Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42.209
Filter
1.
J Immunol Res ; 2024: 8273732, 2024.
Article in English | MEDLINE | ID: mdl-39359694

ABSTRACT

Oxidative stress is crucial in ulcerative colitis (UC) and colitis-associated colorectal cancer (CAC). Intestinal epithelial cells (IECs) are an important component of the intestinal barrier. In previous studies, we have demonstrated that suppressing microRNA-222-3p (miR-222-3p) can protect against oxidative stress in IECs, which ameliorates colonic injuries in UC mice and prevents the conversion of UC to CAC. In this case, we hope to explore whether moxibustion can alleviate UC and CAC by inhibiting miR-222-3p based on mouse models of UC and CAC. After herb-partitioned moxibustion (HPM) intervention, the disease activity index (DAI) and colon macroscopic damage index (CMDI) were significantly reduced in UC mice, and the number and volume of intestinal tumors were decreased considerably in CAC mice. Meanwhile, we found that HPM suppressed miR-222-3p expression and upregulated the mRNA and protein expression of Brahma-related gene 1 (BRG1), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), while inhibiting Kelch-like ECH-associated protein 1 (Keap1) expression in IECs of UC and CAC mice. With changes in reactive oxygen species (ROS), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), and inflammatory cytokines interleukin (IL)-1ß and tumor necrosis factor (TNF)-α), we verified that HPM protects against oxidative stress and inflammation in IECs of UC and CAC mice. The effect of HPM was inhibited in miR-222-3p overexpression mice, further demonstrating that the protective effect of HPM on UC and CAC mice was through inhibiting miR-222-3p. In summary, HPM regulates the BRG1/Nrf2/HO-1 pathway by inhibiting miR-222-3p to attenuate oxidative stress in IECs in UC and CAC.


Subject(s)
Colitis, Ulcerative , Disease Models, Animal , Heme Oxygenase-1 , MicroRNAs , Moxibustion , NF-E2-Related Factor 2 , Oxidative Stress , Signal Transduction , Transcription Factors , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Colitis, Ulcerative/therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/genetics , Mice , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , DNA Helicases/metabolism , DNA Helicases/genetics , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Humans
2.
Mol Autism ; 15(1): 42, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39350244

ABSTRACT

BACKGROUND: SETBP1 Haploinsufficiency Disorder (SETBP1-HD) is characterised by mild to moderate intellectual disability, speech and language impairment, mild motor developmental delay, behavioural issues, hypotonia, mild facial dysmorphisms, and vision impairment. Despite a clear link between SETBP1 mutations and neurodevelopmental disorders the precise role of SETBP1 in neural development remains elusive. We investigate the functional effects of three SETBP1 genetic variants including two pathogenic mutations p.Glu545Ter and SETBP1 p.Tyr1066Ter, resulting in removal of SKI and/or SET domains, and a point mutation p.Thr1387Met in the SET domain. METHODS: Genetic variants were introduced into induced pluripotent stem cells (iPSCs) and subsequently differentiated into neurons to model the disease. We measured changes in cellular differentiation, SETBP1 protein localisation, and gene expression changes. RESULTS: The data indicated a change in the WNT pathway, RNA polymerase II pathway and identified GATA2 as a central transcription factor in disease perturbation. In addition, the genetic variants altered the expression of gene sets related to neural forebrain development matching characteristics typical of the SETBP1-HD phenotype. LIMITATIONS: The study investigates changes in cellular function in differentiation of iPSC to neural progenitor cells as a human model of SETBP1 HD disorder. Future studies may provide additional information relevant to disease on further neural cell specification, to derive mature neurons, neural forebrain cells, or brain organoids. CONCLUSIONS: We developed a human SETBP1-HD model and identified perturbations to the WNT and POL2RA pathway, genes regulated by GATA2. Strikingly neural cells for both the SETBP1 truncation mutations and the single nucleotide variant displayed a SETBP1-HD-like phenotype.


