Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Blood Adv ; 8(9): 2217-2234, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38457926

ABSTRACT

ABSTRACT: Multiple myeloma (MM) cells are addicted to MYC and its direct transactivation targets IRF4 for proliferation and survival. MYC and IRF4 are still considered "undruggable," as most small-molecule inhibitors suffer from low potency, suboptimal pharmacokinetic properties, and undesirable off-target effects. Indirect inhibition of MYC/IRF4 emerges as a therapeutic vulnerability in MM. Here, we uncovered an unappreciated tumor-suppressive role of C-terminal binding protein 2 (CTBP2) in MM via strong inhibition of the MYC-IRF4 axis. In contrast to epithelial cancers, CTBP2 is frequently downregulated in MM, in association with shortened survival, hyperproliferative features, and adverse clinical outcomes. Restoration of CTBP2 exhibited potent antitumor effects against MM in vitro and in vivo, with marked repression of the MYC-IRF4 network genes. Mechanistically, CTBP2 impeded the transcription of MYC and IRF4 by histone H3 lysine 27 deacetylation (H3K27ac) and indirectly via activation of the MYC repressor IFIT3. In addition, activation of the interferon gene signature by CTBP2 suggested its concomitant immunomodulatory role in MM. Epigenetic studies have revealed the contribution of polycomb-mediated silencing and DNA methylation to CTBP2 inactivation in MM. Notably, inhibitors of Enhance of zeste homolog 2, histone deacetylase, and DNA methyltransferase, currently under evaluation in clinical trials, were effective in restoring CTBP2 expression in MM. Our findings indicated that the loss of CTBP2 plays an essential role in myelomagenesis and deciphers an additional mechanistic link to MYC-IRF4 dysregulation in MM. We envision that the identification of novel critical regulators will facilitate the development of selective and effective approaches for treating this MYC/IRF4-addicted malignancy.


Subject(s)
Alcohol Oxidoreductases , Co-Repressor Proteins , Interferon Regulatory Factors , Multiple Myeloma , Proto-Oncogene Proteins c-myc , Animals , Humans , Mice , Alcohol Oxidoreductases/metabolism , Alcohol Oxidoreductases/antagonists & inhibitors , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects , Tumor Suppressor Proteins/metabolism , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/metabolism
2.
Cancer Res ; 82(20): 3830-3844, 2022 10 17.
Article in English | MEDLINE | ID: mdl-35950923

ABSTRACT

Most patients with estrogen receptor alpha-positive (ER+) breast cancers initially respond to treatment but eventually develop therapy resistance with disease progression. Overexpression of oncogenic ER coregulators, including proline, glutamic acid, and leucine-rich protein 1 (PELP1), are implicated in breast cancer progression. The lack of small molecules that inhibits PELP1 represents a major knowledge gap. Here, using a yeast-two-hybrid screen, we identified novel peptide inhibitors of PELP1 (PIP). Biochemical assays demonstrated that one of these peptides, PIP1, directly interacted with PELP1 to block PELP1 oncogenic functions. Computational modeling of PIP1 revealed key residues contributing to its activity and facilitated the development of a small-molecule inhibitor of PELP1, SMIP34, and further analyses confirmed that SMIP34 directly bound to PELP1. In breast cancer cells, SMIP34 reduced cell growth in a dose-dependent manner. SMIP34 inhibited proliferation of not only wild-type (WT) but also mutant (MT) ER+ and therapy-resistant breast cancer cells, in part by inducing PELP1 degradation via the proteasome pathway. RNA sequencing analyses showed that SMIP34 treatment altered the expression of genes associated with estrogen response, cell cycle, and apoptosis pathways. In cell line-derived and patient-derived xenografts of both WT and MT ER+ breast cancer models, SMIP34 reduced proliferation and significantly suppressed tumor progression. Collectively, these results demonstrate SMIP34 as a first-in-class inhibitor of oncogenic PELP1 signaling in advanced breast cancer. SIGNIFICANCE: Development of a novel inhibitor of oncogenic PELP1 provides potential therapeutic avenues for treating therapy-resistant, advanced ER+ breast cancer.


