Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 431
Filter
1.
Nat Commun ; 15(1): 7186, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169036

ABSTRACT

Nucleoli are fundamentally essential sites for ribosome biogenesis in cells and formed by liquid-liquid phase separation (LLPS) for a multilayer condensate structure. How the nucleoli integrity is maintained remains poorly understood. Here, we reveal that METTL3/METTL14, the typical methyltransferase complex catalyzing N6-methyladnosine (m6A) on mRNAs maintain nucleoli integrity in human embryonic stem cells (hESCs). METTL3/METTL14 deficiency impairs nucleoli and leads to the complete loss of self-renewal in hESCs. We further show that SUV39H1/H2 protein, the methyltransferases catalyzing H3K9me3 were dramatically elevated in METTL3/METTL14 deficient cells, which causes an accumulation and infiltration of H3K9me3 across the whole nucleolus and impairs the LLPS. Mechanistically, METTL3/METTL14 complex serves as an essential adapter for CRL4 E3 ubiquitin ligase targeting SUV39H1/H2 for polyubiquitination and proteasomal degradation and therefore prevents H3K9me3 accumulation in nucleoli. Together, these findings uncover a previously unknown role of METTL3/METTL14 to maintain nucleoli integrity by facilitating SUV39H1/H2 degradation in human cells.


Subject(s)
Cell Nucleolus , Methyltransferases , Repressor Proteins , Humans , Methyltransferases/metabolism , Methyltransferases/genetics , Cell Nucleolus/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , Histones/metabolism , Ubiquitination , Human Embryonic Stem Cells/metabolism , Proteolysis , HEK293 Cells , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Histone-Lysine N-Methyltransferase
2.
Orphanet J Rare Dis ; 19(1): 292, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135054

ABSTRACT

Ankyrin repeat domain containing-protein 11 (ANKRD11), a transcriptional factor predominantly localized in the cell nucleus, plays a crucial role in the expression regulation of key genes by recruiting chromatin remodelers and interacting with specific transcriptional repressors or activators during numerous biological processes. Its pathogenic variants are strongly linked to the pathogenesis and progression of multisystem disorder known as KBG syndrome. With the widespread application of high-throughput DNA sequencing technologies in clinical medicine, numerous pathogenic variants in the ANKRD11 gene have been reported. Patients with KBG syndrome usually exhibit a broad phenotypic spectrum with a variable degree of severity, even if having identical variants. In addition to distinctive dental, craniofacial and neurodevelopmental abnormalities, patients often present with skeletal anomalies, particularly postnatal short stature. The relationship between ANKRD11 variants and short stature is not well-understood, with limited knowledge regarding its occurrence rate or underlying biological mechanism involved. This review aims to provide an updated analysis of the molecular spectrum associated with ANKRD11 variants, investigate the prevalence of the short stature among patients harboring these variants, evaluate the efficacy of recombinant human growth hormone in treating children with short stature and ANKRD11 variants, and explore the biological mechanisms underlying short stature from both scientific and clinical perspectives. Our investigation indicated that frameshift and nonsense were the most frequent types in 583 pathogenic or likely pathogenic variants identified in the ANKRD11 gene. Among the 245 KBGS patients with height data, approximately 50% displayed short stature. Most patients showed a positive response to rhGH therapy, although the number of patients receiving treatment was limited. ANKRD11 deficiency potentially disrupts longitudinal bone growth by affecting the orderly differentiation of growth plate chondrocytes. Our review offers crucial insights into the association between ANKRD11 variants and short stature and provides valuable guidance for precise clinical diagnosis and treatment of patients with KBG syndrome.


Subject(s)
Phenotype , Repressor Proteins , Humans , Repressor Proteins/genetics , Abnormalities, Multiple/genetics , Intellectual Disability/genetics , Tooth Abnormalities/genetics , Tooth Abnormalities/pathology , Craniosynostoses/genetics , Craniosynostoses/pathology , Chromosome Deletion , Bone Diseases, Developmental , Facies
3.
Cell Death Dis ; 15(8): 597, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154024