Subject(s)
Carrier Proteins , Cell Differentiation , Haploinsufficiency , Induced Pluripotent Stem Cells , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Humans , Carrier Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Mutation , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Neurons/metabolism , Neural Stem Cells/metabolism , Wnt Signaling Pathway/genetics , Intellectual Disability/genetics , Phenotype
3.
Proc Natl Acad Sci U S A ; 121(41): e2321378121, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39352925

ABSTRACT

Progerin causes Hutchinson-Gilford progeria syndrome (HGPS), but how progerin accelerates aging is still an interesting question. Here, we provide evidence linking nuclear envelope (NE) budding and accelerated aging. Mechanistically, progerin disrupts nuclear lamina to induce NE budding in concert with lamin A/C, resulting in transport of chromatin into the cytoplasm where it is removed via autophagy, whereas emerin antagonizes this process. Primary cells from both HGPS patients and mouse models express progerin and display NE budding and chromatin loss, and ectopically expressing progerin in cells can mimic this process. More excitingly, we screen a NE budding inhibitor chaetocin by high-throughput screening, which can dramatically sequester progerin from the NE and prevent this NE budding through sustaining ERK1/2 activation. Chaetocin alleviates NE budding-induced chromatin loss and ameliorates HGPS defects in cells and mice and significantly extends lifespan of HGPS mice. Collectively, we propose that progerin-induced NE budding participates in the induction of progeria, highlight the roles of chaetocin and sustained ERK1/2 activation in anti-aging, and provide a distinct avenue for treating HGPS.


Subject(s)
Lamin Type A , Nuclear Envelope , Nuclear Proteins , Progeria , Progeria/metabolism , Progeria/drug therapy , Progeria/pathology , Progeria/genetics , Animals , Lamin Type A/metabolism , Lamin Type A/genetics , Mice , Humans , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Aging/metabolism , Aging/drug effects , Chromatin/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Disease Models, Animal , Autophagy/drug effects
5.
Article in Russian | MEDLINE | ID: mdl-39269294

ABSTRACT

OBJECTIVE: To analyze clinical and genetic characteristics of patients with the verified rare forms of autosomal recessive spinocerebellar ataxias, ATX-ANO10 and ATX-SYNE1. MATERIAL AND METHODS: Six unrelated patients with established diagnoses were examined: 4 patients with ATX-ANO10 and 2 patients with ATX-SYNE1. Brain MRI and nerve conduction study were performed. To screen for cognitive impairment, the scale for the Assessment and Rating of Ataxia (SARA), and the Montreal Cognitive Assessment Scale (MoCA) were used. Mutation screening included panel sequencing on the Illumina MiSeq platform. RESULTS: Six variants were found in the ANO10 gene: the previously described pathogenic nonsense mutations c.G1025A (p.W342X) and c.C1244G (p.S415X), as well as novel probably pathogenic variants c.1477-2A>G and c.G101T (p.W34L) and missense mutations c.A110C (p.N37T) and c.T104C (p.L35P) of undetermined significance. A novel nonsense mutation c.C8911T (p.Q2971X) and a previously described pathogenic variant c.C4939T (p.Q1647X) were found in the SYNE1 gene. The clinical presentation of the ATX-ANO10 and ATX-SYNE1 was typical presenting with slowly progressive cerebellar ataxia with pyramidal signs, with young onset and cerebellar atrophy according to brain MRI study. CONCLUSION: We provided first-ever data on clinical features and mutation spectrum In Russian patients with ATX-ANO10 and ATX-SYNE1. The phenotype of these ataxias is nonspecific, so the method of choice for molecular diagnostics is massive parallel sequencing.


Subject(s)
Anoctamins , Cytoskeletal Proteins , Nerve Tissue Proteins , Spinocerebellar Ataxias , Humans , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/diagnostic imaging , Male , Female , Nerve Tissue Proteins/genetics , Adult , Cytoskeletal Proteins/genetics , Anoctamins/genetics , Mutation , Nuclear Proteins/genetics , Middle Aged , Magnetic Resonance Imaging , Codon, Nonsense , Mutation, Missense , Adolescent , Young Adult
6.
BMC Biol ; 22(1): 195, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256730

ABSTRACT

BACKGROUND: iPSC reprogramming technology exhibits significant promise in the realms of clinical therapeutics, disease modeling, pharmaceutical drug discovery, and various other applications. However, the extensive utilization of this technology has encountered impediments in the form of inefficiency, prolonged procedures, and ambiguous biological processes. Consequently, in order to improve this technology, it is of great significance to delve into the underlying mechanisms involved in iPSC reprogramming. The BET protein BRD4 plays a crucial role in the late stage of reprogramming; however, its precise function in the early stage remains unclear. RESULTS: Our study aims to investigate BRD4's role in the early stages of iPSC reprogramming. Our investigation reveals that early inhibition of BRD4 substantially enhances iPSC reprogramming, whereas its implementation during the middle-late stage impedes the process. During the reprogramming, ribosome DNA expression initially increases before decreasing and then gradually recovers. Early inhibition of BRD4 improved the decline and restoration of rDNA expression in the early and middle-late stages, respectively. Additionally, we uncovered the mechanism of BRD4's regulation of rDNA transcription throughout reprogramming. Specifically, BRD4 interacts with UBF and co-localizes to both the rDNA promoter and enhancer regions. Ultimately, BRD4 facilitates rDNA transcription by promoting the enrichment of histone H3 lysine 27 acetylation in the surrounding chromatin. Moreover, we also discovered that early inhibition of BRD4 facilitates cells' transition out of the somatic cell state and activate pluripotent genes. CONCLUSIONS: In conclusion, our results demonstrate that early inhibition of BRD4 promotes sequential dynamic expression of rDNA, which improves iPSC reprogramming efficiency.