Subject(s)
Breast Neoplasms , Co-Repressor Proteins , Transcription Factors , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/metabolism , Estrogen Receptor alpha/genetics , Estrogens , Female , Glutamic Acid , Humans , Leucine , Proline , Proteasome Endopeptidase Complex , Receptors, Estrogen/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
3.
Biochim Biophys Acta Mol Cell Res ; 1869(4): 119200, 2022 04.
Article in English | MEDLINE | ID: mdl-34968577

ABSTRACT

Tubulointerstitial fibrosis is a common pathway of chronic kidney disease (CKD) and is closely related to the progression of CKD. LMCD1, acting as an intermediary, has been reported to play a role in cardiac fibrosis. However, its role in renal fibrosis is yet to be deciphered. Based on the GEO database, we found the expression of LMCD1 is increased in kidney tissues of CKD patients and in human proximal tubular epithelial (HK-2) cells treated with transforming growth factor-ß1 (TGF-ß1), suggesting that LMCD1 may be involved in tubulointerstitial fibrosis. Herein, we investigated the role of LMCD1 in mice with unilateral ureteral obstruction (UUO) and in TGF-ß1-stimulated HK-2 cells. In the UUO model, the expression of LMCD1 was upregulated. UUO-induced renal histopathological changes were mitigated by knockdown of LMCD1. LMCD1 silence alleviated renal interstitial fibrosis in UUO mice by decreasing the expression of TGF-ß1, fibronectin, collagen I, and collagen III. LMCD1 deficiency suppressed cell apoptosis in kidney to prevent UUO-triggered renal injury. Furthermore, LMCD1 deficiency blocked the activation of ERK signaling in UUO mice. In vitro, LMCD1 was upregulated in HK-2 cells after TGF-ß1 stimulation. LMCD1 silence abrogated TGF-ß1-mediated upregulation of fibrotic genes. Treatment of HK-2 cells with ERK-specific inhibitor SCH772984 and agonist TPA validated LMCD1 exerted its function via activating ERK signaling. Together, our findings suggest that inhibition of LMCD1 protects against renal interstitial fibrosis by impeding ERK activation.


Subject(s)
Co-Repressor Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , LIM Domain Proteins/metabolism , Nephritis, Interstitial/pathology , Animals , Apoptosis , Cell Line , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Indazoles/pharmacology , Kidney/metabolism , Kidney/pathology , LIM Domain Proteins/antagonists & inhibitors , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Nephritis, Interstitial/etiology , Nephritis, Interstitial/metabolism , Piperazines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , Ureteral Obstruction/complications
4.
Theranostics ; 11(14): 6983-7004, 2021.
Article in English | MEDLINE | ID: mdl-34093866

ABSTRACT

Rationale: We recently demonstrated that the 'Metabesity' factor HMG20A regulates islet beta-cell functional maturity and adaptation to physiological stress such as pregnancy and pre-diabetes. HMG20A also dictates central nervous system (CNS) development via inhibition of the LSD1-CoREST complex but its expression pattern and function in adult brain remains unknown. Herein we sought to determine whether HMG20A is expressed in the adult CNS, specifically in hypothalamic astrocytes that are key in glucose homeostasis and whether similar to islets, HMG20A potentiates astrocyte function in response to environmental cues. Methods: HMG20A expression profile was assessed by quantitative PCR (QT-PCR), Western blotting and/or immunofluorescence in: 1) the hypothalamus of mice exposed or not to either a high-fat diet or a high-fat high-sucrose regimen, 2) human blood leukocytes and adipose tissue obtained from healthy or diabetic individuals and 3) primary mouse hypothalamic astrocytes exposed to either high glucose or palmitate. RNA-seq and cell metabolic parameters were performed on astrocytes treated or not with a siHMG20A. Astrocyte-mediated neuronal survival was evaluated using conditioned media from siHMG20A-treated astrocytes. The impact of ORY1001, an inhibitor of the LSD1-CoREST complex, on HMG20A expression, reactive astrogliosis and glucose metabolism was evaluated in vitro and in vivo in high-fat high-sucrose fed mice. Results: We show that Hmg20a is predominantly expressed in hypothalamic astrocytes, the main nutrient-sensing cell type of the brain. HMG20A expression was upregulated in diet-induced obesity and glucose intolerant mice, correlating with increased transcript levels of Gfap and Il1b indicative of inflammation and reactive astrogliosis. Hmg20a transcript levels were also increased in adipose tissue of obese non-diabetic individuals as compared to obese diabetic patients. HMG20A silencing in astrocytes resulted in repression of inflammatory, cholesterol biogenesis and epithelial-to-mesenchymal transition pathways which are hallmarks of reactive astrogliosis. Accordingly, HMG20A depleted astrocytes exhibited reduced mitochondrial bioenergetics and increased susceptibility to apoptosis. Neuron viability was also hindered in HMG20A-depleted astrocyte-derived conditioned media. ORY1001 treatment rescued expression of reactive astrogliosis-linked genes in HMG20A ablated astrocytes while enhancing cell surface area, GFAP intensity and STAT3 expression in healthy astrocytes, mimicking the effect of HMG20A. Furthermore, ORY1001 treatment protected against obesity-associated glucose intolerance in mice correlating with a regression of hypothalamic HMG20A expression, indicative of reactive astrogliosis attenuation with improved health status. Conclusion: HMG20A coordinates the astrocyte polarization state. Under physiological pressure such as obesity and insulin resistance that induces low grade inflammation, HMG20A expression is increased to induce reactive astrogliosis in an attempt to preserve the neuronal network and re-establish glucose homeostasis. Nonetheless, a chronic metabesity state or functional mutations will result in lower levels of HMG20A, failure to promote reactive astrogliosis and increase susceptibility of neurons to stress-induced apoptosis. Such effects could be reversed by ORY1001 treatment both in vitro and in vivo, paving the way for a new therapeutic approach for Type 2 Diabetes Mellitus.


