Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.931
Filter
1.
Blood ; 144(14): 1465-1467, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39361299
2.
J Biochem Mol Toxicol ; 38(10): e23864, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39318028

ABSTRACT

Previous research has indicated the highly expressed lysine-specific histone demethylase 1A (KDM1A) in several human malignancies, including triple-negative breast cancer (TNBC). However, its detailed mechanisms in TNBC development remain poorly understood. The mRNA levels of KDM1A and Yin Yang 1 (YY1) were determined by RT-qPCR analysis. Western blot was performed to measure KDM1A and ubiquitin-specific protease 1 (USP1) protein expression. Cell proliferation, apoptosis, invasion, migration and stemness were evaluated by MTT assay, EdU assay, flow cytometry, transwell invasion assay, wound-healing assay and sphere-formation assay, respectively. ChIP and dual-luciferase reporter assays were conducted to determine the relationship between YY1 and KDM1A. Xenograft tumor experiment and IHC were carried out to investigate the roles of USP1 and KDM1A in TNBC development in vivo. The highly expressed KDM1A was demonstrated in TNBC tissues and cells, and KDM1A knockdown significantly promoted cell apoptosis, and hampered cell proliferation, invasion, migration, and stemness in TNBC cells. USP1 could increase the stability of KDM1A via deubiquitination, and USP1 depletion restrained the progression of TNBC cells through decreasing KDM1A expression. Moreover, YY1 transcriptionally activated KDM1A expression by directly binding to its promoter in TNBC cells. Additionally, USP1 inhibition reduced KDM1A expression to suppress tumor growth in TNBC mice in vivo. In conclusion, YY1 upregulation increased KDM1A expression via transcriptional activation. USP1 stabilized KDM1A through deubiquitination to promote TNBC progression.


Subject(s)
Histone Demethylases , Triple Negative Breast Neoplasms , Ubiquitin-Specific Proteases , Ubiquitination , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/genetics , Humans , Female , Animals , Cell Line, Tumor , Mice , Histone Demethylases/metabolism , Histone Demethylases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics , Disease Progression , Cell Proliferation , Mice, Nude , YY1 Transcription Factor/metabolism , YY1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , Apoptosis , Cell Movement
3.
J Med Chem ; 67(18): 16165-16184, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39264726

ABSTRACT

LSD1 (histone lysine-specific demethylase 1) has been gradually disclosed to act as an immunomodulator to enhance antitumor immune response. Despite the identification of numerous potent LSD1 inhibitors, there remains a lack of LSD1 inhibitors approved for marketing. Novel LSD1 inhibitors with different mechanisms are therefore needed. Herein, we reported a series of novel quinazoline-based LSD1 inhibitors. Among them, compound Z-1 exhibited the best LSD1 inhibitory activity (IC50 = 0.108 µM). Z-1 also acted as a selective and cellular active as an LSD1 inhibitor. Furthermore, Z-1 promoted response of gastric cancer cells to T-cell killing effect by decreasing PD-L1 expression and further attenuated the PD-1/PD-L1 interaction. In vivo, Z-1 exhibited significant suppression effect on the growth of gastric cancer cells without obvious toxicity. Therefore, Z-1 represents a potential novel immunomodulator that targets LSD1, providing a lead compound with new function mechanism for gastric cancer treatment.


Subject(s)
Histone Demethylases , Stomach Neoplasms , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Structure-Activity Relationship , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Quinazolines/pharmacology , Quinazolines/chemistry , Quinazolines/chemical synthesis , Mice , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Drug Discovery , Molecular Docking Simulation
4.
Nat Commun ; 15(1): 7758, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237615

ABSTRACT

Lysine-specific histone demethylase 1 (LSD1), which demethylates mono- or di- methylated histone H3 on lysine 4 (H3K4me1/2), is essential for early embryogenesis and development. Here we show that LSD1 is dispensable for mouse embryonic stem cell (ESC) self-renewal but is required for mouse ESC growth and differentiation. Reintroduction of a catalytically-impaired LSD1 (LSD1MUT) recovers the proliferation capability of mouse ESCs, yet the enzymatic activity of LSD1 is essential to ensure proper differentiation. Indeed, increased H3K4me1 in Lsd1 knockout (KO) mouse ESCs does not lead to major changes in global gene expression programs related to stemness. However, ablation of LSD1 but not LSD1MUT results in decreased DNMT1 and UHRF1 proteins coupled to global hypomethylation. We show that both LSD1 and LSD1MUT control protein stability of UHRF1 and DNMT1 through interaction with HDAC1 and the ubiquitin-specific peptidase 7 (USP7), consequently, facilitating the deacetylation and deubiquitination of DNMT1 and UHRF1. Our studies elucidate a mechanism by which LSD1 controls DNA methylation in mouse ESCs, independently of its lysine demethylase activity.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Histone Demethylases , Mice, Knockout , Mouse Embryonic Stem Cells , Ubiquitin-Protein Ligases , Animals , Histone Demethylases/metabolism , Histone Demethylases/genetics , Mice , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Mouse Embryonic Stem Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Histones/metabolism , Cell Proliferation , Ubiquitination
5.
Eur J Med Chem ; 278: 116801, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39241481

