient cells. Taken together, our findings reveal a regulatory role of autophagy and autophagy receptor SQSTM1 in rRNA synthesis and may provide novel mechanisms for the hyperactivated rDNA transcription in autophagy-related human diseases.
- Published
- 2022
32. LncRNA KCNQ1OT1 contributes to hydrogen peroxide‐induced apoptosis, inflammation, and oxidative stress of cardiomyocytes via miR‐130a‐3p/ZNF791 axis
- Author
-
Hong Xin, Chengliang Li, Tianzhi Cai, Jinlong Cao, and Meixue Wang
- Subjects
Inflammation ,MicroRNAs ,Oxidative Stress ,Myocardial Infarction ,Humans ,Nuclear Proteins ,Apoptosis ,Myocytes, Cardiac ,RNA, Long Noncoding ,Zinc Fingers ,Hydrogen Peroxide ,Cell Biology ,General Medicine - Abstract
It has been reported that long noncoding RNA (lncRNA) KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1) played an important role in myocardial infarction (MI). However, the regulatory network behind KCNQ1OT1 in MI is largely unknown. Quantitative real time polymerase chain reaction (qRT-PCR) was applied to detect the enrichment of KCNQ1OT1, microRNA-130a-3p (miR-130a-3p) and zinc finger 791 (ZNF791). The viability and apoptosis of AC16 cells were measured by (4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry. Enzyme-linked immunosorbent assay (ELISA) was conducted to assess the inflammation and oxidative stress status of AC16 cells. The targeted relationship between miR-130a-3p and KCNQ1OT1 or ZNF791 was predicted by StarBase bioinformatic database, and dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were carried out to verify these predictions. Hydrogen peroxide (H
- Published
- 2022
33. The <scp>BRCA2</scp> and <scp>CDKN1A</scp> ‐interacting protein ( <scp>BCCIP</scp> ) stabilizes stalled replication forks and prevents degradation of nascent <scp>DNA</scp>
- Author
-
Bhawna Singh, Shalini Roy Chowdhury, Mohammad Shoab Mansuri, Saraswathi Jayarajan Pillai, and Sonam Mehrotra
- Subjects
BRCA2 Protein ,Cyclin-Dependent Kinase Inhibitor p21 ,DNA Replication ,MRE11 Homologue Protein ,DNA Repair ,Calcium-Binding Proteins ,Biophysics ,Nuclear Proteins ,Cell Cycle Proteins ,DNA ,Cell Biology ,Biochemistry ,Genomic Instability ,Structural Biology ,Genetics ,Humans ,DNA Breaks, Double-Stranded ,Rad51 Recombinase ,Molecular Biology ,Transcription Factors - Abstract
DNA replication stress is characterized by impaired replication fork progression, causing some of the replication forks to collapse and form DNA breaks. It is a primary cause of genomic instability leading to oncogenic transformation. The repair-independent functions of the proteins RAD51 and BRCA2, which are involved in homologous recombination (HR)-mediated DNA repair, are crucial for protecting nascent DNA strands from nuclease-mediated degradation. The BRCA2 and CDKN1A-interacting protein (BCCIP) associates with BRCA2 and RAD51 during HR-mediated DNA repair. Here, we investigated the role of BCCIP during the replication stress response. We find that in the presence of replication stress, BCCIP deficiency increases replication fork stalling and results in DNA double-strand break formation. We show that BCCIP is recruited to stalled replication forks and prevents MRE11 nuclease-mediated degradation of nascent DNA strands.