ABSTRACT

The metastasis-associated protein (MTA) family plays a crucial role in the development of breast cancer, a common malignancy with a high incidence rate among women. However, the mechanism by which each member of the MTA family contributes to breast cancer progression is poorly understood. In this study, we aimed to investigate the roles of MTA1, MTA3, and tripartite motif-containing 21 (TRIM21) in the proliferation, invasion, epithelial-mesenchymal transition (EMT), and stem cell-like properties of breast cancer cells in vivo and in vitro. The molecular mechanisms of the feedback loop between MTA1 and MTA3/TRIM21 regulated by estrogen were explored using Chromatin immunoprecipitation (ChIP), luciferase reporter, immunoprecipitation (IP), and ubiquitination assays. These findings demonstrated that MTA1 acts as a driver to promote the progression of breast cancer by repressing the transcription of tumor suppressor genes, including TRIM21 and MTA3. Conversely, MTA3 inhibited MTA1 transcription and TRIM21 regulated MTA1 protein stability in breast cancer. Estrogen disrupted the balance between MTA1 and MTA3, as well as between MTA1 and TRIM21, thereby affecting stemness and the EMT processes in breast cancer. These findings suggest that MTA1 plays a vital role in stem cell fate and the hierarchical regulatory network of EMT through negative feedback loops with MTA3 or TRIM21 in response to estrogen, supporting MTA1, MTA3, and TRIM21 as potential prognostic biomarkers and MTA1 as a treatment target for future breast cancer therapies.


Subject(s)
Breast Neoplasms , Epithelial-Mesenchymal Transition , Estrogens , Histone Deacetylases , Neoplastic Stem Cells , Repressor Proteins , Trans-Activators , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Repressor Proteins/metabolism , Repressor Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Estrogens/pharmacology , Estrogens/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Animals , Gene Expression Regulation, Neoplastic/drug effects , Mice , Cell Proliferation/drug effects , Cell Line, Tumor , Feedback, Physiological/drug effects , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Mice, Nude , MCF-7 Cells , Mice, Inbred BALB C , Neoplasm Proteins
4.
Front Immunol ; 15: 1444937, 2024.
Article in English | MEDLINE | ID: mdl-39156895

ABSTRACT

Histone deacetylases 1 and 2 play a major role in the transcriptional regulation of T-regulatory (Treg) cells via interactions with a myriad of coregulatory factors. Sin3a has been well established as a Hdac1/2 cofactor, while its role within Tregs has not been established. In this study, the effects of conditional deletion of Sin3a within Foxp3+ Tregs were evaluated. Developmental deletion of Sin3a from Foxp3+ Tregs resulted in the rapid onset of fatal autoimmunity. Treg numbers were greatly reduced, while residual Tregs had impaired suppressive function. Mice also showed effector T-cell activation, autoantibody production, and widespread tissue injury. Mechanistically, Sin3a deletion resulted in decreased transcription of Foxp3 with a complete lack of CNS2 CpG demethylation. In addition, Foxp3 protein stability was impaired with an increased ex-Treg population. Thus, Sin3a plays a critical role in the maintenance of Treg identity and function and is essential for the expression and stability of Foxp3.


Subject(s)
Forkhead Transcription Factors , Sin3 Histone Deacetylase and Corepressor Complex , T-Lymphocytes, Regulatory , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Animals , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Mice , Mice, Knockout , Repressor Proteins/genetics , Repressor Proteins/metabolism , Gene Expression Regulation , Autoimmunity , Mice, Inbred C57BL , Lymphocyte Activation/immunology
6.
Clin Transl Med ; 14(8): e1806, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39143739