ABSTRACT

The rational installation of pharmacophores targeting HSP90 and LSD1 axes has achieved significant anti-cancer capacity in prostate and colorectal cancer. Among the series of hybrids, inhibitor 6 exhibited remarkable anti-proliferative activity against prostate cancer cell lines PC-3 and DU145, with GI50 values of 0.24 and 0.30 µM, respectively. It demonstrated notable efficacy in combinatorial attack and cell death initiation towards apoptosis. The cell death process was mediated by PARP induction and γH2AX signaling, and was also characterized as caspase-dependent and Bcl-xL/Bax-independent. Notably, no difference in eye size or morphology was observed in the zebrafish treated with compound 6 compared to the reference group (AUY922). The profound treatment response in docetaxel-resistant PC-3 cells highlighted the dual inhibitory ability in improving docetaxel sensitivity. Additionally, at a minimum concentration of 1.25 µM, compound 6 effectively inhibited the growth of patient-derived colorectal cancer (CRC) organoids for up to 10 days in vitro. Together, the designed HSP90/LSD1 inhibitors present a novel route and significant clinical value for anti-cancer drug therapy.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Colorectal Neoplasms , Drug Screening Assays, Antitumor , HSP90 Heat-Shock Proteins , Histone Demethylases , Organoids , Prostatic Neoplasms , Animals , Humans , Male , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Dose-Response Relationship, Drug , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Molecular Structure , Organoids/drug effects , Organoids/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Structure-Activity Relationship , Zebrafish
6.
Sci Rep ; 14(1): 20817, 2024 09 06.
Article in English | MEDLINE | ID: mdl-39242625

ABSTRACT

Metastasis accounts for almost 90% of breast cancer-related fatalities, making it frequent malignancy and the main reason of tumor mortality globally among women. LSD1 is a histone demethylase, which plays an important role in breast cancer. In order to explore the effect of LSD1 on invasion and migration of breast cancer, we treated breast cancer cells with MCF7 and T47D exosomes knocked down by LSD1, and the invasion and migration of breast cancer cells were significantly enhanced. This phenomenon indicates that LSD1 can inhibit the invasion and migration of breast cancer cells. miR-1290 expression was downregulated in LSD1 knockdown MCF7 exosomes. By analyzing the database of miR-1290 target gene NAT1, we verified that miR-1290 could regulate the expression of NAT1. These data provide fresh insights into the biology of breast cancer therapy by demonstrating how the epigenetic factor LSD1 stimulates the breast cancer cells' invasion and migration via controlling exosomal miRNA.


Subject(s)
Breast Neoplasms , Cell Movement , Exosomes , Gene Expression Regulation, Neoplastic , Histone Demethylases , MicroRNAs , Neoplasm Invasiveness , Humans , Histone Demethylases/metabolism , Histone Demethylases/genetics , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Exosomes/metabolism , Cell Movement/genetics , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , MCF-7 Cells
7.
Proc Natl Acad Sci U S A ; 121(40): e2321182121, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39325426

ABSTRACT

Senescence is a cell fate driven by different types of stress that results in exit from the cell cycle and expression of an inflammatory senescence-associated secretory phenotype (SASP). Here, we demonstrate that stable overexpression of miR-96-5p was sufficient to induce cellular senescence in the absence of genotoxic stress, inducing expression of certain markers of early senescence including SASP factors while repressing markers of deep senescence including LINE-1 and type 1 interferons. Stable miR-96-5p overexpression led to genome-wide changes in heterochromatin followed by epigenetic activation of p16Ink4a, p21Cip1, and SASP expression, induction of a marker of DNA damage, and induction of a transcriptional signature similar to other senescent lung and endothelial cell types. Expression of miR-96-5p significantly increased following senescence induction in culture cells and with aging in tissues from naturally aged and Ercc1-/Δ progeroid mice. Mechanistically, miR-96-5p directly suppressed expression of SIN3B and SIN3 corepressor complex constituents KDM5A and MORF4L2, and siRNA-mediated knockdown of these transcriptional regulators recapitulated the senescent phenotype. In addition, pharmacologic inhibition of the SIN3 complex suppressed senescence and SASP markers. These results clearly demonstrate that a single microRNA is sufficient to drive early senescence in the absence of genotoxic stress through targeting epigenetic and transcriptional regulators, identifying novel targets for the development of senotherapeutics.


