Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28.360
Filter
1.
Mol Biol Rep ; 51(1): 712, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824221

ABSTRACT

INTRODUCTION: Coronary artery disease (CAD) in young adults can have devastating consequences. The cardiac developmental gene MEIS1 plays important roles in vascular networks and heart development. This gene effects on the regeneration capacity of the heart. Considering role of MEIS1 in cardiac tissue development and the progression of myocardial infarction this study investigated the expression levels of the MEIS1, HIRA, and Myocardin genes in premature CAD patients compared to healthy subjects and evaluated the relationships between these genes and possible inflammatory factors. METHODS AND RESULTS: The study conducted a case-control design involving 35 CAD patients and 35 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were collected, and gene expression analysis was performed using real-time PCR. Compared with control group, the number of PBMCs in the CAD group exhibited greater MEIS1 and HIRA gene expression, with fold changes of 2.45 and 3.6. The expression of MEIS1 exhibited a negative correlation with IL-10 (r= -0.312) expression and positive correlation with Interleukin (IL)-6 (r = 0.415) and tumor necrosis factor (TNF)-α (r = 0.534) gene expression. Moreover, there was an inverse correlation between the gene expression of HIRA and that of IL-10 (r= -0.326), and a positive correlation was revealed between the expression of this gene and that of the IL-6 (r = 0.453) and TNF-α (r = 0.572) genes. CONCLUSION: This research demonstrated a disparity in expression levels of MEIS1, HIRA, and Myocardin, between CAD and healthy subjects. The results showed that, MEIS1 and HIRA play significant roles in regulating the synthesis of proinflammatory cytokines, namely, TNF-α and IL-6.


Subject(s)
Coronary Artery Disease , Myeloid Ecotropic Viral Integration Site 1 Protein , Nuclear Proteins , Trans-Activators , Adult , Female , Humans , Male , Middle Aged , Case-Control Studies , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Coronary Artery Disease/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , Interleukin-10/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Leukocytes, Mononuclear/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
Genome Biol ; 25(1): 143, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822412

ABSTRACT

BACKGROUND: Targeted therapies exploiting vulnerabilities of cancer cells hold promise for improving patient outcome and reducing side-effects of chemotherapy. However, efficacy of precision therapies is limited in part because of tumor cell heterogeneity. A better mechanistic understanding of how drug effect is linked to cancer cell state diversity is crucial for identifying effective combination therapies that can prevent disease recurrence. RESULTS: Here, we characterize the effect of G2/M checkpoint inhibition in acute lymphoblastic leukemia (ALL) and demonstrate that WEE1 targeted therapy impinges on cell fate decision regulatory circuits. We find the highest inhibition of recovery of proliferation in ALL cells with KMT2A-rearrangements. Single-cell RNA-seq and ATAC-seq of RS4;11 cells harboring KMT2A::AFF1, treated with the WEE1 inhibitor AZD1775, reveal diversification of cell states, with a fraction of cells exhibiting strong activation of p53-driven processes linked to apoptosis and senescence, and disruption of a core KMT2A-RUNX1-MYC regulatory network. In this cell state diversification induced by WEE1 inhibition, a subpopulation transitions to a drug tolerant cell state characterized by activation of transcription factors regulating pre-B cell fate, lipid metabolism, and pre-BCR signaling in a reversible manner. Sequential treatment with BCR-signaling inhibitors dasatinib, ibrutinib, or perturbing metabolism by fatostatin or AZD2014 effectively counteracts drug tolerance by inducing cell death and repressing stemness markers. CONCLUSIONS: Collectively, our findings provide new insights into the tight connectivity of gene regulatory programs associated with cell cycle and cell fate regulation, and a rationale for sequential administration of WEE1 inhibitors with low toxicity inhibitors of pre-BCR signaling or metabolism.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Histone-Lysine N-Methyltransferase/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Myeloid-Lymphoid Leukemia Protein/genetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Core Binding Factor Alpha 2 Subunit/genetics
3.
Biochemistry (Mosc) ; 89(4): 585-600, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38831498