ABSTRACT

BACKGROUND: The induction of mitochondrial quality control (MQC) mechanisms is essential for the re-establishment of mitochondrial homeostasis and cellular bioenergetics during periods of stress. Although MQC activation has cardioprotective effects in various cardiovascular diseases, its precise role and regulatory mechanisms in alcoholic cardiomyopathy (ACM) remain incompletely understood. METHODS: We explored whether two mitochondria-related proteins, phosphoglycerate mutase 5 (Pgam5) and prohibitin 2 (Phb2), influence MQC in male mice during ACM. RESULTS: Myocardial Pgam5 expression was upregulated in a male mouse model of ACM. Notably, following ACM induction, heart dysfunction was markedly reversed in male cardiomyocyte-specific Pgam5 knockout (Pgam5cKO) mice. Meanwhile, in alcohol-treated male mouse-derived neonatal cardiomyocytes, Pgam5 depletion preserved cell survival and restored mitochondrial dynamics, mitophagy, mitochondrial biogenesis and the mitochondrial unfolded protein response (mtUPR). We further found that in alcohol-treated cardiomyocyte, Pgam5 binds Phb2 and induces its dephosphorylation at Ser91. Alternative transduction of phospho-mimetic (Phb2S91D) and phospho-defective (Phb2S9A) Phb2 mutants attenuated and enhanced, respectively, alcohol-related mitochondrial dysfunction in cardiomyocytes. Moreover, transgenic male mice expressing Phb2S91D were resistant to alcohol-induced heart dysfunction. CONCLUSIONS: We conclude that ACM-induced Pgam5 upregulation results in Pgam5-dependent Phb2S91 dephosphorylation, leading to MQC destabilisation and mitochondrial dysfunction in heart. Therefore, modulating the Pgam5/Phb2 interaction could potentially offer a novel therapeutic strategy for ACM in male mice. HIGHLIGHTS: Pgam5 knockout attenuates alcohol-induced cardiac histopathology and heart dysfunction in male mice. Pgam5 KO reduces alcohol-induced myocardial inflammation, lipid peroxidation and metabolic dysfunction in male mice. Pgam5 depletion protects mitochondrial function in alcohol-exposed male mouse cardiomyocytes. Pgam5 depletion normalises MQC in ACM. EtOH impairs MQC through inducing Phb2 dephosphorylation at Ser91. Pgam5 interacts with Phb2 and induces Phb2 dephosphorylation. Transgenic mice expressing a Ser91 phospho-mimetic Phb2 mutant are resistant to ACM.


Subject(s)
Cardiomyopathy, Alcoholic , Prohibitins , Repressor Proteins , Animals , Male , Mice , Cardiomyopathy, Alcoholic/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Disease Models, Animal , Phosphorylation , Mitochondria/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/genetics , Myocytes, Cardiac/metabolism , Mice, Knockout
7.
J Hazard Mater ; 477: 135093, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39088948

ABSTRACT

Exposure to particulate matter (PM) can cause airway inflammation and worsen various airway diseases. However, the underlying molecular mechanism by which PM triggers airway inflammation has not been completely elucidated, and effective interventions are lacking. Our study revealed that PM exposure increased the expression of histone deacetylase 9 (HDAC9) in human bronchial epithelial cells and mouse airway epithelium through the METTL3/m6A methylation/IGF2BP3 pathway. Functional assays showed that HDAC9 upregulation promoted PM-induced airway inflammation and activation of MAPK signaling pathway in vitro and in vivo. Mechanistically, HDAC9 modulated the deacetylation of histone 4 acetylation at K12 (H4K12) in the promoter region of dual specificity phosphatase 9 (DUSP9) to repress the expression of DUSP9 and resulting in the activation of MAPK signaling pathway, thereby promoting PM-induced airway inflammation. Additionally, HDAC9 bound to MEF2A to weaken its anti-inflammatory effect on PM-induced airway inflammation. Then, we developed a novel inhaled lipid nanoparticle system for delivering HDAC9 siRNA to the airway, offering an effective treatment for PM-induced airway inflammation. Collectively, we elucidated the crucial regulatory mechanism of HDAC9 in PM-induced airway inflammation and introduced an inhaled therapeutic approach targeting HDAC9. These findings contribute to alleviating the burden of various airway diseases caused by PM exposure.


Subject(s)
Epigenesis, Genetic , Histone Deacetylases , Particulate Matter , Up-Regulation , Animals , Particulate Matter/toxicity , Humans , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Epigenesis, Genetic/drug effects , Up-Regulation/drug effects , Mice , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Inflammation , Nanoparticles/chemistry , Nanoparticles/toxicity , Mice, Inbred C57BL , Cell Line , MAP Kinase Signaling System/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Male
8.
Microbiol Res ; 287: 127859, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39098095

ABSTRACT

Biofilms are common living states for microorganisms, allowing them to adapt to environmental changes. Numerous Bacillus strains can form complex biofilms that play crucial roles in biocontrol processes. However, our current understanding of the molecular mechanisms of biofilm formation in Bacillus is mainly based on studies of Bacillus subtilis. Knowledge regarding the biofilm formation of other Bacillus species remains limited. In this study, we identified a novel transcriptional regulator, BmfR, belonging to the GntR family, that regulates biofilm formation in marine-derived Bacillus methylotrophicus B-9987. We demonstrated that BmfR induces biofilm formation by activating the extracellular polysaccharide structural genes epsA-O and negatively regulating the matrix gene repressor, SinR; of note it positively affects the expression of the master regulator of sporulation, Spo0A. Furthermore, database mining for BmfR homologs has revealed their widespread distribution among many bacterial species, mainly Firmicutes and Proteobacteria. This study advances our understanding of the biofilm regulatory network of Bacillus strains, and provides a new target for exploiting and manipulating biofilm formation.