- Published
- 2022
34. RhoGDI1 interacts with PHLDA2, suppresses the proliferation, migration, and invasion of trophoblast cells, and participates in the pathogenesis of preeclampsia
- Author
-
Guiyu Song and Feng Jin
- Subjects
rho Guanine Nucleotide Dissociation Inhibitor alpha ,Cancer Research ,Pre-Eclampsia ,Cell Movement ,Pregnancy ,Placenta ,Humans ,Nuclear Proteins ,Female ,Cell Biology ,Cell Proliferation ,Trophoblasts - Abstract
Preeclampsia (PE) is a pregnancy-associated disease, which is the major cause of mortality on maternity and perinatal infants. It is hypothesized that PE is a consequence of the dysfunction of the trophoblast cells. Pleckstrin homology-like domain, family A, member 2 (PHLDA2) was shown to inhibit the proliferation, migration, and invasion of trophoblast cells in our previous studies. However, the mechanism by which PHLDA2 affects trophoblast cell function has not been clarified. In the current study, co-immunoprecipitation (Co-IP) with mass spectroscopy analysis was used to explore the proteins that interacted with PHLDA2. A total of 291 candidate proteins were found to be associated with PHLDA2. The interaction between PHLDA2 and Rho guanine nucleotide dissociation inhibitor (RhoGDI) 1 was identified by Co-IP and immunofluorescence staining. Western blot analysis indicated that overexpression of PHLDA2 resulted in upregulation of the RhoGDI1 protein levels, which were stabilized in the presence of cycloheximide. Similarly, overexpression of RhoGDI1 promoted PHLDA2 expression and its stability. Furthermore, pull-down and Co-IP results indicated that PHLDA2 repressed the activity of Rho guanosine triphosphate hydrolase family proteins by regulating RhoGDI1 expression. In addition, RhoGDI1 expression was upregulated in the placental tissues of patients with PE. The effects of the suppression of PHLDA2 expression on proliferation, migration, and invasion of trophoblast cells were partly abrogated following knockdown of RhoGDI1. Taken together, the data indicated that RhoGDI1 mediated regulation of PHLDA2 on the biological behavior of trophoblast cells and may participate in the pathophysiology of PE.
- Published
- 2022
35. E3 ubiquitin ligase RBX1 drives the metastasis of triple negative breast cancer through a FBXO45-TWIST1-dependent degradation mechanism
- Author
-
Jun, Shao, Qian, Feng, Weifan, Jiang, Yuting, Yang, Zhiqiang, Liu, Liang, Li, Wenlong, Yang, and Yufeng, Zou
- Subjects
Gene Expression Regulation, Neoplastic ,Aging ,Epithelial-Mesenchymal Transition ,Cell Line, Tumor ,F-Box Proteins ,Ubiquitin-Protein Ligases ,Twist-Related Protein 1 ,Humans ,Nuclear Proteins ,Triple Negative Breast Neoplasms ,Cell Biology ,Carrier Proteins - Abstract
Triple-negative breast cancer (TNBC) patients are at high risk of recurrence and metastasis in the early stages, although receiving standard treatment. However, the underlying mechanism of TNBC remains unclear. Here, the critical effect of E3 ubiquitin ligase RBX1 in the metastasis of TNBC was reported for the first time. We discovered that RBX1 expression was evidently raised in the tissues of TNBC. Our clinical research displayed that high RBX1 expression was markedly related to poor distant invasion and survival. Functional analysis exhibited that RBX1 facilitated metastasis of TNBC cells through increasing EMT. Furthermore, we demonstrated that RBX1 knockdown increased the levels of the Twist family bHLH transcription factor 1 (TWIST1), is a significant regulator in the EMT process in some cancers. It can be observed an evident positive correlation between the TWIST1 and RBX1 levels, further confirming that EMT induced by RBX1 in TNBC cells is determined by TWIST1. Mechanistically, RBX1 modulates the expression of TWIST1 via modulating FBXO45, directly binding to FBXO45, and facilitating its degradation and ubiquitination. Briefly, our findings confirm that RBX1 is probably a new biomarker of TNBC carcinogenesis, thus suggesting that targeting the RBX1/FBXO45/TWIST1 axis may be an underlying strategy for TNBC treatment.
- Published
- 2022
36. SUMO-specific protease SENP3 enhances MDM2-mediated ubiquitination of PARIS/ZNF746 in HeLa cells
- Author
-
Tamotsu Nishida
- Subjects
Ubiquitin-Protein Ligases ,Ubiquitination ,Biophysics ,Nuclear Proteins ,Neurodegenerative Diseases ,Proto-Oncogene Proteins c-mdm2 ,Cell Biology ,Biochemistry ,Repressor Proteins ,Cysteine Endopeptidases ,Phosphatidylinositol 3-Kinases ,Endopeptidases ,Humans ,Molecular Biology ,HeLa Cells ,Peptide Hydrolases ,Transcription Factors - Abstract
The transcriptional repressor PARIS, a substrate of the ubiquitin E3 ligase parkin, represses the expression of the transcriptional co-activator, PGC-1α gene, and is involved in several pathological processes, including neurodegenerative disease and cancers. We have previously shown that SUMOylation of PARIS play an important role in its transcriptional repression activity. In addition, RNF4-mediated ubiquitination of SUMO2/3-conjugated PARIS is required for the control of PARIS-mediated transcriptional repression in HeLa cells that lack parkin expression. However, little is known about how PARIS ubiquitination and degradation are regulated in parkin-deficient cells. Here, we report that the deSUMOylase SENP3 interacted with PARIS and enhanced the ubiquitination of PARIS independently of its SUMOylation in HeLa cells. SENP3-enhanced PARIS ubiquitination mainly contributed to its proteasomal degradation, and required the oncogenic E3 ubiquitin ligase MDM2. MDM2 knockdown by small interfering RNA or expression of a dominant-negative MDM2 mutant inhibited the ubiquitination of PARIS. We further found that MDM2 activation via the PI3K/AKT pathway was involved in PARIS ubiquitination. Taken together, these results suggest that PARIS ubiquitination through SENP3-mediated MDM2 activation may control its functions in parkin-deficient cells.