Subject(s)
Cellular Reprogramming , DNA, Ribosomal , Induced Pluripotent Stem Cells , Transcription Factors , Induced Pluripotent Stem Cells/metabolism , Cellular Reprogramming/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , DNA, Ribosomal/genetics , Animals , Humans , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Mice , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Bromodomain Containing Proteins
7.
Stem Cell Res Ther ; 15(1): 292, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39256792

ABSTRACT

BACKGROUND: Hepatic progenitor cells serve not only as the origin of combined hepatocellular cholangiocarcinoma (cHCC-CCA) but are also responsible for malignancy recurrence after surgical resection. Nucleophosmin 1 (NPM1) has been implicated in cancer metastasis and poor prognosis. This study aimed to determine the expression of NPM1 by hepatic progenitor cells in cHCC-CCA and the effects of targeting NPM1 on hepatic progenitor cells and BEL-7402 cells with characteristics of both progenitor cells and cHCC-CCA. METHODS: First, NPM1 was detected by RT‒PCR, western blotting, and double-immunofluorescence staining in cHCC-CCA tissues. NPM1 expression was subsequently analysed in rat hepatic progenitor cells cultured in vitro and in interleukin 6 (IL6)-treated cells. The effects and mechanism of NPM1 on hepatic progenitor cells were determined by knocking down NPM1 and performing RNA sequencing analysis. Finally, NSC348884, a small-molecule inhibitor that disrupts NPM1 dimer formation, was used to confirm the function of NPM1 in BEL-7402 cells. RESULTS: Both human hepatic progenitor cells in cHCC-CCA tissues and rat in vitro cultured hepatic progenitor cells highly expressed NPM1. IL6, a cytokine involved in the malignant transformation of hepatic progenitor cells, dose-dependently increased NPM1 and PCNA expression. Knocking down NPM1 reduced IL6R transcription (P < 0.0001) and inhibited the proliferation (P = 0.0065) of hepatic progenitor cells by suppressing the mTOR signalling pathway and activating the apoptosis pathway. Furthermore, knocking down NPM1 in hepatic progenitor cells resulted in more apoptotic cells (7.33 ± 0.09% vs. 3.76 ± 0.13%, P < 0.0001) but fewer apoptotic cells in the presence of NSC348884 (47.57 ± 0.49% vs. 63.40 ± 0.05%, P = 0.0008) than in the control cells, suggesting that low-NPM1-expressing cells are more resistant to NSC348884. In addition, NSC348884 induced the apoptosis of BEL-7402 cells with an IC50 of 2.77 µmol/L via the downregulation of the IL-6R and mTOR signalling pathways and inhibited the growth of BEL-7402 cells in a subcutaneous xenograft tumour model (P = 0.0457). CONCLUSIONS: Targeting NPM1 inhibits proliferation and induces apoptosis in hepatic progenitor cells and BEL-7402 cells, thus serving as a potential therapy for cHCC-CCA.


Subject(s)
Apoptosis , Cell Proliferation , Nuclear Proteins , Nucleophosmin , Signal Transduction , Stem Cells , TOR Serine-Threonine Kinases , Humans , Apoptosis/drug effects , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Cell Proliferation/drug effects , Animals , Signal Transduction/drug effects , Rats , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Stem Cells/metabolism , Stem Cells/cytology , Male , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Cell Line, Tumor , Mice
8.
Sci Rep ; 14(1): 21926, 2024 09 20.
Article in English | MEDLINE | ID: mdl-39300184

ABSTRACT

Despite extensive research, the molecular role of AGR2 in the progression and metastasis of colorectal cancer (CRC) has not been fully characterized. We used quantitative mass spectrometry (SWATH MS) to identify differentially expressed proteins in paired CRC cell models of the SW480 and SW620 cell lines in response to AGR2 protein level manipulation. Relying on the results from SWATH MS and subsequent immunochemical validation, we selected NMP3 as the top candidate protein associated with AGR2 in CRC tumour cells in our screen. RT‒qPCR and immunochemical analysis confirmed the involvement of AGR2-mediated regulation of NPM3 at the transcriptional and posttranscriptional levels. Since PD-L1 is a constituent of the NPM3 regulatory axis, we aimed to correlate the changes in PD-L1 to the differential expression of AGR2 in our cell models. We found that AGR2 positively regulates PD-L1 levels in both SW480 and SW620 cell lines; additionally, several different CRC patient transcriptome cohorts confirmed the association of AGR2 with PD-L1. Our work reveals a new AGR2-NPM3 regulatory axis and the involvement of AGR2 in the regulation of PD-L1, which paves the way for the association of AGR2 with immune evasion in CRC cells.