Subject(s)
Bacillus , Bacterial Proteins , Biofilms , Gene Expression Regulation, Bacterial , Biofilms/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacillus/genetics , Bacillus/physiology , Bacillus/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Polysaccharides, Bacterial/metabolism , Aquatic Organisms/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 121(33): e2401217121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39102544

ABSTRACT

X-linked dystonia-parkinsonism (XDP) is a severe neurodegenerative disorder resulting from an inherited intronic SINE-Alu-VNTR (SVA) retrotransposon in the TAF1 gene that causes dysregulation of TAF1 transcription. The specific mechanism underlying this dysregulation remains unclear, but it is hypothesized to involve the formation of G-quadruplexes (G4) structures within the XDP-SVA that impede transcription. In this study, we show that ZNF91, a critical repressor of SVA retrotransposons, specifically binds to G4-forming DNA sequences. Further, we found that genetic deletion of ZNF91 exacerbates the molecular phenotype associated with the XDP-SVA insertion in patient cells, while no difference was observed when ZNF91 was deleted from isogenic control cells. Additionally, we observed a significant age-related reduction in ZNF91 expression in whole blood and brain, indicating a progressive loss of repression of the XDP-SVA in XDP. These findings indicate that ZNF91 plays a crucial role in controlling the molecular phenotype associated with XDP. Since ZNF91 binds to G4-forming DNA sequences in SVAs, this suggests that interactions between ZNF91 and G4-forming sequences in the XDP-SVA minimize the severity of the molecular phenotype. Our results showing that ZNF91 expression levels significantly decrease with age provide a potential explanation for the age-related progressive neurodegenerative character of XDP. Collectively, our study provides important insights into the protective role of ZNF91 in XDP pathogenesis and suggests that restoring ZNF91 expression, destabilization of G4s, or targeted repression of the XDP-SVA could be future therapeutic strategies to prevent or treat XDP.


Subject(s)
Dystonic Disorders , Genetic Diseases, X-Linked , Phenotype , Humans , Dystonic Disorders/genetics , Dystonic Disorders/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , G-Quadruplexes , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Male , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retroelements/genetics , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism
10.
FASEB J ; 38(16): e23888, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39157983

ABSTRACT

Maf1, originally described as a repressor of RNA polymerase III (RNAP III) transcription in yeast, participates in multiple functions across eukaryotes. However, the knowledge about Maf1 in protozoan parasites is scarce. To initiate the study of Maf1 in Leishmania major, we generated a cell line that overexpresses this protein. Overexpression of Maf1 led to a significant reduction in the abundance of tRNAs, 5S rRNA, and U4 snRNA, demonstrating that Maf1 regulates RNAP III activity in L. major. To further explore the roles played by Maf1 in this microorganism, global transcriptomic and proteomic changes due to Maf1 overexpression were determined using RNA-sequencing and label-free quantitative mass spectrometry. Compared to wild-type cells, differential expression was observed for 1082 transcripts (615 down-regulated and 467 up-regulated) and 205 proteins (132 down-regulated and 73 up-regulated) in the overexpressing cells. A correlation of 44% was found between transcriptomic and proteomic results. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that the differentially expressed genes and proteins are mainly involved in transcription, cell cycle regulation, lipid metabolism and transport, ribosomal biogenesis, carbohydrate metabolism, autophagy, and cytoskeleton modification. Thus, our results suggest the involvement of Maf1 in the regulation of all these processes in L. major, as reported in other species, indicating that the functions performed by Maf1 were established early in eukaryotic evolution. Notably, our data also suggest the participation of L. major Maf1 in mRNA post-transcriptional control, a role that, to the best of our knowledge, has not been described in other organisms.