- Published
- 2022
37. Exosomes from human adipose-derived mesenchymal stromal/stem cells accelerate angiogenesis in wound healing: implication of the EGR-1/lncRNA-SENCR/DKC1/VEGF-A axis
- Author
-
Yang, Sun, Yikun, Ju, and Bairong, Fang
- Subjects
Vascular Endothelial Growth Factor A ,Wound Healing ,Cancer Research ,Neovascularization, Physiologic ,Nuclear Proteins ,Cell Cycle Proteins ,Mesenchymal Stem Cells ,Cell Biology ,Exosomes ,Mice ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,RNA, Long Noncoding ,Cell Proliferation - Abstract
Exosomes (Exos) extracted from human adipose mesenchymal stromal/stem cells (hAD-MSCs) have been reported as therapeutic tools for tissue repair, but how they regulate angiogenesis of endothelial cells remains unknown. In this study, hAD-MSCs were isolated, and early growth response factor-1, Smooth muscle and endothelial cell enriched migration/differentiation-associated long-noncoding RNA (lncRNA-SENCR), and vascular endothelial growth factor-A (VEGF-A) overexpression or knockdown was achieved. Exos extracted from hAD-MSCs (hADSC-Exos) were co-cultured with human umbilical vein endothelial cells (HUVECs) to detect the effects of EGR-1, lncRNA-SENCR, and VEGF-A on angiogenesis and the relationships between EGR-1, lncRNA-SENCR, Dyskerin pseudouridine synthase 1 (DKC1), and VEGF-A. An in vivo experiment verified the effect of hADSC-Exos on the wound healing process. hADSC-Exos substantially promoted the proliferation, migration, and angiogenesis of HUVECs, which could be reversed by short-hairpin RNA SENCR (shSENCR) transfection. hADSC-Exos had elevated expression of EGR-1, which bound to the lncRNA-SENCR promoter. The suppressive effect of Exo-shEGR1 on HUVECs was counteracted by SENCR overexpression. LncRNA-SENCR was shown to interact with DKC1. Overexpression of DKC1 or lncRNA-SENCR maintained stable VEGF-A expression. Overexpression of VEGF-A reversed the suppressive effect of shSENCR on HUVECs. Consistent results were obtained in mice in vivo. Overall, hADSC-Exo EGR-1 upregulates lncRNA-SENCR expression to activate the DKC1/VEGF-A axis, facilitating the wound-healing process by increasing angiogenesis.
- Published
- 2022
38. Mice lacking DCAF2 in placenta die at the gastrulation stage
- Author
-
Man Yang, Mei Liu, Zongting Wang, and Cong Zhang
- Subjects
Mice, Knockout ,Mice ,Histology ,Pregnancy ,Placenta ,Gastrulation ,Animals ,Nuclear Proteins ,Cell Cycle Proteins ,Female ,Cell Biology ,Trophoblasts ,Pathology and Forensic Medicine - Abstract
UV-damaged DNA-binding protein 1 (DDB1) and cullin 4-associated factor 2 (DCAF2, also known as DTL or CDT2) is an evolutionarily highly conserved substrate recognition factor in the cullin 4 RING E3 ubiquitin ligase (CRL4) complex. This complex degrades multiple DNA replication and cell cycle-associated proteins to maintain genome stability. To clarify the function of DCAF2 in vivo, we used Cre recombinase driven by the Elf5 promoter to generate knockout mouse model that was specifically deleted Dcaf2 in the trophoblast lineage (Elf5-Cre; Dcaf2