Subject(s)
Cellular Senescence , DNA Damage , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Cellular Senescence/genetics , Mice , Humans , Repressor Proteins/metabolism , Repressor Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Senescence-Associated Secretory Phenotype/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Heterochromatin/metabolism , Heterochromatin/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Epigenesis, Genetic , Histone Demethylases/metabolism , Histone Demethylases/genetics , Gene Expression Regulation , Endonucleases
8.
FASEB J ; 38(18): e70072, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39323294

ABSTRACT

The inability to efficiently metabolize homocysteine (Hcy) due to nutritional and genetic deficiencies, leads to hyperhomocysteinemia (HHcy) and endothelial dysfunction, a hallmark of atherosclerosis which underpins cardiovascular disease (CVD). PHF8 is a histone demethylase that demethylates H4K20me1, which affects the mammalian target of rapamycin (mTOR) signaling and autophagy, processes that play important roles in CVD. PHF8 is regulated by microRNA (miR) such as miR-22-3p and miR-1229-3p. Biochemically, HHcy is characterized by elevated levels of Hcy, Hcy-thiolactone and N-Hcy-protein. Here, we examined the effects of these metabolites on miR-22-3p, miR-1229-3p, and their target PHF8, as well as on the downstream consequences of these effects on H4K20me1, mTOR-, and autophagy-related proteins and mRNAs expression in human umbilical vein endothelial cells (HUVEC). We found that treatments with N-Hcy-protein, Hcy-thiolactone, or Hcy upregulated miR-22-3p and miR-1229-3p, attenuated PHF8 expression, upregulated H4K20me1, mTOR, and phospho-mTOR. Autophagy-related proteins (BECN1, ATG5, ATG7, lipidated LC3-II, and LC3-II/LC3-I ratio) were significantly downregulated by at least one of these metabolites. We also found similar changes in the expression of miR-22-3p, Phf8, mTOR- and autophagy-related proteins/mRNAs in vivo in hearts of Cbs-/- mice, which show severe HHcy and endothelial dysfunction. Treatments with inhibitors of miR-22-3p or miR-1229-3p abrogated the effects of Hcy-thiolactone, N-Hcy-protein, and Hcy on miR expression and on PHF8, H4K20me1, mTOR-, and autophagy-related proteins/mRNAs in HUVEC. Taken together, these findings show that Hcy metabolites upregulate miR-22-3p and miR-1229-3p expression, which then dysregulate the PHF8/H4K20me1/mTOR/autophagy pathway, important for vascular homeostasis.


Subject(s)
Autophagy , Histone Demethylases , Homocysteine , Human Umbilical Vein Endothelial Cells , MicroRNAs , TOR Serine-Threonine Kinases , Transcription Factors , Animals , Female , Humans , Mice , Histone Demethylases/metabolism , Histone Demethylases/genetics , Homocysteine/metabolism , Homocysteine/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Up-Regulation
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1620-1630, 2024 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-39276059

ABSTRACT

OBJECTIVE: To explore the inhibitory effect ORY-1001, a lysine-specific histone demethylase 1 (LSD1) inhibitor, on growth of glioblastoma (GBM) and the underlying mechanism. METHODS: We analyzed LSD1 expressions in GBM and normal brain tissues based on data from TCGA and HPA databases. Female BALB/c mouse models bearing xenografts derived from U87 cells or cells with lentivirus-mediated LSD1 silencing or Notch overexpression were treated with saline or 400 µg/kg ORY-1001 by gavage every 7 days, and GBM formation and survival time of the mice were recorded. The effect of ORY-1001 on GBM cell viability was assessed, and its effect on LSD1 expression was analyzed with Western blotting. The genes and pathways associated with LSD1 were analyzed using bioinformatics methods. Western blotting and qRT-PCR were used to detect Notch/HES1 pathway expression after LSD1 silencing and ORY-1001 treatment. The impact of ORY-1001 on viability of U87 cells with Notch1 silencing or overexpression was assessed, and the regulatory effects of ORY-1001 on Notch/HES1 pathway were analyzed using chromatin immunoprecipitation assay. RESULTS: A high expression of LSD1 in GBM was negatively correlated with patient survival (P < 0.001). ORY-1001 and LSD1 silencing obviously reduced tumor burden and prolonged the survival time of GBM-bearing mice. ORY-1001 treatment significantly inhibited the viability and dose-dependently decreased LSD1 expression in GBM cells, and such inhibitory effect of ORY-1001 was attenuated by LSD1 silencing. The Notch pathway enriched the differential genes related to LSD1, and Notch/HES1 pathway expression was significantly down-regulated after LSD1 silencing and ORY-1001 treatment. Notch1 overexpression significantly attenuated the anti-tumor effect of ORY-1001 on GBM. Mechanistically, ORY-1001 disrupted the interaction between LSD1 and the Notch pathway target genes including Notch3, HES1 and CR2. CONCLUSION: ORY-1001 down-regulates the Notch/HES1 pathway by inhibiting LSD1 expression to suppress the growth of GBM in mice.