Subject(s)
Leishmania major , Proteome , Transcriptome , Leishmania major/metabolism , Leishmania major/genetics , Proteome/metabolism , Humans , RNA Polymerase III/metabolism , RNA Polymerase III/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Gene Expression Regulation
11.
Epigenetics ; 19(1): 2381849, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39109527

ABSTRACT

Gametogenetin binding protein 2 (GGNBP2) was indispensable in normal spermatids for transformation into mature spermatozoa in mice, and when Gametogenetin binding protein 2 is bound to BRCC36 and RAD51, the complex participates in repairing DNA double-strand breaks (DSB) during the meiotic progression of spermatocytes. Ggnbp2 knockout resulted in the up-regulation of H2AK119ubi and down-regulation of H2BK120ubi in GC-2 cells (mouse spermatogonia-derived cell line) and postnatal day 18 testis lysate. Our results also demonstrated that Gametogenetin binding protein 2 inducedASXL1 to activate the deubiquitinating enzyme BAP1 in deubiquitinating H2A, while Gametogenetin binding protein 2 knockout disrupted the interaction between ASXL1 and BAP1, resulting in BAP1 localization change. Furthermore, the Gametogenetin binding protein 2 deletion reduced H2B ubiquitination by affecting E2 enzymes and E3 ligase binding. Gametogenetin binding protein 2 regulated H2A and H2B ubiquitination levels and controlled H3K27 and H3K79 methylation by PRC2 subunits and histone H3K79 methyltransferase. Altogether, our results suggest that Ggnbp2 knockout increased DNA damage response by promoting H2A ubiquitination and H3K27trimethylation (H3K27me3) and reduced nucleosome stability by decreasing H2B ubiquitination and H3K79 dimethylation (H3K79me2), revealing new mechanisms of epigenetic phenomenon during spermatogenesis. Gametogenetin binding protein 2 seems critical in regulating histone modification and chromatin structure in spermatogenesis.


Subject(s)
Histones , Spermatogenesis , Ubiquitination , Male , Animals , Spermatogenesis/genetics , Histones/metabolism , Mice , Methylation , Mice, Knockout , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Cell Line
12.
J Exp Clin Cancer Res ; 43(1): 217, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39098911

ABSTRACT

Aberrant alternative splicing events play a critical role in cancer biology, contributing to tumor invasion, metastasis, epithelial-mesenchymal transition, and drug resistance. Recent studies have shown that alternative splicing is a key feature for transcriptomic variations in colorectal cancer, which ranks third among malignant tumors worldwide in both incidence and mortality. Long non-coding RNAs can modulate this process by acting as trans-regulatory agents, recruiting splicing factors, or driving them to specific targeted genes. LncH19 is a lncRNA dis-regulated in several tumor types and, in colorectal cancer, it plays a critical role in tumor onset, progression, and metastasis. In this paper, we found, that in colorectal cancer cells, the long non-coding RNA H19 can bind immature RNAs and splicing factors as hnRNPM and RBFOX2. Through bioinformatic analysis, we identified 57 transcripts associated with lncH19 and containing binding sites for both splicing factors, hnRNPM, and RBFOX2. Among these transcripts, we identified the mRNA of the GTPase-RAC1, whose alternatively spliced isoform, RAC1B, has been ascribed several roles in the malignant transformation. We confirmed, in vitro, the binding of the splicing factors to both the transcripts RAC1 and lncH19. Loss and gain of expression experiments in two colorectal cancer cell lines (SW620 and HCT116) demonstrated that lncH19 is required for RAC1B expression and, through RAC1B, it induces c-Myc and Cyclin-D increase. In vivo, investigation from biopsies of colorectal cancer patients showed higher levels of all the explored genes (lncH19, RAC1B, c-Myc and Cyclin-D) concerning the healthy counterpart, thus supporting our in vitro model. In addition, we identified a positive correlation between lncH19 and RAC1B in colorectal cancer patients. Finally, we demonstrated that lncH19, as a shuttle, drives the splicing factors RBFOX2 and hnRNPM to RAC1 allowing exon retention and RAC1B expression. The data shown in this paper represent the first evidence of a new mechanism of action by which lncH19 carries out its functions as an oncogene by prompting colorectal cancer through the modulation of alternative splicing.