- Published
- 2022
39. Nuclear envelope-localized torsinA-LAP1 complex regulates hepatic VLDL secretion and steatosis.
- Author
-
Ji-Yeon Shin, Hernandez-Ono, Antonio, Fedotova, Tatyana, Östlund, Cecilia, Lee, Michael J., Gibeley, Sarah B., Chun-Chi Liang, Dauer, William T., Ginsberg, Henry N., Worman, Howard J., Shin, Ji-Yeon, and Liang, Chun-Chi
- Subjects
- *
FATTY liver , *SECRETION , *NUCLEAR membranes , *METABOLIC regulation , *NUCLEAR proteins , *ANIMALS , *CARRIER proteins , *CELL membranes , *EPITHELIAL cells , *GENETIC disorders , *LIPID metabolism disorders , *LIPOPROTEINS , *MEMBRANE proteins , *MICE , *MOLECULAR chaperones - Abstract
Deciphering novel pathways regulating liver lipid content has profound implications for understanding the pathophysiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Recent evidence suggests that the nuclear envelope is a site of regulation of lipid metabolism but there is limited appreciation of the responsible mechanisms and molecular components within this organelle. We showed that conditional hepatocyte deletion of the inner nuclear membrane protein lamina-associated polypeptide 1 (LAP1) caused defective VLDL secretion and steatosis, including intranuclear lipid accumulation. LAP1 binds to and activates torsinA, an AAA+ ATPase that resides in the perinuclear space and continuous main ER. Deletion of torsinA from mouse hepatocytes caused even greater reductions in VLDL secretion and profound steatosis. Both of these mutant mouse lines developed hepatic steatosis and subsequent steatohepatitis on a regular chow diet in the absence of whole-body insulin resistance or obesity. Our results establish an essential role for the nuclear envelope-localized torsinA-LAP1 complex in hepatic VLDL secretion and suggest that the torsinA pathway participates in the pathophysiology of nonalcoholic fatty liver disease. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
40. The Jembrana disease virus Rev protein: Identification of nuclear and novel lentiviral nucleolar localization and nuclear export signals.
- Author
-
Marchand, Claude, Lemay, Guy, and Archambault, Denis
- Subjects
- *
PROTEOMICS , *VIRAL proteins , *VIRUS diseases , *NUCLEAR nonproliferation , *NUCLEAR proteins , *NUCLEOPHOSMIN - Abstract
The lentiviral Rev protein, which is a regulatory protein essential for virus replication, has been first studied in the human immunodeficiency virus type 1 (HIV-1). The main function of Rev is to mediate the nuclear exportation of viral RNAs. To fulfill its function, Rev shuttles between the cytoplasm and the nucleus. The Jembrana disease virus (JDV), a lentivirus, is the etiologic agent of the Jembrana disease which was first described in Bali cattle in Indonesia in 1964. Despite the high mortality rate associated with JDV, this virus remains poorly studied. Herein the subcellular distribution of JDV Rev, the nuclear and nucleolar localization signals (NLS and NoLS, respectively) and the nuclear export signal (NES) of the protein were examined. JDV Rev fused to the enhanced green fluorescent protein (EGFP) predominantly localized to the cytoplasm and nucleolus of transfected cells, as determined by fluorescence microscopy analyses. Through transfection of a series of deletion mutants of JDV Rev, it was possible to localize the NLS/NoLS region between amino acids (aa) 74 to 105. By substituting basic residues with alanine within this sequence, we demonstrated that the JDV Rev NLS encompasses aa 76 to 86, and is exclusively composed of arginine residues, whereas a bipartite NoLS was observed for the first time in any retroviral Rev/Rev-like proteins. Finally, a NES was identified downstream of the NLS/NoLS and encompasses aa 116 to 128 of the JDV Rev protein. The JDV Rev NES was found to be of the protein kinase A inhibitor (PKI) class instead of the HIV-1 Rev class. It also corresponds to the most optimal consensus sequence of PKI NES and, as such, is novel among lentiviral Rev NES. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
41. The implication of the crosstalk of Nrf2 with NOXs, and HMGB1 in ethanol-induced gastric ulcer: Potential protective effect is afforded by Raspberry Ketone.
- Author
-
Badr, Amira M., EL- Orabi, Naglaa F., and Ali, Rehab A.