Subject(s)
Cell Proliferation , Glioblastoma , Histone Demethylases , Mice, Inbred BALB C , Transcription Factor HES-1 , Histone Demethylases/metabolism , Histone Demethylases/genetics , Animals , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Mice , Cell Line, Tumor , Female , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Humans , Cell Proliferation/drug effects , Signal Transduction , Receptors, Notch/metabolism , Down-Regulation , Brain Neoplasms/metabolism , Brain Neoplasms/pathology
10.
J Clin Immunol ; 45(1): 7, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39264387

ABSTRACT

OBJECTIVE: To analyze the lymphocyte subsets in individuals with Kabuki syndrome for better characterizing the immunological phenotype of this rare congenital disorder. METHODS: We characterized the immunological profile including B-, T- and natural killer-cell subsets in a series (N = 18) of individuals with Kabuki syndrome. RESULTS: All 18 individuals underwent genetic analysis: 15 had a variant in KMT2D and 3 a variant in KDM6A. Eleven of the 18 individuals (61%) had recurrent infections and 9 (50%) respiratory infections. Three (17%) had autoimmune diseases. On immunological analysis, 6 (33%) had CD4 T-cell lymphopenia, which was preferentially associated with the KMT2D truncating variant (5/9 individuals). Eight of 18 individuals (44%) had a humoral deficiency and eight (44%) had B lymphopenia. We found abnormal distributions of T-cell subsets, especially a frequent decrease in recent thymic emigrant CD4 + naive T-cell count in 13/16 individuals (81%). CONCLUSION: The immunological features of Kabuki syndrome showed variable immune disorders with CD4 + T-cell deficiency in one third of cases, which had not been previously reported. In particular, we found a reduction in recent thymic emigrant naïve CD4 + T-cell count in 13 of 16 individuals, representing a novel finding that had not previously been reported.


Subject(s)
Abnormalities, Multiple , DNA-Binding Proteins , Face , Histone Demethylases , Neoplasm Proteins , Vestibular Diseases , Humans , Vestibular Diseases/genetics , Vestibular Diseases/immunology , Face/abnormalities , Female , Male , Abnormalities, Multiple/genetics , Abnormalities, Multiple/immunology , Child , DNA-Binding Proteins/genetics , Adolescent , Histone Demethylases/genetics , Child, Preschool , Adult , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Young Adult , Infant , Lymphopenia/immunology , Lymphopenia/genetics , Phenotype , Hematologic Diseases/genetics , Hematologic Diseases/immunology , Mutation , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Immunophenotyping
11.
Int J Med Sci ; 21(11): 2158-2169, 2024.
Article in English | MEDLINE | ID: mdl-39239542