Subject(s)
B7-H1 Antigen , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Mucoproteins , Nucleophosmin , Oncogene Proteins , Proteins , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Mucoproteins/metabolism , Mucoproteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Oncogene Proteins/metabolism , Oncogene Proteins/genetics , Proteins/metabolism , Proteins/genetics
9.
Science ; 385(6715): 1366-1375, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39298589

ABSTRACT

Faithful chromosome segregation requires biorientation, where the pair of kinetochores on the chromosome establish bipolar microtubule attachment. The integrity of the kinetochore, a macromolecular complex built on centromeric DNA, is required for biorientation, but components sufficient for biorientation remain unknown. Here, we show that tethering the outer kinetochore heterodimer NDC80-NUF2 to the surface of apolar microbeads establishes their biorientation-like state in mouse cells. NDC80-NUF2 microbeads align at the spindle equator and self-correct alignment errors. The alignment is associated with stable bipolar microtubule attachment and is independent of the outer kinetochore proteins SPC24-SPC25, KNL1, the Mis12 complex, inner kinetochore proteins, and Aurora. Larger microbeads align more rapidly, suggesting a size-dependent biorientation mechanism. This study demonstrates a biohybrid kinetochore design for synthetic biorientation of microscale particles in cells.


Subject(s)
Cell Cycle Proteins , Chromosome Segregation , Kinetochores , Microspheres , Microtubule-Associated Proteins , Microtubules , Spindle Apparatus , Animals , Mice , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Spindle Apparatus/metabolism
10.
Nat Commun ; 15(1): 8195, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294122

ABSTRACT

Progressive supranuclear palsy (PSP) is an incurable neurodegenerative disease characterized by 4-repeat (0N/4R)-Tau protein accumulation in CNS neurons. We generated transgenic zebrafish expressing human 0N/4R-Tau to investigate PSP pathophysiology. Tau zebrafish replicated multiple features of PSP, including: decreased survival; hypokinesia; impaired optokinetic responses; neurodegeneration; neuroinflammation; synapse loss; and Tau hyperphosphorylation, misfolding, mislocalization, insolubility, truncation, and oligomerization. Using automated assays, we screened 147 small molecules for activity in rescuing neurological deficits in Tau zebrafish. (+)JQ1, a bromodomain inhibitor, improved hypokinesia, survival, microgliosis, and brain synapse elimination. A heterozygous brd4+/- mutant reducing expression of the bromodomain protein Brd4 similarly rescued these phenotypes. Microglial phagocytosis of synaptic material was decreased by (+)JQ1 in both Tau zebrafish and rat primary cortical cultures. Microglia in human PSP brains expressed Brd4. Our findings implicate Brd4 as a regulator of microglial synaptic elimination in tauopathy and provide an unbiased approach for identifying mechanisms and therapeutic targets in PSP.


Subject(s)
Animals, Genetically Modified , Disease Models, Animal , Microglia , Supranuclear Palsy, Progressive , Synapses , Transcription Factors , Zebrafish , tau Proteins , Animals , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , tau Proteins/metabolism , tau Proteins/genetics , Microglia/metabolism , Microglia/pathology , Synapses/metabolism , Supranuclear Palsy, Progressive/metabolism , Supranuclear Palsy, Progressive/genetics , Supranuclear Palsy, Progressive/pathology , Azepines/pharmacology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Triazoles/pharmacology , Rats , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Brain/metabolism , Brain/pathology , Phagocytosis , Neurons/metabolism , Bromodomain Containing Proteins , Cell Cycle Proteins
11.
Medicina (Kaunas) ; 60(9)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39336484