- Subjects
- *
KETONES , *ULCERS , *CROSSTALK , *RASPBERRIES , *NUCLEAR proteins , *OMEPRAZOLE - Abstract
Ethanol consumption is one of the common causative agents implicated in gastric ulcer development. Oxidative stress plays a major role in the induction and development of gastric ulceration. NADPH oxidases (NOXs) and Nuclear factor erythroid 2-related factor 2 (Nrf2) are key players in ethanol-induced ulcers. High-mobility group box 1 (HMGB1), a ubiquitous nuclear protein, mediates various inflammation functions. However, the role of HMGB1 in ethanol-induced gastric ulcer is not yet elucidated. Raspberry Ketone (RK) is a natural phenolic compound with antioxidant and anti-inflammatory properties. In the present study, absolute ethanol (7.5 ml/kg) was used to induce gastric ulceration in rats. Raspberry Ketone (RK) (50 mg/kg) was given orally one hour before the administration of absolute ethanol. Interestingly, ethanol-induced gastric ulcer was associated with Nrf2 downregulation, which was correlated with NOX-1, 2 NOX-4, and HMGB1 upregulation, and was significantly reversed by RK pre-treatment. RK pre-treatment provided 80% gastroprotection. Gastroprotective properties of RK were mediated via antioxidant, anti-inflammatory (suppression of NF-kB and tumor necrosis factor-α), and antiapoptotic activities (reduction of Bax/Bcl2 ratio). Gastroprotective properties of RK were confirmed by histopathological examination. In conclusion, this study is the first to provide evidence to the role of HMGB1 in ethanol-induced gastric ulcer, and the crosstalk of Nrf2, NOXs and HMGB1. It also demonstrates that RK represents a promising gastroprotective activity comparable to omeprazole. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
42. Normal B cell development and Pax5 expression in Thy28/ThyN1-deficient mice.
- Author
-
Kitaura, Fusako, Yuno, Miyuki, Fujita, Toshitsugu, Wakana, Shigeharu, Ueda, Jun, Yamagata, Kazuo, and Fujii, Hodaka
- Subjects
- *
B cells , *NUCLEAR proteins , *LEUCOCYTES , *IMMUNOGLOBULIN producing cells , *TRANSCRIPTION factors , *MICE - Abstract
Thy28, also known as ThyN1, is a highly conserved nuclear protein. We previously showed that in a chicken mature B cell line, Thy28 binds to the promoter of the gene encoding Pax5, a transcription factor essential for B cell development, and positively regulates its expression. Here, we generated a Thy28-deficient mouse line to analyze its potential role in B cell development in mice. Thy28-deficient mice showed normal development of B cells, and the expression of Pax5 was comparable between wild-type and Thy28-deficient primary B cells. Thus, species-specific mechanisms regulate Pax5 expression and B cell development. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
43. Curcumin attenuates oxidative stress in RAW264.7 cells by increasing the activity of antioxidant enzymes and activating the Nrf2-Keap1 pathway.
- Author
-
Lin, Xinyu, Bai, Dingping, Wei, Zixi, Zhang, Ying, Huang, Yifan, Deng, Hui, and Huang, Xiaohong
- Subjects
- *
CURCUMIN , *OXIDATIVE stress , *NUCLEAR proteins , *WESTERN immunoblotting , *ENZYMES - Abstract
Large-scale breeding environments often lead to oxidative stress. Macrophages play an important role in the immune system and are vulnerable to reactive oxygen species (ROS), which result in macrophage death. Curcumin is the main active component of turmeric and exerts antioxidant effects. Here, we measured the activity of some antioxidant enzymes and chose the Nrf2-Keap1 signaling pathway to study the protective effects of curcumin on macrophages under oxidative stress in vitro. We used RAW264.7 cells as a research model, and oxidative damage was induced by hydrogen peroxide (H2O2). Cell viability was measured by an MTT assay. Flow cytometry was used to measure cellular ROS and apoptosis. The effect of curcumin on Nrf2-Keap1 signaling pathway-related genes was analyzed by qRT-PCR. Furthermore, the translocation of Nrf2 protein was also investigated by Western blot analysis of total and nuclear proteins. All curcumin-treated groups exhibited increased activity of catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX). Low- and middle-dose curcumin decreased malondialdehyde (MDA) and ROS levels, but high-dose curcumin increased MDA and ROS production. We found that low-dose curcumin protected cells from apoptosis, while apoptosis in the middle- and high-dose curcumin-treated groups were stagnant in the early stage. Furthermore, middle-dose curcumin upregulated Nrf2 expression after H2O2 treatment for 4 h. Low- and middle-dose curcumin could activate Nrf2 and promote it to migrate into nuclei. The translocation of Nrf2 to the nucleus to upregulate the expression of haemoxygenase-1 (HO-1) was promoted in the low- and middle-dose curcumin-treated groups. The middle-dose curcumin-treated group also exhibited enhanced expression of glutamate-cysteine ligase, a modifier subunit (GLCM), but inhibited transcription of glutamate-cysteine ligase, a catalytic subunit (GCLC). Curcumin resisted oxidants by increasing the activity of antioxidant enzymes and activating the Nrf2-Keap1 pathway, which could potentially promote cell survival. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