Subject(s)
Astrocytes/metabolism , Diabetes Mellitus, Type 2/metabolism , Gliosis/metabolism , High Mobility Group Proteins/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Obesity/metabolism , Adipose Tissue/metabolism , Adult , Animals , Cell Survival/drug effects , Co-Repressor Proteins/antagonists & inhibitors , Diet, High-Fat , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/genetics , Histone Demethylases/antagonists & inhibitors , Humans , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria/genetics , Mitochondria/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , RNA, Small Interfering , RNA-Seq
5.
Sci Rep ; 11(1): 9018, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33907245

ABSTRACT

Histone/protein deacetylases (HDAC) 1 and 2 are typically viewed as structurally and functionally similar enzymes present within various co-regulatory complexes. We tested differential effects of these isoforms in renal ischemia reperfusion injury (IRI) using inducible knockout mice and found no significant change in ischemic tolerance with HDAC1 deletion, but mitigation of ischemic injury with HDAC2 deletion. Restriction of HDAC2 deletion to the kidney via transplantation or PAX8-controlled proximal renal tubule-specific Cre resulted in renal IRI protection. Pharmacologic inhibition of HDAC2 increased histone acetylation in the kidney but did not extend renal protection. Protein analysis demonstrated increased HDAC1-associated CoREST protein in HDAC2-/- versus WT cells, suggesting that in the absence of HDAC2, increased CoREST complex occupancy of HDAC1 can stabilize this complex. In vivo administration of a CoREST inhibitor exacerbated renal injury in WT mice and eliminated the benefit of HDAC2 deletion. Gene expression analysis of endothelin showed decreased endothelin levels in HDAC2 deletion. These data demonstrate that contrasting effects of HDAC1 and 2 on CoREST complex stability within renal tubules can affect outcomes of renal IRI and implicate endothelin as a potential downstream mediator.


Subject(s)
Co-Repressor Proteins/metabolism , Histone Deacetylase 2/metabolism , Kidney Tubules, Proximal/metabolism , Reperfusion Injury/prevention & control , Animals , Co-Repressor Proteins/antagonists & inhibitors , Endothelins/metabolism , Enzyme Inhibitors/pharmacology , Female , Gene Deletion , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Kidney Tubules, Proximal/drug effects , Male , Mice , Mice, Knockout
6.
Drug Des Devel Ther ; 14: 1583-1592, 2020.
Article in English | MEDLINE | ID: mdl-32425504