ABSTRACT

Accurate duplication and separation of long linear genomic DNA molecules is associated with a number of purely mechanical problems. SMC complexes are key components of the cellular machinery that ensures decatenation of sister chromosomes and compaction of genomic DNA during division. Cohesin, one of the essential eukaryotic SMC complexes, has a typical ring structure with intersubunit pore through which DNA molecules can be threaded. Capacity of cohesin for such topological entrapment of DNA is crucial for the phenomenon of post-replicative association of sister chromatids better known as cohesion. Recently, it became apparent that cohesin and other SMC complexes are, in fact, motor proteins with a very peculiar movement pattern leading to formation of DNA loops. This specific process has been called loop extrusion. Extrusion underlies multiple functions of cohesin beyond cohesion, but molecular mechanism of the process remains a mystery. In this review, we summarized the data on molecular architecture of cohesin, effect of ATP hydrolysis cycle on this architecture, and known modes of cohesin-DNA interactions. Many of the seemingly disparate facts presented here will probably be incorporated in a unified mechanistic model of loop extrusion in the not-so-distant future.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cohesins , DNA , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/chemistry , DNA/metabolism , DNA/chemistry , Humans , Animals , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Chromatids/metabolism , Chromatids/chemistry
4.
Biochemistry (Mosc) ; 89(4): 601-625, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38831499

ABSTRACT

The most prominent representatives of multisubunit SMC complexes, cohesin and condensin, are best known as structural components of mitotic chromosomes. It turned out that these complexes, as well as their bacterial homologues, are molecular motors, the ATP-dependent movement of these complexes along DNA threads leads to the formation of DNA loops. In recent years, we have witnessed an avalanche-like accumulation of data on the process of SMC dependent DNA looping, also known as loop extrusion. This review briefly summarizes the current understanding of the place and role of cohesin-dependent extrusion in cell physiology and presents a number of models describing the potential molecular mechanism of extrusion in a most compelling way. We conclude the review with a discussion of how the capacity of cohesin to extrude DNA loops may be mechanistically linked to its involvement in sister chromatid cohesion.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Cohesins , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/chemistry , Humans , Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Multiprotein Complexes/metabolism , Multiprotein Complexes/chemistry , DNA/metabolism , DNA/chemistry , Animals , Chromatids/metabolism
5.
Nat Commun ; 15(1): 4729, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830897

ABSTRACT

Cohesin mediates sister chromatid cohesion to enable chromosome segregation and DNA damage repair. To perform these functions, cohesin needs to be protected from WAPL, which otherwise releases cohesin from DNA. It has been proposed that cohesin is protected from WAPL by SORORIN. However, in vivo evidence for this antagonism is missing and SORORIN is only known to exist in vertebrates and insects. It is therefore unknown how important and widespread SORORIN's functions are. Here we report the identification of SORORIN orthologs in Schizosaccharomyces pombe (Sor1) and Arabidopsis thaliana (AtSORORIN). sor1Δ mutants display cohesion defects, which are partially alleviated by wpl1Δ. Atsororin mutant plants display dwarfism, tissue specific cohesion defects and chromosome mis-segregation. Furthermore, Atsororin mutant plants are sterile and separate sister chromatids prematurely at anaphase I. The somatic, but not the meiotic deficiencies can be alleviated by loss of WAPL. These results provide in vivo evidence for SORORIN antagonizing WAPL, reveal that SORORIN is present in organisms beyond the animal kingdom and indicate that it has acquired tissue specific functions in plants.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Arabidopsis/genetics , Arabidopsis/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Cohesins , Chromosome Segregation , Mutation , Chromatids/metabolism , Chromatids/genetics , Evolution, Molecular , Meiosis/genetics
6.
Cell Mol Life Sci ; 81(1): 251, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847937