ABSTRACT

Background and Objectives: With the advent of novel therapies for nucleophosmin gene (NPM1)-mutated acute myeloid leukemia (AML), there is a growing need for the reliable prediction of NPM1 mutations. This study explored the role of cytomorphological features in the early prediction of NPM1-mutated AML. Materials and Methods: Altogether, 212 de novo AML cases with normal karyotypes, diagnosed and treated at a single institution within 5 years (2018-2023), were retrospectively evaluated. A final diagnosis of NPM1-mutated AML, based on the World Health Organization (WHO) integrated criteria, including real-time based identification of NPM1 mutation and normal karyotype, was established in 83/212 (39.15%) cases. Results: Cup-like blasts (CLBs), a cytomorphological feature suggestive of NPM1-mutated AML, were detected in 56/83 (67%) patients. Most cases (44/56, 78.6%) had CLB ≥ 10%. In total, 27 of 83 AML NPM1-mutated patients had no CLB morphology (missed call). Additionally, two of 212 had CLB morphology without confirmed NPM1 mutation (wrong call). The positive/negative predictive values of cytomorphological evaluation for CLB ≥ 10% were 95.7%/75.6%, with sensitivity/specificity of 53%/98.5%, while the accuracy was 80.7%. We noted an increased percentage of CLBs (≥15%) in 77.8% and 50% of patients with AML without and with granulocytic maturation, respectively (the specificity for NPM1 mutation prediction was 100%). CLB was associated with fms-like tyrosine kinase 3 (FLT3) mutation (p = 0.03), but, without statistical significance for CLB ≥ 10% and CLB ≥ 15%. Conclusions: Our investigation confirmed that the morphological identification of CLB at diagnosis represents a reliable and easily reproducible tool for the early prediction of NPM1 mutations, enabling a streamlined genetic work-up for its confirmation. This may facilitate considering the early administration of individualized therapies by clinicians for specific patients.


Subject(s)
Leukemia, Myeloid, Acute , Mutation , Nuclear Proteins , Nucleophosmin , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Nuclear Proteins/genetics , Male , Female , Middle Aged , Retrospective Studies , Aged , Adult , Aged, 80 and over
12.
Commun Biol ; 7(1): 1196, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39341909

ABSTRACT

Selective elimination of cancer cells without causing deleterious effects on normal cells is an ideal anti-cancer strategy. Here, using Drosophila cancer model, we performed an in vivo RNAi screen for anti-cancer targets that selectively eliminate tumors without affecting normal tissue growth. In Drosophila imaginal epithelium, clones of cells expressing oncogenic Ras with simultaneous mutations in the cell polarity gene scribble (RasV12/scrib-/-) develop into malignant tumors. We found that knockdown of Crk, the Drosophila ortholog of human CRK (CT10 regulatory kinase) and CRKL (Crk-like) adapter proteins, significantly suppresses growth of RasV12/scrib-/- tumors by inducing c-Jun N-terminal kinase (JNK)-mediated apoptosis, while it does not affect growth of normal epithelium. Mechanistically, Crk inhibition blocks Yorkie (Yki)/YAP activity by impairing F-actin accumulation, an upstream event of Yki/YAP activation in tumors. Inhibition of Yki/YAP in tumors causes intracellular JNK signaling to be used for apoptosis induction. Given that molecules and signaling pathways identified in Drosophila are highly conserved and activated in human cancers, our findings would provide a novel, to the best of our knowledge, anti-cancer strategy against YAP-activated cancers.


Subject(s)
Apoptosis , Drosophila Proteins , Trans-Activators , YAP-Signaling Proteins , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Drosophila melanogaster/genetics , Proto-Oncogene Proteins c-crk/metabolism , Proto-Oncogene Proteins c-crk/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , Drosophila/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Protein Serine-Threonine Kinases , Intracellular Signaling Peptides and Proteins
13.
PLoS One ; 19(9): e0310219, 2024.
Article in English | MEDLINE | ID: mdl-39259742

ABSTRACT

Nucleostemin (NS) plays a role in liver regeneration, and aging reduces its expression in the baseline and regenerating livers following 70% partial hepatectomy (PHx). Here we interrogate the mechanism controlling NS expression during liver regeneration and aging. The NS promoter was analyzed by TRANSFAC. Functional studies were performed using cell-based luciferase assay, endogenous NS expression in Hep3B cells, mouse livers with a gain-of-function mutation of C/EBPα (S193D), and mouse livers with C/EBPα knockdown. We found a CAAT box with four C/EBPα binding sites (-1216 to -735) and a GC box with consensus binding sites for c-Myc, E2F1, and p300-associated protein complex (-633 to -1). Age-related changes in NS expression correlated positively with the expression of c-Myc, E2F1, and p300, and negatively with that of C/EBPα and C/EBPß. PHx upregulated NS expression at 1d, coinciding with an increase in E2F1 and a decrease in C/EBPα. C/EBPα bound to the consensus sequences found in the NS promoter in vitro and in vivo, inhibited its transactivational activity in a binding site-dependent manner, and decreased the expression of endogenous NS in Hep3B cells. In vivo activation of C/EBPα by the S193D mutation resulted in a 4th-day post-PHx reduction of NS, a feature shared by 16-m/o livers. Finally, C/EBPα knockdown increased its expression in aged (24-m/o) livers under both baseline and regeneration conditions. This study reports the C/EBPα suppression of NS expression in aged livers, providing a new perspective on the mechanistic orchestration of tissue homeostasis in aging.