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is a common cause of cancer-related deaths. This study identified the regulatory pattern of gallic acid in NSCLC. METHODS: Human NSCLC cells were treated with different doses of gallic acid, after which, MTT assay and flow cytometry were performed to determine the survival and apoptotic rate of human NSCLC cells. Then, co-immunoprecipitation assay was performed to analyze the relationships between gallic acid, epidermal growth factor receptor (EGFR), and CARM1-PELP1. Next, we analyzed whether PELP1, CARM1 and EGFR were associated with the effects of gallic acid on NSCLC cells by conducting rescue experiments. The expression pattern of phosphorylated EGFR, EGFR, Ki67, as well as Fas, FasL and Caspase 3 proteins in cancer cells or xenografts was measured by Western blot analysis. Lastly, the role of gallic acid in the tumor growth was assessed in nude mice. RESULTS: The ideal dose of gallic acid that presented good suppressive effect on NSCLC cells were 30 µM, 50 µM and 75 µM, respectively. Gallic acid played an inhibiting role in the activation of EGFR, which further reduced the formation of CARM1-PELP1 complex, ultimately repressed the proliferation and elevated apoptosis of NSCLC cells. Meanwhile, CARM1 repression led to decreased growth, proliferation and migration abilities of NSCLC cells. Animal experiments confirmed that gallic acid contributed to the inhibition of tumor growth in vivo. CONCLUSION: To sum up, gallic acid could potentially prevent NSCLC progression via inhibition of EGFR activation and impairment of the binding of CARM1 to PELP1, highlighting a novel therapy to dampen NSCLC progression.


Subject(s)
Antineoplastic Agents/pharmacology , CARD Signaling Adaptor Proteins/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Co-Repressor Proteins/antagonists & inhibitors , Disease Progression , Gallic Acid/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , CARD Signaling Adaptor Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Co-Repressor Proteins/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Guanylate Cyclase/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Structure-Activity Relationship , Transcription Factors/metabolism
7.
Int J Biol Sci ; 16(7): 1107-1120, 2020.
Article in English | MEDLINE | ID: mdl-32174788

ABSTRACT

Traumatic brain injury (TBI) induces an acute inflammatory response in the central nervous system that involves both resident and peripheral immune cells. The ensuing chronic neuroinflammation causes cell death and tissue damage and may contribute to neurodegeneration. The molecular mechanisms involved in the maintenance of this chronic inflammation state remain underexplored. C-terminal binding protein (CtBP) 1 and 2 are transcriptional coregulators that repress diverse cellular processes. Unexpectedly, we find that the CtBPs can transactivate a common set of proinflammatory genes both in lipopolysaccharide-activated microglia, astrocytes and macrophages, and in a mouse model of the mild form of TBI. We also find that the expression of these genes is markedly enhanced by a single mild injury in both brain and peripheral blood leukocytes in a severity- and time-dependent manner. Moreover, we were able to demonstrate that specific inhibitors of the CtBPs effectively suppress the expression of the CtBP target genes and thus improve neurological outcome in mice receiving single and repeated mild TBIs. This discovery suggests new avenues for therapeutic modulation of the inflammatory response to brain injury.


Subject(s)
Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Microglia/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Astrocytes/drug effects , Astrocytes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects
8.
Trends Cancer ; 6(3): 247-260, 2020 03.
Article in English | MEDLINE | ID: mdl-32101727

ABSTRACT

Alternative lengthening of telomeres (ALT) is a mechanism of telomere maintenance that is observed in many of the most recalcitrant cancer subtypes. Telomeres in ALT cancer cells exhibit a distinctive nucleoprotein architecture shaped by the mismanagement of chromatin that fosters cycles of DNA damage and replicative stress that activate homology-directed repair (HDR). Mutations in specific chromatin-remodeling factors appear to be key determinants of the emergence and survival of ALT cancer cells. However, these may represent vulnerabilities for the targeted elimination of ALT cancer cells that infiltrate tissues and organs to become devastating tumors. In this review we examine recent findings that provide new insights into the factors and mechanisms that mediate telomere length maintenance and survival of ALT cancer cells.