Subject(s)
Alternative Splicing , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , RNA Splicing Factors , RNA, Long Noncoding , rac1 GTP-Binding Protein , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , RNA, Long Noncoding/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Cell Line, Tumor , Heterogeneous-Nuclear Ribonucleoprotein Group M/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group M/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
14.
Head Neck Pathol ; 18(1): 66, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39101978

ABSTRACT

BACKGROUND: ETV6 gene rearrangement is the molecular hallmark of secretory carcinoma (SC), however; the nature, frequency, and clinical implications of atypical ETV6 signal patterns by fluorescence in situ hybridization (FISH) has not yet been systematically evaluated in salivary gland neoplasms. METHODS: The clinical, histopathologic, immunohistochemical and molecular features of seven salivary SCs, including four cases with atypical ETV6 FISH patterns, were retrospectively analyzed along with a critical appraisal of the literature on unbalanced ETV6 break-apart in SCs. RESULTS: The patients were four males and three females (31-70 years-old). Five presented with a painless neck mass and two patients with recurrent disease had a history of a previously diagnosed acinic cell carcinoma of the buccal mucosa. Histologically, there were varied combinations of microcystic, papillary, tubular, and solid patterns. All tumors were diffusely positive for S100 and/or SOX10, while 2 cases also showed luminal DOG1 staining. Rearrangement of the ETV6 locus was confirmed in 5/7 cases, of which 3 cases showed classic break-apart signals, 1 case further demonstrated duplication of the ETV6 5`end and the other loss of one copy of ETV6. Two cases harbored ETV6 deletion without rearrangement. Two of the 4 cases with atypical ETV6 FISH patterns represented recurrent tumors, one with widespread skeletal muscle involvement, bone and lymphovascular invasion. Surgical treatment resulted in gross-total resection in all 7 cases, with a median follow up of 9.5 months post-surgery for primary (n = 3) and recurrent disease (n = 1). CONCLUSION: Duplication of the distal/telomeric ETV6 probe represented the most common (26/40; 65%) variant ETV6 break-apart FISH pattern in salivary SC reported in the literature and appears indicative of an aggressive clinical course.


Subject(s)
ETS Translocation Variant 6 Protein , Proto-Oncogene Proteins c-ets , Repressor Proteins , Salivary Gland Neoplasms , Humans , Male , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Proto-Oncogene Proteins c-ets/genetics , Female , Adult , Aged , Middle Aged , Repressor Proteins/genetics , In Situ Hybridization, Fluorescence , Gene Rearrangement , Carcinoma/genetics , Carcinoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis
15.
Proc Natl Acad Sci U S A ; 121(34): e2404738121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39141353

ABSTRACT

Most mammalian cells have molecular circadian clocks that generate widespread rhythms in transcript and protein abundance. While circadian clocks are robust to fluctuations in the cellular environment, little is known about the mechanisms by which the circadian period compensates for fluctuating metabolic states. Here, we exploit the heterogeneity of single cells both in circadian period and a metabolic parameter-protein stability-to study their interdependence without the need for genetic manipulation. We generated cells expressing key circadian proteins (CRYPTOCHROME1/2 (CRY1/2) and PERIOD1/2 (PER1/2)) as endogenous fusions with fluorescent proteins and simultaneously monitored circadian rhythms and degradation in thousands of single cells. We found that the circadian period compensates for fluctuations in the turnover rates of circadian repressor proteins and uncovered possible mechanisms using a mathematical model. In addition, the stabilities of the repressor proteins are circadian phase dependent and correlate with the circadian period in a phase-dependent manner, in contrast to the prevailing model.


Subject(s)
Circadian Rhythm , Cryptochromes , Period Circadian Proteins , Single-Cell Analysis , Period Circadian Proteins/metabolism , Period Circadian Proteins/genetics , Circadian Rhythm/physiology , Cryptochromes/metabolism , Cryptochromes/genetics , Animals , Repressor Proteins/metabolism , Repressor Proteins/genetics , Circadian Clocks/physiology , Humans , Mice , Protein Stability
16.
BMC Med Genomics ; 17(1): 195, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103808