ABSTRACT

The Smc5/6 complex is a highly conserved molecular machine involved in the maintenance of genome integrity. While its functions largely depend on restraining the fork remodeling activity of Mph1 in yeast, the presence of an analogous Smc5/6-FANCM regulation in humans remains unknown. We generated human cell lines harboring mutations in the NSE1 subunit of the Smc5/6 complex. Point mutations or truncations in the RING domain of NSE1 result in drastically reduced Smc5/6 protein levels, with differential contribution of the two zinc-coordinating centers in the RING. In addition, nse1-RING mutant cells display cell growth defects, reduced replication fork rates, and increased genomic instability. Notably, our findings uncover a synthetic sick interaction between Smc5/6 and FANCM and show that Smc5/6 controls fork progression and chromosome disjunction in a FANCM-independent manner. Overall, our study demonstrates that the NSE1 RING domain plays vital roles in Smc5/6 complex stability and fork progression through pathways that are not evolutionary conserved.


Subject(s)
Cell Cycle Proteins , DNA Replication , Genomic Instability , Humans , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Protein Domains , Protein Stability , Mutation , Cell Line , DNA Helicases
8.
Folia Biol (Praha) ; 70(1): 53-61, 2024.
Article in English | MEDLINE | ID: mdl-38830123

ABSTRACT

Psoriasis is a chronic non-contagious autoimmune disease. Gallic acid is a natural compound with potential health benefits, including antioxidant, anticancer, antiviral and antibacterial properties. Nevertheless, the influence of gallic acid on psoriasis has not been fully determined. This investigation aimed to discover the effect of gallic acid on psoriasis. Thirty-one pairs of psoriatic skin tissues and healthy adult human skin tissues were collected. Human keratinocytes (HaCaT cells) were transfected with interleukin 17A (IL-17A) to create the psoriatic keratinocyte model. The content of bromodomain-containing protein 4 (BRD4) microRNA was assessed using qRT-PCR testing. The content of BRD4 was detected by Western blotting. Cell migration was evaluated by conducting a wound healing assay. Cell proliferation was determined using an EdU assay. Apoptosis was detected by the TUNEL assay. The contents of interferon gamma (IFN-γ), IL-6, IL-8 and IL-17 were detected by ELISA. BRD4 was up-regulated in psoriatic skin tissues and in the IL-17A group compared to the healthy adult human skin tissues and the control group. Silencing BRD4 inhibited cell migration, proliferation and inflammatory response but induced apoptosis in IL-17A-treated HaCaT cells. Conversely, BRD4 over-expression promoted cell migration, proliferation and inflammatory response but suppressed apoptosis in IL-17A-treated HaCaT cells. Gallic acid repressed cell migration, proliferation and inflammatory response but indu-ced apoptosis in HaCaT cells transfected with IL-17A by down-regulating BRD4. Gallic acid represses cell migration, proliferation and inflammatory response but induces apoptosis in IL-17A-transfected HaCaT cells by down-regulating BRD4.


Subject(s)
Apoptosis , Cell Cycle Proteins , Cell Movement , Cell Proliferation , Gallic Acid , Inflammation , Keratinocytes , Psoriasis , Transcription Factors , Humans , Psoriasis/metabolism , Psoriasis/pathology , Psoriasis/drug therapy , Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Gallic Acid/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Apoptosis/drug effects , Inflammation/pathology , Cell Proliferation/drug effects , Cell Movement/drug effects , Interleukin-17/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Adult , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Male , HaCaT Cells , Female , Gene Expression Regulation/drug effects , Cell Line , Bromodomain Containing Proteins
9.
Int J Biol Sci ; 20(7): 2440-2453, 2024.
Article in English | MEDLINE | ID: mdl-38725860