Subject(s)
Neoplasms/genetics , Telomere Homeostasis , Chromatin/ultrastructure , Clonal Evolution , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/physiology , DNA Damage , DNA Repair , DNA Replication , DNA, Neoplasm/metabolism , DNA, Neoplasm/ultrastructure , Histones/physiology , Homologous Recombination , Humans , Models, Genetic , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/physiology , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms/ultrastructure , Nucleic Acid Conformation , Telomerase/genetics , Telomerase/physiology , X-linked Nuclear Protein/antagonists & inhibitors , X-linked Nuclear Protein/physiology
9.
Cardiovasc Res ; 116(3): 633-644, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31147690

ABSTRACT

AIMS: Regulated cell death is a main contributor of myocardial ischaemia-reperfusion (IR) injury during acute myocardial infarction. In this context, targeting apoptosis could be a potent therapeutical strategy. In a previous study, we showed that DAXX (death-associated protein) was essential for transducing the FAS-dependent apoptotic signal during IR injury. The present study aims at evaluating the cardioprotective effects of a synthetic peptide inhibiting FAS:DAXX interaction. METHODS AND RESULTS: An interfering peptide was engineered and then coupled to the Tat cell penetrating peptide (Tat-DAXXp). Its internalization and anti-apoptotic properties were demonstrated in primary cardiomyocytes. Importantly, an intravenous bolus injection of Tat-DAXXp (1 mg/kg) 5 min before reperfusion in a murine myocardial IR model decreased infarct size by 48% after 24 h of reperfusion. In addition, Tat-DAXXp was still efficient after a 30-min delayed administration, and was completely degraded and eliminated within 24 h thereby reducing risks of potential side effects. Importantly, Tat-DAXXp reduced mouse early post-infarction mortality by 67%. Mechanistically, cardioprotection was supported by both anti-apoptotic and pro-survival effects, and an improvement of myocardial functional recovery as evidenced in ex vivo experiments. CONCLUSIONS: Our study demonstrates that a single dose of Tat-DAXXp injected intravenously at the onset of reperfusion leads to a strong cardioprotection in vivo by inhibiting IR injury validating Tat-DAXXp as a promising candidate for therapeutic application.


Subject(s)
Apoptosis/drug effects , Cell-Penetrating Peptides/pharmacology , Co-Repressor Proteins/antagonists & inhibitors , Molecular Chaperones/antagonists & inhibitors , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Animals , Cell Line , Cell Survival/drug effects , Co-Repressor Proteins/metabolism , Disease Models, Animal , Male , Mice, Inbred C57BL , Molecular Chaperones/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Recovery of Function/drug effects , Signal Transduction , fas Receptor/metabolism
10.
Cells ; 8(10)2019 09 27.
Article in English | MEDLINE | ID: mdl-31569653

ABSTRACT

Spermatogenesis is a process by which haploid cells differentiate from germ cells in the seminiferous tubules of the testes. TLE3, a transcriptional co-regulator that interacts with DNA-binding factors, plays a role in the development of somatic cells. However, no studies have shown its role during germ cell development in the testes. Here, we examined TLE3 expression in the testes during spermatogenesis. TLE3 was highly expressed in mouse testes and was dynamically regulated in different cell types of the seminiferous tubules, spermatogonia, spermatids, and Sertoli cells, but not in the spermatocytes. Interestingly, TLE3 was not detected in Sertoli cells on postnatal day 7 (P7) but was expressed from P10 onward. The microarray analysis showed that the expression of numerous genes changed upon TLE3 knockdown in a Sertoli cell line TM4. These include 1597 up-regulated genes and 1452 down-regulated genes in TLE3-knockdown TM4 cells. Ingenuity Pathway Analysis (IPA) showed that three factors were up-regulated and two genes were down-regulated upon TLE3 knockdown in TM4 cells. The abnormal expression of the three factors is associated with cellular malfunctions such as abnormal differentiation and Sertoli cell formation. Thus, TLE3 is differentially expressed in Sertoli cells and plays a crucial role in regulating cell-specific genes involved in the differentiation and formation of Sertoli cells during testicular development.


Subject(s)
Cell Differentiation , Co-Repressor Proteins/metabolism , Seminiferous Tubules/metabolism , Sertoli Cells/metabolism , Spermatogenesis , Testis/metabolism , Animals , Cells, Cultured , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Seminiferous Tubules/cytology , Sertoli Cells/cytology , Testis/cytology
11.
Eur J Med Chem ; 182: 111656, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31494467