ABSTRACT

Pancreatic cancer (PC) is a challenging and heterogeneous disease with a high mortality rate. Despite advancements in treatment, the prognosis for PC patients remains poor, with a high chance of disease recurrence. Biomarkers are crucial for diagnosing cancer, predicting patient prognosis and selecting treatments. However, the current lack of effective biomarkers for PC could contribute to the insufficiency of existing treatments. These findings underscore the urgent need to develop novel strategies to fight this disease. This study utilized multiple comprehensive bioinformatic analyses to identify potential therapeutic target genes in PC, focusing on histone lysine demethylases (KDMs). We found that high expression levels of KDM family genes, particularly KDM1A, KDM5A and KDM5B, were associated with improved overall survival in the cohort. Furthermore, the infiltration of various immune cells, including B cells, neutrophils, CD8+ T cells, dendritic cells, and macrophages, was positively correlated with KDM1A, KDM5A, and KDM5B expression. Moreover, MetaCore pathway analysis revealed interesting connections between KDM1A and the cell cycle and proliferation, between KDM5A and DNA damage and double-strand break repair through homologous recombination, and between KDM5B and WNT/ß-catenin signaling. These findings suggest that KDM1A, KDM5A and KDM5B may serve as promising biomarkers and therapeutic targets for PC, a disease of high importance due to its aggressive nature and urgent need for novel biomarkers to improve diagnosis and treatment.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Prognosis , Computational Biology , F-Box Proteins/metabolism , F-Box Proteins/genetics , Histone Demethylases/metabolism , Histone Demethylases/genetics , Molecular Targeted Therapy/methods , Retinoblastoma-Binding Protein 2/metabolism , Retinoblastoma-Binding Protein 2/genetics , Wnt Signaling Pathway/genetics , Cell Proliferation/genetics , Nuclear Proteins , Repressor Proteins
12.
Cells ; 13(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39273051

ABSTRACT

Intervertebral disc disease (IDD) is a debilitating spine condition that can be caused by intervertebral disc (IVD) damage which progresses towards IVD degeneration and dysfunction. Recently, human pluripotent stem cells (hPSCs) were recognized as a valuable resource for cell-based regenerative medicine in skeletal diseases. Therefore, adult somatic cells reprogrammed into human induced pluripotent stem cells (hiPSCs) represent an attractive cell source for the derivation of notochordal-like cells (NCs) as a first step towards the development of a regenerative therapy for IDD. Utilizing a differentiation method involving treatment with a four-factor cocktail targeting the BMP, FGF, retinoic acid, and Wnt signaling pathways, we differentiate CRISPR/Cas9-generated mCherry-reporter knock-in hiPSCs into notochordal-like cells. Comprehensive analysis of transcriptomic changes throughout the differentiation process identified regulation of histone methylation as a pivotal driver facilitating the differentiation of hiPSCs into notochordal-like cells. We further provide evidence that specific inhibition of histone demethylases KDM2A and KDM7A/B enhanced the lineage commitment of hiPSCs towards notochordal-like cells. Our results suggest that inhibition of KDMs could be leveraged to alter the epigenetic landscape of hiPSCs to control notochord-specific gene expression. Thus, our study highlights the importance of epigenetic regulators in stem cell-based regenerative approaches for the treatment of disc degeneration.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Jumonji Domain-Containing Histone Demethylases , Notochord , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Notochord/metabolism , Notochord/cytology , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Histone Demethylases/metabolism , Histone Demethylases/genetics , F-Box Proteins
13.
Eur J Endocrinol ; 191(3): 334-344, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39171930

ABSTRACT

BACKGROUND: Primary bilateral macronodular adrenal hyperplasia (PBMAH) is a rare cause of Cushing's syndrome. Individuals with PBMAH and glucose-dependent insulinotropic polypeptide (GIP)-dependent Cushing's syndrome due to ectopic expression of the GIP receptor (GIPR) typically harbor inactivating KDM1A sequence variants. Primary unilateral macronodular adrenal hyperplasia (PUMAH) with concomitant glucocorticoid and androgen excess has never been encountered or studied. METHODS: We investigated a woman with a large, heterogeneous adrenal mass and severe adrenocorticotropic hormone-independent glucocorticoid and androgen excess, a biochemical presentation typically suggestive of adrenocortical carcinoma. The patient presented during pregnancy (22nd week of gestation) and reported an 18-month history of oligomenorrhea, hirsutism, and weight gain. We undertook an exploratory study with detailed histopathological and genetic analysis of the resected adrenal mass and leukocyte DNA collected from the patient and her parents. RESULTS: Histopathology revealed benign macronodular adrenal hyperplasia. Imaging showed a persistently normal contralateral adrenal gland. Whole-exome sequencing of 4 representative nodules detected KDM1A germline variants, benign NM_001009999.3:c.136G > A:p.G46S, and likely pathogenic NM_001009999.3:exon6:c.865_866del:p.R289Dfs*7. Copy number variation analysis demonstrated an additional somatic loss of the KDM1A wild-type allele on chromosome 1p36.12 in all nodules. RNA sequencing of a representative nodule showed low/absent KDM1A expression and increased GIPR expression compared with 52 unilateral sporadic adenomas and 4 normal adrenal glands. Luteinizing hormone/chorionic gonadotropin receptor expression was normal. Sanger sequencing confirmed heterozygous KDM1A variants in both parents (father: p.R289Dfs*7 and mother: p.G46S) who showed no clinical features suggestive of glucocorticoid or androgen excess. CONCLUSIONS: We investigated the first PUMAH associated with severe Cushing's syndrome and concomitant androgen excess, suggesting pathogenic mechanisms involving KDM1A.