ABSTRACT

Glioblastoma is the prevailing and highly malignant form of primary brain neoplasm with poor prognosis. Exosomes derived from glioblastoma cells act a vital role in malignant progression via regulating tumor microenvironment (TME), exosomal tetraspanin protein family members (TSPANs) are important actors of cell communication in TME. Among all the TSPANs, TSPAN6 exhibited predominantly higher expression levels in comparison to normal tissues. Meanwhile, glioblastoma patients with high level of TSPAN6 had shorter overall survival compared with low level of TSPAN6. Furthermore, TSPAN6 promoted the malignant progression of glioblastoma via promoting the proliferation and metastatic potential of glioblastoma cells. More interestingly, TSPAN6 overexpression in glioblastoma cells promoted the migration of vascular endothelial cell, and exosome secretion inhibitor reversed the migrative ability of vascular endothelial cells enhanced by TSPAN6 overexpressing glioblastoma cells, indicating that TSPAN6 might reinforce angiogenesis via exosomes in TME. Mechanistically, TSPAN6 enhanced the malignant progression of glioblastoma by interacting with CDK5RAP3 and regulating STAT3 signaling pathway. In addition, TSPAN6 overexpression in glioblastoma cells enhanced angiogenesis via regulating TME and STAT3 signaling pathway. Collectively, TSPAN6 has the potential to serve as both a therapeutic target and a prognostic biomarker for the treatment of glioblastoma.


Subject(s)
Glioblastoma , STAT3 Transcription Factor , Signal Transduction , Tetraspanins , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Humans , STAT3 Transcription Factor/metabolism , Tetraspanins/metabolism , Tetraspanins/genetics , Cell Line, Tumor , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Animals , Cell Proliferation/genetics , Exosomes/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Movement/genetics , Disease Progression , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice
10.
BMC Cancer ; 24(1): 582, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741069

ABSTRACT

BACKGROUND: Local recurrence after surgery and radiochemotherapy seriously affects the prognosis of locally advanced rectal cancer (LARC) patients. Studies on molecular markers related to the radiochemotherapy sensitivity of cancers have been widely carried out, which might provide valued information for clinicians to carry out individual treatment. AIM: To find potential biomarkers of tumors for predicting postoperative recurrence. METHODS: In this study, LARC patients undergoing surgery and concurrent radiochemotherapy were enrolled. We focused on clinicopathological factors and PTEN, SIRT1, p-4E-BP1, and pS6 protein expression assessed by immunohistochemistry in 73 rectal cancer patients with local recurrence and 76 patients without local recurrence. RESULTS: The expression of PTEN was higher, while the expression of p-4E-BP1 was lower in patients without local recurrence than in patients with local recurrence. Moreover, TNM stage, lymphatic vessel invasion (LVI), PTEN and p-4E-BP1 might be independent risk factors for local recurrence after LARC surgery combined with concurrent radiochemotherapy. CONCLUSIONS: This study suggests that PTEN and p-4E-BP1 might be potential biomarkers for prognostic prediction and therapeutic targets for LARC.


Subject(s)
Adaptor Proteins, Signal Transducing , Biomarkers, Tumor , Cell Cycle Proteins , Chemoradiotherapy , Neoplasm Recurrence, Local , PTEN Phosphohydrolase , Rectal Neoplasms , Humans , Rectal Neoplasms/therapy , Rectal Neoplasms/pathology , Rectal Neoplasms/metabolism , PTEN Phosphohydrolase/metabolism , Male , Female , Middle Aged , Chemoradiotherapy/methods , Biomarkers, Tumor/metabolism , Aged , Prognosis , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Phosphoproteins/metabolism , Adult , Neoplasm Staging
11.
J Cell Biol ; 223(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38722822

ABSTRACT

Cell growth is required for cell cycle progression. The amount of growth required for cell cycle progression is reduced in poor nutrients, which leads to a reduction in cell size. In budding yeast, nutrients can influence cell size by modulating the extent of bud growth, which occurs predominantly in mitosis. However, the mechanisms are unknown. Here, we used mass spectrometry to identify proteins that modulate bud growth in response to nutrient availability. This led to the discovery that nutrients regulate numerous components of the mitotic exit network (MEN), which controls exit from mitosis. A key component of the MEN undergoes gradual multisite phosphorylation during bud growth that is dependent upon bud growth and correlated with the extent of growth. Furthermore, activation of the MEN is sufficient to override a growth requirement for mitotic exit. The data suggest a model in which the MEN ensures that mitotic exit occurs only when an appropriate amount of bud growth has occurred.