ABSTRACT

BACKGROUND: The Cut Homeobox 1 (CUX1) gene has been implicated in a number of developmental processes and has recently emerged as an important cause of developmental delay and impaired intellectual development. Individuals with variants in CUX1 have been described with a variety of co-morbidities including variations in sex development (VSD) although these features have not been closely documented. CASE PRESENTATION: The proband is a 14-year-old male who presented with congenital complex hypospadias, neurodevelopmental differences, and subtle dysmorphism. A family history of neurodevelopmental differences and VSD was noted. Microarray testing and whole exome sequencing found the 46,XY proband had a large heterozygous in-frame deletion of exons 4-10 of the CUX1 gene. CONCLUSIONS: Our review of the literature has revealed that variants in CUX1 are associated with a range of VSD and suggest this gene should be considered in cases where a VSD is noted at birth, especially if there is a familial history of VSD and/or neurodevelopmental differences. Further work is required to fully investigate the role and regulation of CUX1 in sex development.


Subject(s)
Homeodomain Proteins , Repressor Proteins , Humans , Male , Homeodomain Proteins/genetics , Adolescent , Repressor Proteins/genetics , Transcription Factors/genetics , Sexual Development/genetics
17.
Clin Epigenetics ; 16(1): 102, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097736

ABSTRACT

BACKGROUND: Oesophageal cancer remains a challenging disease with high mortality rates and few therapeutic options. In view of these difficulties, epigenetic drugs have emerged as potential alternatives for patient care. The goal of this study was to evaluate the effect and biological consequences of Panobinostat treatment, an HDAC (histone deacetylase) inhibitor already approved for treatment of patients with multiple myeloma, in oesophageal cell lines of normal and malignant origin, with the latter being representative of the two main histological subtypes: adenocarcinoma and squamous cell carcinoma. RESULTS: Panobinostat treatment inhibited growth and hindered proliferation, colony formation and invasion of oesophageal cancer cells. Considering HDAC tissue expression, HDAC1 was significantly upregulated in normal oesophageal epithelium in comparison with tumour tissue, whereas HDAC3 was overexpressed in oesophageal cancer compared to non-malignant mucosa. No differences between normal and tumour tissue were observed for HDAC2 and HDAC8 expression. CONCLUSIONS: Panobinostat exposure effectively impaired malignant features of oesophageal cancer cells. Because HDAC3 was shown to be overexpressed in oesophageal tumour samples, this epigenetic drug may represent an alternative therapeutic option for oesophageal cancer patients.


Subject(s)
Adenocarcinoma , Carcinoma, Squamous Cell , Cell Proliferation , Esophageal Neoplasms , Histone Deacetylase Inhibitors , Histone Deacetylases , Panobinostat , Humans , Panobinostat/pharmacology , Panobinostat/therapeutic use , Panobinostat/administration & dosage , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Cell Line, Tumor , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Cell Proliferation/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Repressor Proteins/genetics , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology
18.
Oncol Res ; 32(7): 1185-1195, 2024.
Article in English | MEDLINE | ID: mdl-38948024

ABSTRACT

Background: Long non-coding RNAs are important regulators in cancer biology and function either as tumor suppressors or as oncogenes. Their dysregulation has been closely associated with tumorigenesis. LINC00265 is upregulated in lung adenocarcinoma and is a prognostic biomarker of this cancer. However, the mechanism underlying its function in cancer progression remains poorly understood. Methods: Here, the regulatory role of LINC00265 in lung adenocarcinoma was examined using lung cancer cell lines, clinical samples, and xenografts. Results: We found that high levels of LINC00265 expression were associated with shorter overall survival rate of patients, whereas knockdown of LINC00265 inhibited proliferation of cancer cell lines and tumor growth in xenografts. Western blot and flow cytometry analyses indicated that silencing of LINC00265 induced autophagy and apoptosis. Moreover, we showed that LINC00265 interacted with and stabilized the transcriptional co-repressor Switch-independent 3a (SIN3A), which is a scaffold protein functioning either as a tumor repressor or as an oncogene in a context-dependent manner. Silencing of SIN3A also reduced proliferation of lung cancer cells, which was correlated with the induction of autophagy. These observations raise the possibility that LINC00265 functions to promote the oncogenic activity of SIN3A in lung adenocarcinoma. Conclusions: Our findings thus identify SIN3A as a LINC00265-associated protein and should help to understand the mechanism underlying LINC00265-mediated oncogenesis.