44. Conserved motifs in nuclear genes encoding predicted mitochondrial proteins in Trypanosoma cruzi.
- Author
-
Becco, Lorena, Smircich, Pablo, and Garat, Beatriz
- Subjects
- *
MITOCHONDRIAL proteins , *PLANT mitochondria , *TRYPANOSOMA cruzi , *CHAGAS' disease , *MITOCHONDRIAL DNA , *NUCLEAR proteins - Abstract
Trypanosoma cruzi, the protozoan parasite that causes Chagas’ disease, exhibits peculiar biological features. Among them, the presence of a unique mitochondrion is remarkable. Even though the mitochondrial DNA constitutes up to 25% of total cellular DNA, the structure and functionality of the mitochondrion are dependent on the expression of the nuclear genome. As in other eukaryotes, specific peptide signals have been proposed to drive the mitochondrial localization of a subset of trypanosomatid proteins. However, there are mitochondrial proteins encoded in the nuclear genome that lack of a peptide signal. In other eukaryotes, alternative protein targeting to subcellular organelles via mRNA localization has also been recognized and specific mRNA localization towards the mitochondria has been described. With the aim of seeking for mitochondrial localization signals in T. cruzi, we developed a strategy to build a comprehensive database of nuclear genes encoding predicted mitochondrial proteins (MiNT) in the TriTryps (T. cruzi, T. brucei and L. major). We found that approximately 15% of their nuclear genome encodes mitochondrial products. In T. cruzi the MiNT database reaches 1438 genes and a conserved peptide signal, M(L/F) R (R/S) SS, named TryM-TaPe is found in 60% of these genes, suggesting that the canonical mRNA guidance mechanism is present. In addition, the search for compositional signals in the transcripts of T. cruzi MiNT genes produce a list, being worth to note a conserved non-translated element represented by the consensus sequence DARRVSG. Taking into account its reported interaction with the T. brucei TRRM3 protein which is enriched in the mitochondrial membrane fraction, we here suggest a putative zip code role for this element. Globally, here we provide an inventory of the mitochondrial proteins in T. cruzi and give evidence for the existence of both peptide and mRNA signals specific to nuclear encoded mitochondrial proteins. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
45. Towards an understanding of C9orf82 protein/CAAP1 function.
- Author
-
Aslam, Muhammad Assad, Alemdehy, Mir Farshid, Pritchard, Colin E. J., Song, Ji-Ying, Muhaimin, Fitriari Izzatunnisa, Wijdeven, Ruud H., Huijbers, Ivo J., Neefjes, Jacques, and Jacobs, Heinz
- Subjects
- *
NUCLEAR proteins , *APOPTOSIS , *DNA topoisomerase II , *PROTEIN expression , *DRUG resistance , *DOXORUBICIN - Abstract
C9orf82 protein, or conserved anti-apoptotic protein 1 or caspase activity and apoptosis inhibitor 1 (CAAP1) has been implicated as a negative regulator of the intrinsic apoptosis pathway by modulating caspase expression and activity. In contrast, an independent genome wide screen for factors capable of driving drug resistance to the topoisomerase II (Topo II) poisons doxorubicin and etoposide, implicated a role for the nuclear protein C9orf82 in delaying DSBs repair downstream of Topo II, hereby sensitizing cells to DSB induced apoptosis. To determine its function in a genetically defined setting in vivo and ex vivo, we here employed CRISPR/Cas9 technology in zygotes to generate a C9orf82 knockout mouse model. C9orf82ko/ko mice were born at a Mendelian ratio and did not display any overt macroscopic or histological abnormalities. DSBs repair dependent processes like lymphocyte development and class switch recombination (CSR) appeared normal, arguing against a link between the C9orf82 encoded protein and V(D)J recombination or CSR. Most relevant, primary pre-B cell cultures and Tp53 transformed mouse embryo fibroblasts (MEFs) derived from C9orf82ko/ko E14.5 and wild type embryos displayed comparable sensitivity to a number of DNA lesions, including DSBs breaks induced by the topoisomerase II inhibitors, etoposide and doxorubicin. Likewise, the kinetics of γH2AX formation and resolution in response to etoposide of C9orf82 protein proficient, deficient and overexpressing MEFs were indistinguishable. These data argue against a direct role of C9orf82 protein in delaying repair of Topo II generated DSBs and regulating apoptosis. The genetically defined systems generated in this study will be of value to determine the actual function of C9orf82 protein. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