Subject(s)
Mitosis , Saccharomyces cerevisiae , Signal Transduction , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Nutrients/metabolism , Phosphorylation , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomycetales/metabolism , Saccharomycetales/growth & development
12.
PeerJ ; 12: e17032, 2024.
Article in English | MEDLINE | ID: mdl-38770093

ABSTRACT

Purpose: This study seeks to identify potential clinical biomarkers for osteoarthritis (OA) using bioinformatics and investigate OA mechanisms through cellular assays. Methods: Differentially Expressed Genes (DEGs) from GSE52042 (four OA samples, four control samples) were screened and analyzed with protein-protein interaction (PPI) analysis. Overlapping genes in GSE52042 and GSE206848 (seven OA samples, and seven control samples) were identified and evaluated using Gene Set Enrichment Analysis (GSEA) and clinical diagnostic value analysis to determine the hub gene. Finally, whether and how the hub gene impacts LPS-induced OA progression was explored by in vitro experiments, including Western blotting (WB), co-immunoprecipitation (Co-IP), flow cytometry, etc. Result: Bioinformatics analysis of DEGs (142 up-regulated and 171 down-regulated) in GSE52042 identified two overlapping genes (U2AF2, TPX2) that exhibit significant clinical diagnostic value. These genes are up-regulated in OA samples from both GSE52042 and GSE206848 datasets. Notably, TPX2, which AUC = 0.873 was identified as the hub gene. In vitro experiments have demonstrated that silencing TPX2 can alleviate damage to chondrocytes induced by lipopolysaccharide (LPS). Furthermore, there is a protein interaction between TPX2 and MMP13 in OA. Excessive MMP13 can attenuate the effects of TPX2 knockdown on LPS-induced changes in OA protein expression, cell growth, and apoptosis. Conclusion: In conclusion, our findings shed light on the molecular mechanisms of OA and suggested TPX2 as a potential therapeutic target. TPX2 could promote the progression of LPS-induced OA by up-regulating the expression of MMP13, which provides some implications for clinical research.


Subject(s)
Cell Cycle Proteins , Chondrocytes , Disease Progression , Lipopolysaccharides , Matrix Metalloproteinase 13 , Microtubule-Associated Proteins , Osteoarthritis , Up-Regulation , Lipopolysaccharides/pharmacology , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/chemically induced , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrocytes/drug effects , Computational Biology , Protein Interaction Maps
13.
Proc Natl Acad Sci U S A ; 121(19): e2401386121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38696471

ABSTRACT

In the meiotic prophase, programmed DNA double-strand breaks are repaired by meiotic recombination. Recombination-defective meiocytes are eliminated to preserve genome integrity in gametes. BRCA1 is a critical protein in somatic homologous recombination, but studies have suggested that BRCA1 is dispensable for meiotic recombination. Here we show that BRCA1 is essential for meiotic recombination. Interestingly, BRCA1 also has a function in eliminating recombination-defective oocytes. Brca1 knockout (KO) rescues the survival of Dmc1 KO oocytes far more efficiently than removing CHK2, a vital component of the DNA damage checkpoint in oocytes. Mechanistically, BRCA1 activates chromosome asynapsis checkpoint by promoting ATR activity at unsynapsed chromosome axes in Dmc1 KO oocytes. Moreover, Brca1 KO also rescues the survival of asynaptic Spo11 KO oocytes. Collectively, our study not only unveils an unappreciated role of chromosome asynapsis in eliminating recombination-defective oocytes but also reveals the dual functions of BRCA1 in safeguarding oocyte genome integrity.