ABSTRACT

Chemical probes of epigenetic 'readers' of histone post-translational modifications (PTMs) have become powerful tools for mechanistic and functional studies of their target proteins in physiology and pathology. However, only limited 'reader' probes have been developed, which restricted our understanding towards these macromolecules and their roles in cells or animals. Here, we reported a structure-guided approach to develop and characterize benzo [d]oxazol-2(3H)-one analogs as the first potent and selective small-molecule inhibitors of chromodomain Y-like (CDYL), a histone methyllysine reader protein. The binding conformation between the chromodomain of CDYL and the modified peptidomimetics was studied via molecular docking and dynamic simulations, facilitating subsequent virtual screening of tens of hits from Specs chemical library validated by SPR technique (KD values: from 271.1 µM to 5.4 µM). Further design and synthesis of 43 compounds helped to interpret the structure-activity relationship (SAR) that lead to the discovery of novel small-molecule inhibitors of CDYL. Compound D03 (KD: 0.5 µM) was discovered and showed excellent selectivity among other chromodomain proteins, including CDYL2 (>140 folds), CDY1 (no observed binding) and CBX7 (>32 folds). Moreover, we demonstrated that D03 engaged with endogenous CDYL in a dose-dependent manner, and perturbed the recruitment of CDYL onto chromatin, resulting in transcriptional derepression of its target genes. Finally, the results showed that D03 promoted the development and branching of neurodendrites by inhibiting CDYL in hippocampal and cortical cultured neurons. This study not only discovers the first selective small-molecule inhibitors of CDYL, but provids a new chemical tool to intervene the dynamic nature of bio-macromolecules involved in epigenetic mechanism.


Subject(s)
Benzoxazoles/pharmacology , Co-Repressor Proteins/antagonists & inhibitors , Hydro-Lyases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Benzoxazoles/chemical synthesis , Benzoxazoles/chemistry , Cell Survival/drug effects , Cells, Cultured , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Mice , Mice, Inbred C57BL , Molecular Structure , Neurons/drug effects , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
12.
J Biochem ; 164(6): 415-426, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30165670

ABSTRACT

A small nuclear protein, C1D, has roles in various cellular processes, transcription regulation, genome stability surveillance, DNA repair and RNA processing, all of which are required to maintain the host life cycles. In the previous report, C1D directly interacts with XPB, a component of the nucleotide excision repair complex, and C1D knockdown reduced cell survival of 27-1 cells, CHO derivative cells, after UV irradiation. To find out the role of C1D in UV-damaged cells, we used human cell lines with siRNA or shRNA to knockdown C1D. C1D knockdown reduced cell survival rates of LU99 and 786-O after UV irradiation, although C1D knockdown did not affect the efficiency of the nucleotide excision repair. Immunostaining data support that C1D is not directly involved in the DNA repair process in UV-damaged cells. However, H2O2 treatment reduced cell viability in LU99 and 786-O cells. We also found that C1D knockdown upregulated DDIT3 expression in LU99 cells and downregulated APEX1 in 786-O cells, suggesting that C1D functions as a co-repressor/activator. The data accounts for the reduction of cell survival rates upon UV irradiation.


Subject(s)
Co-Repressor Proteins/metabolism , DNA Repair/radiation effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA/metabolism , Gene Expression Regulation, Enzymologic/radiation effects , Oxidative Stress/radiation effects , Transcription Factor CHOP/metabolism , Animals , Biomarkers/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , DNA Damage , DNA Repair/drug effects , DNA, Neoplasm/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Hydrogen Peroxide/toxicity , Oxidants/toxicity , Oxidative Stress/drug effects , Pyrimidine Dimers/metabolism , RNA Interference , Radiation Injuries, Experimental/enzymology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Transcription Factor CHOP/agonists , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics
13.
Cell Physiol Biochem ; 45(3): 973-983, 2018.
Article in English | MEDLINE | ID: mdl-29428935

ABSTRACT

BACKGROUND/AIMS: Retinal Müller cells could be induced to differentiate into retinal ganglion cells (RGCs), but RGCs derived from Müller cells have defects in axon growth, leading to a defect in signal conduction. In this study we aimed to explore the role of miR-124 in axon growth of RGCs derived from Müller cells. METHODS: Müller cells were isolated from rat retina and induced to dedifferentiate into retinal stem cells. The stem cells were infected by PGC-FU-Atoh7-GFP lentivirus and then transfected with miR-124 or anti-miR-124, and the length of axon was compared. Furthermore, the cells were injected into the eyes of rat chronic ocular hypertension glaucoma model and axon growth in vivo was examined. The targeting of CoREST by miR-124 was detected by luciferase assay. RESULTS: In retinal stem cells, the length of axon was 1,792±64.54 µm in miR-124 group, 509±21.35 µm in control group, and only 87.9±9.24 µm in anti-miR-124 group. In rat model, miR-124 promoted axon growth of RGCs differentiated from retinal stem cells. Furthermore, we found that miR-124 negatively regulated CoREST via directly targeting the binding site in CoREST 3' UTR. CONCLUSIONS: We provide the first evidence that miR-124 regulates axon growth of RGCs derived from Müller cells, and miR-124 has translational potential for gene therapy of glaucoma.