Subject(s)
Cushing Syndrome , Histone Demethylases , Humans , Female , Adult , Histone Demethylases/genetics , Histone Demethylases/metabolism , Cushing Syndrome/genetics , Cushing Syndrome/pathology , Cushing Syndrome/metabolism , Glucocorticoids , Pregnancy , Androgens/metabolism , Adrenal Glands/pathology , Adrenal Glands/metabolism , Adrenal Glands/diagnostic imaging , Adrenal Hyperplasia, Congenital/genetics , Adrenal Hyperplasia, Congenital/complications , Adrenal Hyperplasia, Congenital/pathology , Adrenal Hyperplasia, Congenital/metabolism
14.
Environ Int ; 191: 108971, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39180775

ABSTRACT

There is no safe level of air pollution for human health. Traffic-related particulate matter (PM2.5) is a major in-utero toxin, mechanisms of action of which are not fully understood. BALB/c dams were exposed to an Australian level of traffic PM2.5 (5 µg/mouse/day, intranasal, 6 weeks before mating, during gestation and lactation). Male offspring had reduced memory in adulthood, whereas memory was normal in female littermates, similar to human responses. Maternal PM2.5 exposure resulted in oxidative stress and abnormal mitochondria in male, but not female, brains. RNA-sequencing analysis showed unique sex-related changes in newborn brains. Two X-chromosome-linked histone lysine demethylases, Kdm6a and Kdm5c, demonstrated higher expression in female compared to male littermates, in addition to upregulated genes with known functions to support mitochondrial function, synapse growth and maturation, cognitive function, and neuroprotection. No significant changes in Kdm6a and Kdm5c were found in male littermates, nor other genes, albeit significantly impaired memory function after birth. In primary foetal cortical neurons, PM2.5 exposure suppressed neuron and synaptic numbers and induced oxidative stress, which was prevented by upregulation of Kdm6a or Kdm5c. Therefore, timely epigenetic adaptation by histone demethylation to open DNA for translation before birth may be the key to protecting females against prenatal PM2.5 exposure-induced neurological disorders, which fail to occur in males associated with their poor cognitive outcomes.


Subject(s)
Air Pollution , Maternal Exposure , Memory , Particulate Matter , Animals , Mice , Mice, Inbred BALB C , Particulate Matter/toxicity , Male , Female , Sex Characteristics , Neurons/cytology , Brain/pathology , Mitochondria/pathology , Gene Expression , Animals, Newborn , Histone Demethylases/genetics , Cells, Cultured
15.
Int Immunopharmacol ; 141: 112896, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39146782

ABSTRACT

Colorectal cancer (CRC), as a fatal cancer, is one of the most common cancers worldwide. Although the standard treatment for colorectal cancer is well researched and established, long-term patient survival remains poor, and mortality remains high. Therefore, more and more effective treatment options are needed. To evaluate the efficacy of bevacizumab, the histone demethylase inhibitor IOX1, or their combination for the treatment of colorectal cancer, we examined the effects of IOX1, bevacizumab, and IOX1 combined with bevacizumab on cell activity, proliferation, and migration of colorectal cancer cell lines HCT116, RKO, and CT26 by CCK8, colony formation assay, wound healing assay, and transwell assay. The effects of the drugs alone as well as in combination on apoptosis in colorectal cancer cell lines were examined by flow cytometry and further validated by Western blotting for apoptosis-related proteins. The antitumor effects of treatment alone or in combination on colorectal cancer cells were examined in animal models. Mice were injected subcutaneously with CT26 cells and the growth and immune infiltration in tumor tissues were detected by IHC after drug treatment. We found that IOX1 could effectively inhibit the activity of CRC cells and had a significant inhibitory effect on the proliferation and migration of CRC cells. The apoptosis rate increased in a dose-dependent manner after IOX1 treatment on colorectal cancer cells, and the expression of apoptosis-related proteins changed accordingly. Further combination with bevacizumab revealed that the combination had a more significant effect on the proliferation, migration, and apoptosis of CRC cells than either IOX1 or bevacizumab alone. In vivo experiments have found that both alone and combination drugs can inhibit the growth of mouse tumors, but the effect of combination inhibition is the most obvious. Combination therapy significantly inhibited the expression of proliferative marker (Ki67) in tumor xenograft models, and increased content of antigen-specific CD4+, CD8+T cell growth, and granzymeB (GZMB), which is associated with T cell cytotoxicity, was detected in combination therapy. Immunoassays suppressed the expression of relevant PD-1 and decreased. The anticancer drug bevacizumab and the histone demethylase inhibitor IOX1 may inhibit colon cancer cell growth by regulating apoptosis. The inhibitory effect of combination therapy on tumor growth may be achieved, in part, through upregulation of infiltration-mediated tumor immunity by T lymphocytes. The combination of IOX1 and bevacizumab produced significant synergistic effects. This study aims to provide a new direction for CRC combination therapy.