Subject(s)
BRCA1 Protein , Cell Cycle Proteins , Mice, Knockout , Oocytes , Oocytes/metabolism , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Female , Mice , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Meiosis/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/deficiency , DNA Breaks, Double-Stranded , Chromosome Pairing/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Phosphate-Binding Proteins/metabolism , Phosphate-Binding Proteins/genetics , Recombination, Genetic , Homologous Recombination , Genomic Instability
14.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732206

ABSTRACT

Breast cancer stands out as one of the most prevalent malignancies worldwide, necessitating a nuanced understanding of its molecular underpinnings for effective treatment. Hormone receptors in breast cancer cells substantially influence treatment strategies, dictating therapeutic approaches in clinical settings, serving as a guide for drug development, and aiming to enhance treatment specificity and efficacy. Natural compounds, such as curcumin, offer a diverse array of chemical structures with promising therapeutic potential. Despite curcumin's benefits, challenges like poor solubility and rapid metabolism have spurred the exploration of analogs. Here, we evaluated the efficacy of the curcumin analog NC2603 to induce cell cycle arrest in MCF-7 breast cancer cells and explored its molecular mechanisms. Our findings reveal potent inhibition of cell viability (IC50 = 5.6 µM) and greater specificity than doxorubicin toward MCF-7 vs. non-cancer HaCaT cells. Transcriptome analysis identified 12,055 modulated genes, most notably upregulation of GADD45A and downregulation of ESR1, implicating CDKN1A-mediated regulation of proliferation and cell cycle genes. We hypothesize that the curcumin analog by inducing GADD45A expression and repressing ESR1, triggers the expression of CDKN1A, which in turn downregulates the expression of many important genes of proliferation and the cell cycle. These insights advance our understanding of curcumin analogs' therapeutic potential, highlighting not just their role in treatment, but also the molecular pathways involved in their activity toward breast cancer cells.


Subject(s)
Breast Neoplasms , Cell Cycle Checkpoints , Curcumin , Cyclin-Dependent Kinase Inhibitor p21 , Gene Expression Regulation, Neoplastic , Humans , Curcumin/pharmacology , Curcumin/analogs & derivatives , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , MCF-7 Cells , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cell Cycle Checkpoints/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Up-Regulation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Antineoplastic Agents/pharmacology , GADD45 Proteins
15.
Medicine (Baltimore) ; 103(19): e38066, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38728485

ABSTRACT

CDCA3, a cell cycle regulator gene that plays a catalytic role in many tumors, was initially identified as a regulator of cell cycle progression, specifically facilitating the transition from the G2 phase to mitosis. However, its role in glioma remains unknown. In this study, bioinformatics analyses (TCGA, CGGA, Rembrandt) shed light on the upregulation and prognostic value of CDCA3 in gliomas. It can also be included in a column chart as a parameter predicting 3- and 5-year survival risk (C index = 0.86). According to Gene Set Enrichment Analysis and gene ontology analysis, the biological processes of CDCA3 are mainly concentrated in the biological activities related to cell cycle such as DNA replication and nuclear division. CDCA3 is closely associated with many classic glioma biomarkers (CDK4, CDK6), and inhibitors of CDK4 and CDK6 have been shown to be effective in tumor therapy. We have demonstrated that high expression of CDCA3 indicates a higher malignancy and poorer prognosis in gliomas.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Cell Cycle Proteins , Glioma , Humans , Glioma/genetics , Glioma/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Prognosis , Molecular Targeted Therapy/methods , Up-Regulation , Computational Biology/methods
16.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731817