Subject(s)
Axons/metabolism , Ependymoglial Cells/cytology , MicroRNAs/metabolism , Retinal Ganglion Cells/metabolism , Animals , Antagomirs/metabolism , Base Sequence , Binding Sites , Cell Dedifferentiation , Chronic Disease , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Disease Models, Animal , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glaucoma/pathology , Glaucoma/therapy , Ki-67 Antigen/metabolism , Lentivirus/genetics , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Rats , Retinal Ganglion Cells/cytology , Sequence Alignment , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism
14.
Nat Commun ; 9(1): 53, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29302039

ABSTRACT

Here we report corin, a synthetic hybrid agent derived from the class I HDAC inhibitor (entinostat) and an LSD1 inhibitor (tranylcypromine analog). Enzymologic analysis reveals that corin potently targets the CoREST complex and shows more sustained inhibition of CoREST complex HDAC activity compared with entinostat. Cell-based experiments demonstrate that corin exhibits a superior anti-proliferative profile against several melanoma lines and cutaneous squamous cell carcinoma lines compared to its parent monofunctional inhibitors but is less toxic to melanocytes and keratinocytes. CoREST knockdown, gene expression, and ChIP studies suggest that corin's favorable pharmacologic effects may rely on an intact CoREST complex. Corin was also effective in slowing tumor growth in a melanoma mouse xenograft model. These studies highlight the promise of a new class of two-pronged hybrid agents that may show preferential targeting of particular epigenetic regulatory complexes and offer unique therapeutic opportunities.


Subject(s)
Benzamides/pharmacology , Co-Repressor Proteins/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Melanoma/drug therapy , Nerve Tissue Proteins/antagonists & inhibitors , Pyridines/pharmacology , Tranylcypromine/pharmacology , Aged , Animals , Antineoplastic Agents , Cell Line, Tumor , Cell Proliferation , Co-Repressor Proteins/metabolism , Drug Design , Drug Screening Assays, Antitumor , Female , Histone Deacetylases/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Skin Neoplasms/drug therapy , Xenograft Model Antitumor Assays
15.
J Biol Chem ; 292(31): 12885-12894, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28607151

ABSTRACT

Satellite cells are skeletal muscle stem cells that provide myonuclei for postnatal muscle growth, maintenance, and repair/regeneration in adults. Normally, satellite cells are mitotically quiescent, but they are activated in response to muscle injury, in which case they proliferate extensively and exhibit up-regulated expression of the transcription factor MyoD, a master regulator of myogenesis. MyoD forms a heterodimer with E proteins through their basic helix-loop-helix domain, binds to E boxes in the genome and thereby activates transcription at muscle-specific promoters. The central role of MyoD in muscle differentiation has increased interest in finding potential MyoD regulators. Here we identified transducin-like enhancer of split (TLE3), one of the Groucho/TLE family members, as a regulator of MyoD function during myogenesis. TLE3 was expressed in activated and proliferative satellite cells in which increased TLE3 levels suppressed myogenic differentiation, and, conversely, reduced TLE3 levels promoted myogenesis with a concomitant increase in proliferation. We found that, via its glutamine- and serine/proline-rich domains, TLE3 interferes with MyoD function by disrupting the association between the basic helix-loop-helix domain of MyoD and E proteins. Our findings indicate that TLE3 participates in skeletal muscle homeostasis by dampening satellite cell differentiation via repression of MyoD transcriptional activity.