46. BRD4 inhibitor MZ1 exerts anti-cancer effects by targeting MYCN and MAPK signaling in neuroblastoma
- Author
-
Xianbing, Zhang, Xinyi, Guo, Ran, Zhuo, Yanfang, Tao, Wenxia, Liang, Randong, Yang, Yanling, Chen, Haibo, Cao, Siqi, Jia, Juanjuan, Yu, Xinmei, Liao, Xiaolu, Li, Fang, Fang, Gen, Li, Di, Wu, Yunyun, Xu, Zhiheng, Li, Jian, Pan, and Jian, Wang
- Subjects
N-Myc Proto-Oncogene Protein ,Biophysics ,Nuclear Proteins ,Cell Cycle Proteins ,Dipeptides ,Cell Biology ,Biochemistry ,Gene Expression Regulation, Neoplastic ,Neuroblastoma ,Cell Line, Tumor ,Humans ,Child ,Heterocyclic Compounds, 3-Ring ,Molecular Biology ,Transcription Factors - Abstract
Neuroblastoma(NB) is a common childhood solid tumor, and most patients in the high-risk group with MYCN gene amplification have a poor prognosis. Inhibition of bromodomain and extra terminal (BET) proteins has shown considerable promise in the investigation of MYCN-driven malignancies in recent years. MZ1 is a novel BET inhibitor that employs proteolytic-targeting chimera (PROTAC) technology for proteasomal degradation of target proteins and has shown excellent effects in some tumors, but its role in neuroblastoma remains poorly understood. Herein, we observed that MZ1 suppressed MYC-amplified NB cell proliferation and normal cell cycle, while simultaneously boosting cell apoptosis. MZ1 also provides a significant therapeutic impact in vivo. Mechanistically, MZ1 exhibits anti-tumor effect in NB cells by suppressing the expression of N-Myc or C-Myc as well as the MAPK signaling pathway. Overall, our data imply that MZ1 might be exploited as a possible therapeutic method for NB therapy.
- Published
- 2022
47. Mutations equivalent to Drosophila mago nashi mutants imply reduction of Magoh protein incorporation into exon junction complex
- Author
-
Saya Oshizuki, Eri Matsumoto, Satoshi Tanaka, and Naoyuki Kataoka
- Subjects
RNA Splicing ,Mutation ,Genetics ,Animals ,Nuclear Proteins ,Drosophila ,Exons ,RNA, Messenger ,Cell Biology - Abstract
Pre-mRNA splicing imprints mRNAs by depositing multi-protein complexes, termed exon junction complexes (EJCs). The EJC core consists of four proteins, eIF4AIII, MLN51, Y14 and Magoh. Magoh is a human homolog of Drosophila mago nashi protein, which is involved in oskar mRNA localization in Drosophila oocytes. Here we determined the effects of Magoh mutations equivalent to those of Drosophila mago nashi mutant proteins that cause mis-localization of oskar mRNA. We found that Magoh I90T mutation caused mis-localization of Magoh protein in the cytoplasm by reducing its binding activity to Y14. On the other hand, G18R mutation did not affect its binding to Y14, but this mutation reduced its association with spliced mRNAs. Our results strongly suggest that Magoh mutations equivalent to Drosophila mago nashi mutants cause improper EJC formation by reducing incorporation of Magoh into EJC.
- Published
- 2022
48. Impact of FLT3 internal tandem duplication and NPM1 mutations in acute myeloid leukemia treated with allogeneic hematopoietic cell transplantation
- Author
-
Mary Lynn Savoie, Jan Storek, Megan Kinzel, Mona Shafey, Rehan M. Faridi, Faisal Khan, Rutvij A. Khanolkar, and Kareem Jamani
- Subjects
Adult ,Oncology ,FLT3 Internal Tandem Duplication ,Cancer Research ,medicine.medical_specialty ,NPM1 ,medicine.medical_treatment ,Immunology ,medicine.disease_cause ,Recurrence ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,Cumulative incidence ,Genetics (clinical) ,Transplantation ,Mutation ,Chemotherapy ,business.industry ,Proportional hazards model ,Hematopoietic Stem Cell Transplantation ,Nuclear Proteins ,Myeloid leukemia ,Cell Biology ,Leukemia, Myeloid, Acute ,fms-Like Tyrosine Kinase 3 ,business ,Nucleophosmin - Abstract