ABSTRACT

MCPH1 has been identified as the causal gene for primary microcephaly type 1, a neurodevelopmental disorder characterized by reduced brain size and delayed growth. As a multifunction protein, MCPH1 has been reported to repress the expression of TERT and interact with transcriptional regulator E2F1. However, it remains unclear whether MCPH1 regulates brain development through its transcriptional regulation function. This study showed that the knockout of Mcph1 in mice leads to delayed growth as early as the embryo stage E11.5. Transcriptome analysis (RNA-seq) revealed that the deletion of Mcph1 resulted in changes in the expression levels of a limited number of genes. Although the expression of some of E2F1 targets, such as Satb2 and Cdkn1c, was affected, the differentially expressed genes (DEGs) were not significantly enriched as E2F1 target genes. Further investigations showed that primary and immortalized Mcph1 knockout mouse embryonic fibroblasts (MEFs) exhibited cell cycle arrest and cellular senescence phenotype. Interestingly, the upregulation of p19ARF was detected in Mcph1 knockout MEFs, and silencing p19Arf restored the cell cycle and growth arrest to wild-type levels. Our findings suggested it is unlikely that MCPH1 regulates neurodevelopment through E2F1-mediated transcriptional regulation, and p19ARF-dependent cell cycle arrest and cellular senescence may contribute to the developmental abnormalities observed in primary microcephaly.


Subject(s)
Cell Cycle Checkpoints , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16 , Mice, Knockout , Microcephaly , Animals , Mice , Cellular Senescence/genetics , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Cell Cycle Checkpoints/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/deficiency , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Fibroblasts/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
17.
Eur J Med Chem ; 271: 116444, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38691889

ABSTRACT

The NAPRT-induced increase in NAD+ levels was proposed as a mechanism contributing to hepatocellular carcinoma (HCC) resistance to NAMPT inhibitors. Thus, concurrently targeting NAMPT and NAPRT could be considered to overcome drug resistance. A BRD4 inhibitor downregulates the expression of NAPRT in HCC, and the combination of NAMPT inhibitors with BRD4 inhibitors simultaneously blocks NAD+ generation via salvage and the PH synthesis pathway. Moreover, the combination of the two agents significantly downregulated the expression of tumor-promoting genes and strongly promoted apoptosis. The present work identified various NAMPT/BRD4 dual inhibitors based on the multitargeted drug rationale. Among them, compound A2, which demonstrated the strongest effect, exhibited potent inhibition of NAMPT and BRD4 (IC50 = 35 and 58 nM, respectively). It significantly suppressed the growth and migration of HCC cells and facilitated their apoptosis. Furthermore, compound A2 also manifested a robust anticancer effect in HCCLM3 xenograft mouse models, with no apparent toxic effects. Our findings in this study provide an effective approach to target NAD+ metabolism for HCC treatment.


Subject(s)
Antineoplastic Agents , Apoptosis , Carcinoma, Hepatocellular , Cell Cycle Proteins , Cell Proliferation , Cytokines , Liver Neoplasms , Nicotinamide Phosphoribosyltransferase , Transcription Factors , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Mice , Apoptosis/drug effects , Structure-Activity Relationship , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cytokines/metabolism , Cytokines/antagonists & inhibitors , Drug Discovery , Drug Screening Assays, Antitumor , Molecular Structure , Dose-Response Relationship, Drug , Mice, Nude , Cell Line, Tumor , Mice, Inbred BALB C , Bromodomain Containing Proteins
18.
Life Sci Alliance ; 7(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38724194

ABSTRACT

NUT carcinoma (NC) is an aggressive cancer with no effective treatment. About 70% of NUT carcinoma is associated with chromosome translocation events that lead to the formation of a BRD4::NUTM1 fusion gene. Because the BRD4::NUTM1 gene is unequivocally cytotoxic when ectopically expressed in cell lines, questions remain on whether the fusion gene can initiate NC. Here, we report the first genetically engineered mouse model for NUT carcinoma that recapitulates the human t(15;19) chromosome translocation in mice. We demonstrated that the mouse t(2;17) syntenic chromosome translocation, forming the Brd4::Nutm1 fusion gene, could induce aggressive carcinomas in mice. The tumors present histopathological and molecular features similar to human NC, with enrichment of undifferentiated cells. Similar to the reports of human NC incidence, Brd4::Nutm1 can induce NC from a broad range of tissues with a strong phenotypical variability. The consistent induction of poorly differentiated carcinoma demonstrated a strong reprogramming activity of BRD4::NUTM1. The new mouse model provided a critical preclinical model for NC that will lead to better understanding and therapy development for NC.