Subject(s)
Aging , GTP-Binding Proteins , Liver Regeneration , Nuclear Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc , Animals , Liver Regeneration/genetics , Liver Regeneration/physiology , Mice , Aging/metabolism , Aging/physiology , Aging/genetics , Humans , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Hepatectomy , Binding Sites , Liver/metabolism , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation , Transcription, Genetic , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , Male , Carrier Proteins/metabolism , Carrier Proteins/genetics , Mice, Inbred C57BL , Cell Line, Tumor , RNA-Binding Proteins
14.
Exp Mol Med ; 56(9): 2016-2032, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39218980

ABSTRACT

Micronuclei (MN) can form through many mechanisms, including the breakage of aberrant cytokinetic chromatin bridges. The frequent observation of MN in tumors suggests that they might not merely be passive elements but could instead play active roles in tumor progression. Here, we propose a mechanism through which the presence of micronuclei could induce specific phenotypic and functional changes in cells and increase the invasive potential of cancer cells. Through the integration of diverse in vitro imaging and molecular techniques supported by clinical samples from patients with prostate cancer (PCa) defined as high-risk by the D'Amico classification, we demonstrate that the resolution of chromosome bridges can result in the accumulation of Emerin and the formation of Emerin-rich MN. These structures are negative for Lamin A/C and positive for the Lamin-B receptor and Sec61ß. MN can act as a protein sinks and result in the pauperization of Emerin from the nuclear envelope. The Emerin mislocalization phenotype is associated with a molecular signature that is correlated with a poor prognosis in PCa patients and is enriched in metastatic samples. Emerin mislocalization corresponds with increases in the migratory and invasive potential of tumor cells, especially in a collagen-rich microenvironment. Our study demonstrates that the mislocalization of Emerin to MN results in increased cell invasiveness, thereby worsening patient prognosis.


Subject(s)
Chromatin , Collagen , Membrane Proteins , Neoplasm Invasiveness , Nuclear Proteins , Prostatic Neoplasms , Tumor Microenvironment , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Chromatin/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Cell Line, Tumor , Collagen/metabolism , Nuclear Envelope/metabolism , Micronuclei, Chromosome-Defective , Cell Movement
15.
Cell Death Differ ; 31(10): 1242-1254, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39266717

ABSTRACT

The proteasome, the catalytic arm of the ubiquitin system, is regulated via its dynamic compartmentation between the nucleus and the cytoplasm, among other mechanisms. Under amino acid shortage, the proteolytic complex is translocated to the cytoplasm, where it stimulates proteolysis to supplement recycled amino acids for essential protein synthesis. This response is mediated via the mTOR pathway and the lack of the three aromatic amino acids Tyr, Trp, and Phe (YWF). mTOR activation by supplementation of the triad inhibits proteasome translocation, leading to cell death. We now show that tumoral inherent stress conditions result in translocation of the proteasome from the nucleus to the cytosol. We further show that the modulation of the signaling cascade governed by YWF is applicable also to non-starved cells by using higher concentration of the triad to achieve a surplus relative to all other amino acids. Based on these two phenomena, we found that the modulation of stress signals via the administration of YWF leads to nuclear proteasome sequestration and inhibition of growth of xenograft, spontaneous, and metastatic mouse tumor models. In correlation with the observed effect of YWF on tumors, we found - using transcriptomic and proteomic analyses - that the triad affects various cellular processes related to cell proliferation, migration, and death. In addition, Sestrin3-a mediator of YWF sensing upstream of mTOR-is essential for proteasome translocation, and therefore plays a pro-tumorigenic role, positioning it as a potential oncogene. This newly identified approach for hijacking the cellular "satiety center" carries therefore potential therapeutic implications for cancer.