Subject(s)
Apoptosis , Autophagy , Cell Proliferation , Lung Neoplasms , RNA, Long Noncoding , Sin3 Histone Deacetylase and Corepressor Complex , Humans , RNA, Long Noncoding/genetics , Autophagy/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Apoptosis/genetics , Animals , Mice , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Repressor Proteins/genetics , Repressor Proteins/metabolism , Gene Expression Regulation, Neoplastic , Protein Stability , Gene Silencing , Oncogenes , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Xenograft Model Antitumor Assays
19.
Virol J ; 21(1): 152, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970084

ABSTRACT

BACKGROUND: High-risk human papillomavirus (HR-HPV) infection is an important factor for the development of cervical cancer. HPV18 is the second most common HR-HPV after HPV16. METHODS: In this study, MEGA11 software was used to analyze the variation and phylogenetic tree of HPV18 E6-E7 and L1 genes. The selective pressure to E6, E7 and L1 genes was estimated using pamlX. In addition, the B cell epitopes of L1 amino acid sequences and T cell epitopes of E6-E7 amino acid sequences in HPV18 were predicted by ABCpred server and IEDB website, respectively. RESULTS: A total of 9 single nucleotide variants were found in E6-E7 sequences, of which 2 were nonsynonymous variants and 7 were synonymous variants. Twenty single nucleotide variants were identified in L1 sequence, including 11 nonsynonymous variants and 9 synonymous variants. Phylogenetic analysis showed that E6-E7 and L1 sequences were all distributed in A lineage. In HPV18 E6, E7 and L1 sequences, no positively selected site was found. The nonconservative substitution R545C in L1 affected hypothetical B cell epitope. Two nonconservative substitutions, S82A in E6, and R53Q in E7, impacted multiple hypothetical T cell epitopes. CONCLUSION: The sequence variation data of HPV18 may lay a foundation for the virus diagnosis, further study of cervical cancer and vaccine design in central China.


Subject(s)
Genetic Variation , Human papillomavirus 18 , Oncogene Proteins, Viral , Papillomavirus E7 Proteins , Phylogeny , Oncogene Proteins, Viral/genetics , China , Humans , Human papillomavirus 18/genetics , Human papillomavirus 18/classification , Papillomavirus E7 Proteins/genetics , Capsid Proteins/genetics , Female , Epitopes, T-Lymphocyte/genetics , Papillomavirus Infections/virology , Repressor Proteins/genetics , Epitopes, B-Lymphocyte/genetics , DNA-Binding Proteins
20.
Protein Sci ; 33(8): e5118, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39022984

ABSTRACT

Proper protein arginine methylation by protein arginine methyltransferase 1 (PRMT1) is critical for maintaining cellular health, while dysregulation is often associated with disease. How the activity of PRMT1 is regulated is therefore paramount, but is not clearly understood. Several studies have observed higher order oligomeric species of PRMT1, but it is unclear if these exist at physiological concentrations and there is confusion in the literature about how oligomerization affects activity. We therefore sought to determine which oligomeric species of PRMT1 are physiologically relevant, and quantitatively correlate activity with specific oligomer forms. Through quantitative western blotting, we determined that concentrations of PRMT1 available in a variety of human cell lines are in the sub-micromolar to low micromolar range. Isothermal spectral shift binding data were modeled to a monomer/dimer/tetramer equilibrium with an EC50 for tetramer dissociation of ~20 nM. A combination of sedimentation velocity and Native polyacrylamide gel electrophoresis experiments directly confirmed that the major oligomeric species of PRMT1 at physiological concentrations would be dimers and tetramers. Surprisingly, the methyltransferase activity of a dimeric PRMT1 variant is similar to wild type, tetrameric PRMT1 with some purified substrates, but dimer and tetramer forms of PRMT1 show differences in catalytic efficiencies and substrate specificity for other substrates. Our results define an oligomerization paradigm for PRMT1, show that the biophysical characteristics of PRMT1 are poised to support a monomer/dimer/tetramer equilibrium in vivo, and suggest that the oligomeric state of PRMT1 could be used to regulate substrate specificity.


Subject(s)
Protein Multimerization , Protein-Arginine N-Methyltransferases , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Humans , Substrate Specificity , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Repressor Proteins/genetics , Methylation
SELECTION OF CITATIONS
SEARCH DETAIL