Subject(s)
Nuclear Proteins , Oncogene Proteins, Fusion , Transcription Factors , Animals , Mice , Oncogene Proteins, Fusion/genetics , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Disease Models, Animal , Carcinoma/genetics , Carcinoma/metabolism , Translocation, Genetic/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Bromodomain Containing Proteins
19.
FASEB J ; 38(9): e23633, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38690712

ABSTRACT

Recent reports suggest that the Hippo signaling pathway regulates testis development, though its exact roles in Sertoli cell differentiation remain unknown. Here, we examined the functions of the main Hippo pathway kinases, large tumor suppressor homolog kinases 1 and 2 (Lats1 and Lats2) in developing mouse Sertoli cells. Conditional inactivation of Lats1/2 in Sertoli cells resulted in the disorganization and overgrowth of the testis cords, the induction of a testicular inflammatory response and germ cell apoptosis. Stimulated by retinoic acid 8 (STRA8) expression in germ cells additionally suggested that germ cells may have been preparing to enter meiosis prior to their loss. Gene expression analyses of the developing testes of conditional knockout animals further suggested impaired Sertoli cell differentiation, epithelial-to-mesenchymal transition, and the induction of a specific set of genes associated with Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ)-mediated integrin signaling. Finally, the involvement of YAP/TAZ in Sertoli cell differentiation was confirmed by concomitantly inactivating Yap/Taz in Lats1/2 conditional knockout model, which resulted in a partial rescue of the testicular phenotypic changes. Taken together, these results identify Hippo signaling as a crucial pathway for Sertoli cell development and provide novel insight into Sertoli cell fate maintenance.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Protein Serine-Threonine Kinases , Sertoli Cells , Tumor Suppressor Proteins , YAP-Signaling Proteins , Animals , Sertoli Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Male , Mice , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Cell Differentiation/physiology , Mice, Knockout , Signal Transduction , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Testis/metabolism , Epithelial-Mesenchymal Transition/physiology , Transcription Factors/metabolism , Transcription Factors/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics
20.
Klin Onkol ; 38(2): 95-101, 2024.
Article in English | MEDLINE | ID: mdl-38697817

ABSTRACT

BACKGROUND: The N-myc downstream-regulated gene 1 (NDRG1) has been discovered as a significant gene in the progression of cancers. However, the regulatory mechanism of NDRG1 remained obscure in prostate cancer (PCa). METHODS: The miR-96-5p and NDRG1 expression levels were evaluated in PCa cell lines, and prostate tissues, and validated in public databases by real-time polymerase chain reaction, western blot analysis, and immunohistochemistry. The function of miR-96-5p and NDRG1 were investigated by scratch assay and transwell assays in vitro, and mouse xenograft assay in vivo. The candidate pathway regulated by NDRG1 was conducted by the next-generation gene sequencing technique. Immunofluorescence and luciferase assays were used to detect the relation between miR-96-5p, NDRG1, and NF-kB pathway. RESULTS: Overexpressing NDRG1 suppresses the migration, invasion, and epithelial-mesenchymal transition (EMT) in vitro, and inhibits metastasis in vivo. Moreover, miR-96-5p contributes to NDRG1 deficiency and promotes PCa cell migration and invasion. Furthermore, NDRG1 loss activates the NF-kB pathway, which stimulates p65 and IKBa phosphorylation and induces EMT in PCa. CONCLUSIONS: MiR-96-5p promotes the migration and invasion of PCa by targeting NDRG1 and regulating the NF-kB pathway.


Subject(s)
Cell Cycle Proteins , Intracellular Signaling Peptides and Proteins , MicroRNAs , NF-kappa B , Neoplasm Invasiveness , Prostatic Neoplasms , MicroRNAs/genetics , Humans , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , NF-kappa B/metabolism , Animals , Cell Line, Tumor , Mice , Epithelial-Mesenchymal Transition , Cell Movement , Gene Expression Regulation, Neoplastic
SELECTION OF CITATIONS
SEARCH DETAIL
...