Subject(s)
Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Animals , Humans , Mice , Amino Acids, Aromatic/metabolism , Cell Nucleus/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Protein Transport , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
16.
Proc Natl Acad Sci U S A ; 121(39): e2319666121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39288176

ABSTRACT

Mammalian Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) and Drosophila Yorkie (Yki) are transcription cofactors of the highly conserved Hippo signaling pathway. It has been long assumed that the YAP/TAZ/Yki signaling drives cell proliferation during organ growth. However, its instructive role in regulating developmentally programmed organ growth, if any, remains elusive. Out-of-context gain of YAP/TAZ/Yki signaling often turns oncogenic. Paradoxically, mechanically strained, and differentiated squamous epithelia display developmentally programmed constitutive nuclear YAP/TAZ/Yki signaling. The unknown, therefore, is how a growth-promoting YAP/TAZ/Yki signaling restricts proliferation in differentiated squamous epithelia. Here, we show that reminiscent of a tumor suppressor, Yki negatively regulates the cell growth-promoting PI3K/Akt/TOR signaling in the squamous epithelia of Drosophila tubular organs. Thus, downregulation of Yki signaling in the squamous epithelium of the adult male accessory gland (MAG) up-regulates PI3K/Akt/TOR signaling, inducing cell hypertrophy, exit from their cell cycle arrest, and, finally, culminating in squamous cell carcinoma (SCC). Thus, blocking PI3K/Akt/TOR signaling arrests Yki loss-induced MAG-SCC. Further, MAG-SCCs, like other lethal carcinomas, secrete a cachectin, Impl2-the Drosophila homolog of mammalian IGFBP7-inducing cachexia and shortening the lifespan of adult males. Moreover, in the squamous epithelium of other tubular organs, like the dorsal trunk of larval tracheal airways or adult Malpighian tubules, downregulation of Yki signaling triggers PI3K/Akt/TOR-induced cell hypertrophy. Our results reveal that Yki signaling plays an instructive, antiproliferative role in the squamous epithelia of tubular organs.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Nuclear Proteins , Protein Serine-Threonine Kinases , Signal Transduction , Trans-Activators , YAP-Signaling Proteins , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Male , Drosophila melanogaster/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Epithelium/metabolism , Cell Proliferation , Phosphatidylinositol 3-Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics
17.
Eur J Cancer ; 211: 114308, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39288736

ABSTRACT

The Cell Division Cycle Associated 2 (CDCA2) gene is responsible for encoding a targeting subunit of cell-cycle associated protein. CDCA2 plays a crucial role in various cellular processes, including chromosome segregation and decondensation, nuclear envelope reassembly, microtubule assembly, and DNA damage response. Additionally, CDCA2 is involved in multiple signaling pathways such as the PI3K/Akt pathway and p53 pathway. Undoubtedly, there exists a strong association between CDCA2 and cancer. Numerous studies have reported that elevated levels of CDCA2 are correlated with poor prognosis and several clinicopathological characteristics like tumor size and TNM stage across different types of cancer. Therefore, CDCA2 holds great potential as both a biomarker for diagnosis and a therapeutic target for interventions such as targeted therapies or immunotherapy. Given its promising prospects in scientific research and clinical applications, it is imperative for researchers to delve into the underlying mechanisms of CDCA2 and explore its utilization.


Subject(s)
Biomarkers, Tumor , Cell Cycle Proteins , Neoplasms , Humans , Neoplasms/genetics , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Molecular Targeted Therapy/methods , Carrier Proteins
18.
CNS Neurosci Ther ; 30(9): e70033, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39267282

ABSTRACT

AIMS: Ischemic stroke is a major cause of disability and mortality worldwide. Transcranial direct current stimulation (tDCS) and isoflurane (ISO) preconditioning exhibit neuroprotective properties. However, it remains unclear whether tDCS enhances the protective effect of ISO preconditioning on ischemic stroke, and the underlying mechanisms are yet to be clarified. METHOD: A model of middle cerebral artery occlusion (MCAO), a rat ischemia-reperfusion (I/R) injury model, and an in vitro oxygen-glucose deprivation/re-oxygenation (O/R) model of ischemic injury were developed. ISO preconditioning and tDCS were administered daily for 7 days before MCAO modeling. Triphenyltetrazolium chloride staining, modified neurological severity score, and hanging-wire test were conducted to assess infarct volume and neurological outcomes. Untargeted metabolomic experiments, adeno-associated virus, lentiviral vectors, and small interfering RNA techniques were used to explore the underlying mechanisms. RESULTS: tDCS/DCS enhanced the protective effects of ISO pretreatment on I/R injury-induced brain damage. This was evidenced by reduced infarct volume and improved neurological outcomes in rats with MCAO, as well as decreased cortical neuronal death after O/R injury. Untargeted metabolomic experiments identified oxidative phosphorylation (OXPHOS) as a critical pathological process for ISO-mediated neuroprotection from I/R injury. The combination of tDCS/DCS with ISO preconditioning significantly inhibited I/R injury-induced OXPHOS. Mechanistically, Akirin2, a small nuclear protein that regulates cell proliferation and differentiation, was found to decrease in the cortex of rats with MCAO and in cortical primary neurons subjected to O/R injury. Akirin2 functions upstream of phosphatase and tensin homolog deleted on chromosome 10 (PTEN). tDCS/DCS was able to further upregulate Akirin2 levels and activate the Akirin2/PTEN signaling pathway in vivo and in vitro, compared with ISO pretreatment alone, thereby contributing to the improvement of cerebral I/R injury. CONCLUSION: tDCS treatment enhances the neuroprotective effects of ISO preconditioning on ischemic stroke by inhibiting oxidative stress and activating Akirin2-PTEN signaling pathway, highlighting potential of combination therapy in ischemic stroke.