Background aims The internal tandem duplication of FLT3 (FLT3ITD) and NPM1 mutations (NPM1mut) are well-established prognostic factors in cytogenetically intermediate-risk acute myeloid leukemia (AML) when treated with chemotherapy alone. However, their prognostic value in the setting of allogeneic hematopoietic cell transplantation (HCT) is controversial. Methods FLT3 and NPM1 mutational status was determined at diagnosis using single-gene polymerase chain reaction or next-generation sequencing in 247 adult patients with cytogenetically intermediate-risk AML who underwent myeloablative HCT. Multivariate Fine–Gray and Cox regression was used to analyze the cumulative incidence of relapse (CIR), relapse-free survival (RFS) and overall survival (OS). Results FLT3ITD and NPM1mut were present in 74 of 247 (30%) and 79 of 247 (32%) patients, respectively. There was no significant difference between patients without a FLT3ITD or NPM1mut (FLT3NONITD/NPM1WT) and patients with a FLT3ITD mutation alone (FLT3ITD/NPM1WT) with regard to CIR (P = 0.60), RFS (P = 0.91) or OS (P = 0.66). Similarly, there was no significant difference between FLT3NONITD/NPM1WT and FLT3NONITD/NPM1mut patients with regard to CIR (P = 0.70), RFS (P = 0.75) or OS (P = 0.95). The presence of a concurrent mutation in NPM1 did not appear to modify the impact of having a FLT3ITD mutation. Conclusions In contrast to chemotherapy-only treatment, FLT3 and NPM1 mutational status does not appear to predict outcomes in patients with cytogenetically intermediate-risk AML following HCT. These results suggest that HCT may ameliorate the poor prognostic effect of FLT3ITD mutation and that HCT should be considered over chemotherapy-only treatment in FLT3ITD-mutated AML.
- Published
- 2022
49. BRD4 promotes the migration and invasion of bladder cancer cells through the Sonic hedgehog signaling pathway and enhances cisplatin resistance
- Author
-
Tiantian, Liu, Ze, Zhang, Chong, Wang, Houbao, Huang, and Yawei, Li
- Subjects
Nuclear Proteins ,Cell Cycle Proteins ,Cell Biology ,Biochemistry ,Urinary Bladder Neoplasms ,Cell Movement ,Cell Line, Tumor ,Humans ,Hedgehog Proteins ,Cisplatin ,skin and connective tissue diseases ,Molecular Biology ,Cell Proliferation ,Transcription Factors - Abstract
Platinum-based chemotherapy is a widely used strategy for bladder cancer (BCa) treatment. However, its clinical efficacy is affected by chemotherapy resistance via complex molecular mechanisms. Therefore, there is an urgent need to explore new targets for BCa therapy. Here, we showed that bromodomain-4 protein (BRD4) expression is upregulated in BCa tissues and cells. Inhibition of BRD4 attenuated the migration and invasion of BCa cells, which was rescued by the Sonic hedgehog (SHH) pathway activator recombinant human Sonic hedgehog peptide (rhSHH). We further found that cisplatin (DDP) suppressed the migration and invasion of BCa cells in vitro and inhibited tumor growth in vivo. However, overexpression of BRD4 weakened the pharmacological effects of DDP. In brief, our research revealed that BRD4 promotes migration and invasion by positively regulating the SHH pathway, drives DDP resistance in BCa, and is a novel therapeutic target for the treatment of BCa.
- Published
- 2022
50. LINC00624/TEX10/NF-κB axis promotes proliferation and migration of human prostate cancer cells
- Author
-
Jian Zhou, Juan Li, Chunya Qian, Feng Qiu, Qianghua Shen, Ruiqing Tong, Qian Yang, Jinfu Xu, Bo Zheng, Jinxing Lv, and Jianquan Hou
- Subjects
Male ,NF-kappa B ,Biophysics ,Nuclear Proteins ,Prostatic Neoplasms ,Oncogenes ,Cell Biology ,Biochemistry ,Cell Movement ,Cell Line, Tumor ,Humans ,RNA, Long Noncoding ,Molecular Biology ,Cell Proliferation ,Signal Transduction - Abstract
Prostate cancer (PCa) is a malignant epithelial tumor with a high rate of biochemical or local recurrence. Studies have suggested that LINC00624 plays an important oncogenic role in liver cancer. However, whether it exerts similar effects in PCa progression remains unknown. In this study, we explored the effects of LINC00624 on the malignant progression of PCa and sought to identify the relevant signaling pathways. The results showed that LINC00624 was highly expressed in PCa tissues and cells and was associated with poor prognosis in PCa patients. In vitro and in vivo assays further showed that LINC00624 knockdown could decrease the proliferative and migratory ability of PCa cells. Mechanistically, we found that LINC00624 and TEX10 formed a co-regulatory axis that stimulated NF-κB activity. Our data suggest that LINC00624 acts as an oncogene in PCa progression and has potential as a novel biomarker for PCa.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.