Subject(s)
Apoptosis , Bevacizumab , Cell Proliferation , Colorectal Neoplasms , Animals , Bevacizumab/therapeutic use , Bevacizumab/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Humans , Apoptosis/drug effects , Mice , Cell Proliferation/drug effects , Cell Line, Tumor , Xenograft Model Antitumor Assays , Cell Movement/drug effects , Mice, Inbred BALB C , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Mice, Nude , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism
16.
Int J Biol Macromol ; 278(Pt 4): 134798, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153678

ABSTRACT

Histone lysine demethylase (KDM), AlkB homolog (ALKBH), and Ten-Eleven Translocation (TET) proteins are members of the 2-Oxoglutarate (2OG) and ferrous iron-dependent oxygenases, each of which harbors a catalytic domain centered on a double-stranded ß-helix whose topology restricts the regions directly involved in substrate binding. However, they have different catalytic functions, and the deeply structural biological reasons are not yet clear. In this review, the catalytic domain features of the three protein families are summarized from both sequence and structural perspectives. The construction of the phylogenetic tree and comparison of the structure show ten relatively conserved ß-sheets and three key regions with substantial structural differences. We summarize the relationship between three key regions of remarkable differences and the substrate compatibility of the three protein families. This review facilitates research into substrate-selective inhibition and bioengineering by providing new insights into the catalytic domains of KDM, ALKBH, and TET proteins.


Subject(s)
Catalytic Domain , Ketoglutaric Acids , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/chemistry , Humans , Models, Molecular , Phylogeny , Substrate Specificity , Iron/chemistry , Iron/metabolism , Animals , Histone Demethylases/chemistry , Histone Demethylases/metabolism , Amino Acid Sequence
17.
EMBO J ; 43(19): 4197-4227, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39160277

ABSTRACT

In mammals, the transition from mitosis to meiosis facilitates the successful production of gametes. However, the regulatory mechanisms that control meiotic initiation remain unclear, particularly in the context of complex histone modifications. Herein, we show that KDM2A, acting as a lysine demethylase targeting H3K36me3 in male germ cells, plays an essential role in modulating meiotic entry and progression. Conditional deletion of Kdm2a in mouse pre-meiotic germ cells results in complete male sterility, with spermatogenesis ultimately arrested at the zygotene stage of meiosis. KDM2A deficiency disrupts H3K36me2/3 deposition in c-KIT+ germ cells, characterized by a reduction in H3K36me2 but a dramatic increase in H3K36me3. Furthermore, KDM2A recruits the transcription factor E2F1 and its co-factor HCFC1 to the promoters of key genes required for meiosis entry and progression, such as Stra8, Meiosin, Spo11, and Sycp1. Collectively, our study unveils an essential role for KDM2A in mediating H3K36me2/3 deposition and controlling the programmed gene expression necessary for the transition from mitosis to meiosis during spermatogenesis.


Subject(s)
E2F1 Transcription Factor , Jumonji Domain-Containing Histone Demethylases , Meiosis , Spermatogenesis , Animals , Male , Mice , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Spermatogenesis/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Host Cell Factor C1/metabolism , Host Cell Factor C1/genetics , Histones/metabolism , Histones/genetics , Mice, Knockout , Infertility, Male/genetics , Infertility, Male/metabolism , Histone Demethylases
18.
Nucleic Acids Res ; 52(17): 10297-10310, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39087553