Subject(s)
Infarction, Middle Cerebral Artery , Isoflurane , Rats, Sprague-Dawley , Reperfusion Injury , Transcranial Direct Current Stimulation , Animals , Isoflurane/pharmacology , Male , Reperfusion Injury/prevention & control , Rats , Transcranial Direct Current Stimulation/methods , Ischemic Preconditioning/methods , Brain Ischemia/prevention & control , Neuroprotective Agents/pharmacology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Anesthetics, Inhalation/pharmacology
19.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273420

ABSTRACT

Radiation therapy continues to be the cornerstone treatment for malignant brain tumors, the majority of which express wild-type p53. Therefore, the identification of drugs that promote the ionizing radiation (IR)-induced activation of p53 is expected to increase the efficacy of radiation therapy for these tumors. The growth inhibitory effects of CEP-1347, a known inhibitor of MDM4 expression, on malignant brain tumor cell lines expressing wild-type p53 were examined, alone or in combination with IR, by dye exclusion and/or colony formation assays. The effects of CEP-1347 on the p53 pathway, alone or in combination with IR, were examined by RT-PCR and Western blot analyses. The combination of CEP-1347 and IR activated p53 in malignant brain tumor cells and inhibited their growth more effectively than either alone. Mechanistically, CEP-1347 and IR each reduced MDM4 expression, while their combination did not result in further decreases. CEP-1347 promoted IR-induced Chk2 phosphorylation and increased p53 expression in concert with IR in a Chk2-dependent manner. The present results show, for the first time, that CEP-1347 is capable of promoting Chk2-mediated p53 activation by IR in addition to inhibiting the expression of MDM4 and, thus, CEP-1347 has potential as a radiosensitizer for malignant brain tumors expressing wild-type p53.


Subject(s)
Brain Neoplasms , Checkpoint Kinase 2 , Radiation, Ionizing , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Checkpoint Kinase 2/metabolism , Checkpoint Kinase 2/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Brain Neoplasms/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Phosphorylation/drug effects , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects
20.
Nat Commun ; 15(1): 7646, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223123

ABSTRACT

Despite their prevalent cancer implications, the in vivo dynamics of SWI/SNF chromatin remodelers and how misregulation of such dynamics underpins cancer remain poorly understood. Using live-cell single-molecule tracking, we quantify the intranuclear diffusion and chromatin-binding of three key subunits common to all major human SWI/SNF remodeler complexes (BAF57, BAF155 and BRG1), and resolve two temporally distinct stable binding modes for the fully assembled complex. Super-resolved density mapping reveals heterogeneous, nanoscale remodeler binding "hotspots" across the nucleoplasm where multiple binding events (especially longer-lived ones) preferentially cluster. Importantly, we uncover distinct roles of the bromodomain in modulating chromatin binding/targeting in a DNA-accessibility-dependent manner, pointing to a model where successive longer-lived binding within "hotspots" leads to sustained productive remodeling. Finally, systematic comparison of six common BRG1 mutants implicated in various cancers unveils alterations in chromatin-binding dynamics unique to each mutant, shedding insight into a multi-modal landscape regulating the spatio-temporal organizational dynamics of SWI/SNF remodelers.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin , Chromosomal Proteins, Non-Histone , DNA Helicases , Neoplasms , Nuclear Proteins , Single Molecule Imaging , Transcription Factors , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Single Molecule Imaging/methods , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , DNA Helicases/metabolism , DNA Helicases/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromatin/metabolism , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , DNA/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Protein Binding , Mutation , Cell Line, Tumor , Protein Domains , Adenosine Triphosphatases
SELECTION OF CITATIONS
SEARCH DETAIL