ABSTRACT

Transient halting of transcription activity on the damaged chromatin facilitates DNA double-strand break (DSB) repair. However, the molecular mechanisms that facilitate transcription recovery following DSB repair remain largely undefined. Notably, failure to restore gene expression in a timely manner can compromise transcriptome signatures and may impose deleterious impacts on cell identity and cell fate. Here, we report PHF8 as the major demethylase that reverses transcriptionally repressive epigenetic modification laid down by the DYRK1B-EHMT2 pathway. We found that PHF8 concentrates at laser-induced DNA damage tracks in a DYRK1B-dependent manner and promotes timely resolution of local H3K9me2 to facilitate the resumption of transcription. Moreover, PHF8 also assists in the recovery of ribosomal DNA (rDNA) transcription following the repair of nucleolar DSBs. Taken together, our findings uncover PHF8 as a key mediator that coordinates transcription activities during the recovery phase of DSB responses.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Transcription Factors , Transcription, Genetic , Transcription Factors/metabolism , Transcription Factors/genetics , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Histones/metabolism , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Chromatin/metabolism , Chromatin/genetics , Histone Demethylases/metabolism , Histone Demethylases/genetics , Epigenesis, Genetic
19.
Cancer Res Commun ; 4(9): 2320-2334, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39113632

ABSTRACT

Radiotherapy (RT) for prostate cancer has been associated with an increased risk for the development of bladder cancer. We aimed to integrate clinical and genomic data to better understand the development of RT-associated bladder cancer. A retrospective analysis was performed to identify control patients (CTRL; n = 41) and patients with RT-associated bladder cancer (n = 41). RT- and CTRL-specific features were then identified through integration and analysis of the genomic sequencing data and clinical variables. RT-associated bladder tumors were significantly enriched for alterations in KDM6A and ATM, whereas CTRL tumors were enriched for CDKN2A mutation. Globally, there were an increased number of variants within RT tumors, albeit at a lower variant allele frequency. Mutational signature analysis revealed three predominate motif patterns, with similarity to SBS2/13 (APOBEC3A), SBS5 (ERCC2/smoking), and SBS6/15 (MMR). Poor prognostic factors in the RT cohort include a short tumor latency, smoking status, the presence of the smoking and X-ray therapy mutational signatures, and CDKN2A copy number loss. Based on the clinical and genomic findings, we suggest at least two potential pathways leading to RT-associated bladder cancer: The first occurs in the setting of field cancerization related to smoking or preexisting genetic alterations and leads to the development of more aggressive bladder tumors, and the second involves RT initiating the oncogenic process in otherwise healthy urothelium, leading to a longer latency and less aggressive disease. SIGNIFICANCE: Clinicogenomic analysis of radiation-associated bladder cancer uncovered mutational signatures that, in addition to a short tumor latency, smoking, and CDKN2A loss, are associated with a poor outcome. These clinical and genomic features provide a potential method to identify patients with prostate cancer who are at an increased risk for the development of aggressive bladder cancer following prostate RT.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16 , Mutation , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/radiotherapy , Urinary Bladder Neoplasms/etiology , Male , Prognosis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Aged , Retrospective Studies , Middle Aged , Female , Histone Demethylases/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/epidemiology , Aged, 80 and over
20.
Crit Rev Eukaryot Gene Expr ; 34(8): 25-34, 2024.
Article in English | MEDLINE | ID: mdl-39180205

ABSTRACT

Lysine demethylase 6A (KDM6A) is abnormally expressed in various cancer. This study aimed to investigate the potential of KDM6A in pancreatic cancer (PC). mRNA expression was calculated by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR). Protein expression was detected by Western blot. Cell viability was measured by Cell Counting Kit (CCK-8) assay. Cell angiogenesis was determined by tube formation assay. Cell migration and invasion were determined by Transwell assay. We found that KDM6A was upregulated in PC patients and cells. Interestingly, KDM6A deficiency inhibited the proliferation and angiogenesis of PC cells. Moreover, KDM6A knockdown suppressed the migration and invasion of PC cells. Additionally, KDM6A upregulated the expression of lysosomal associated membrane protein 3 (LAMP3) via driving demethylation of H3K27me3. Overexpression of LAMP3 reversed the effects of KDM6A knockdown and contributed to the angiogenesis and aggressiveness of PC cells. In summary, KDM6A-mediated demethylation of tri-methylation at lysine 27 of histone H3 (H3K27me3) promotes the transcription of LAMP3, resulting the angiogenesis and aggressiveness of PC. Therefore, targeting KDM6A may be an anti-angiogenetic strategy for PC.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Histone Demethylases , Lysosomal Membrane Proteins , Neoplasm Invasiveness , Neovascularization, Pathologic , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Cell Movement/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Histone Demethylases/metabolism , Histone Demethylases/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/genetics , Angiogenesis , Neoplasm Proteins , Lysosomal-Associated Membrane Protein 3
SELECTION OF CITATIONS
SEARCH DETAIL