Subject(s)
Co-Repressor Proteins/metabolism , Gene Expression Regulation, Developmental , Muscle Development , Muscle Fibers, Skeletal/metabolism , MyoD Protein/antagonists & inhibitors , Myoblasts/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Cell Proliferation , Cells, Cultured , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/genetics , Gene Deletion , Helix-Loop-Helix Motifs , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/cytology , MyoD Protein/chemistry , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Satellite Cells, Skeletal Muscle/cytology
16.
Cell Biol Int ; 41(2): 204-212, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27935220

ABSTRACT

Nuclear co-repressor (NCoR) regulates peripheral insulin sensitivity; however, its role in modulating insulin sensitivity in skeletal muscle remains elusive. Present study investigated protein expression and effect of NCoR on insulin sensitivity in murine skeletal muscle cell line C2 C12 . Myotubes as compared to myoblasts of C2 C12 cells were found to be more sensitive in response to insulin as increase in insulin-stimulated phosphorylation of AKT at serine 473 residue (pAKTS473 ) was significantly higher in myotubes. Incidentally, reduced protein level of NCoR coincided with differentiation of myoblasts into myotubes of C2 C12 cells. However, insulin stimulation per se failed to affect protein level of NCoR either in myoblasts or myotubes of C2 C12 cells. To assess the role of NCoR on insulin sensitivity, NCoR was transiently knocked down using siRNA in myotubes of C2 C12 . In fact, transient silencing of NCoR led to significant reduction in insulin-stimulated pAKTS473 and impaired glucose uptake. This observation is in contrast to published studies where NCoR has been reported to negatively regulate insulin signaling cascade. Furthermore, transient silencing of NCoR failed to improve insulin sensitivity in chronic hyperinsulinemia-induced insulin-resistant model of C2 C12 cells. Importantly, inhibition of lysosomal protein degradation pathway using ammonium chloride restored protein level of NCoR but failed to increase glucose uptake in serum-starved C2 C12 myotubes. Collectively, data from present study show differential protein level of NCoR under different cell state (myoblast and myotubes) of C2 C12 cells and NCoR proves to be vital for maintaining insulin sensitivity in C2 C12 myotubes.


Subject(s)
Co-Repressor Proteins/metabolism , Insulin/metabolism , Ammonium Chloride , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Line , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Glucose/metabolism , Insulin/pharmacology , Insulin Resistance , Leupeptins/pharmacology , Lysosomes/metabolism , Mice , Models, Biological , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism
17.
FEBS Lett ; 590(1): 43-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26763127

ABSTRACT

Macrophages display heterogeneous phenotypes, including the classical M1 proinflammatory and the alternative M2 anti-inflammatory polarization states. The transducin-like enhancer of split-1 (TLE1) is a transcriptional corepressor whose functions in macrophages have not been studied yet. We report that TLE1 is highly expressed in human alternative macrophages in vitro and in atherosclerotic plaques as well as in adipose tissue M1/M2 mixed macrophages. TLE1 silencing in alternative macrophages decreases the expression of the M2 markers IL-1Ra and IL-10, while it exacerbates TNFα and CCL3 induction by lipopolysaccharide. Hence, TLE1 is expressed in human macrophages where it has potential anti-inflammatory and alternative phenotype promoting properties.


Subject(s)
Co-Repressor Proteins/metabolism , Macrophage Activation , Macrophages/metabolism , Repressor Proteins/metabolism , Animals , Biomarkers/metabolism , Body Mass Index , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Female , Gene Expression Regulation/drug effects , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-10/antagonists & inhibitors , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Interleukin-4/pharmacology , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice, 129 Strain , Obesity/blood , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , RNA Interference , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics
18.
Breast Cancer Res Treat ; 150(3): 487-99, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25788226

ABSTRACT

Triple-negative breast cancer (TNBC), the most aggressive breast cancer subtype, occurs in younger women and is associated with poor prognosis. Gain-of-function mutations in TP53 are a frequent occurrence in TNBC and have been demonstrated to repress apoptosis and up-regulate cell cycle progression. Even though TNBC responds to initial chemotherapy, resistance to chemotherapy develops and is a major clinical problem. Tumor recurrence eventually occurs and most patients die from their disease. An urgent need exists to identify molecular-targeted therapies that can enhance chemotherapy response. In the present study, we report that targeting PELP1, an oncogenic co-regulator molecule, could enhance the chemotherapeutic response of TNBC through the inhibition of cell cycle progression and activation of apoptosis. We demonstrate that PELP1 interacts with MTp53, regulates its recruitment, and alters epigenetic marks at the target gene promoters. PELP1 knockdown reduced MTp53 target gene expression, resulting in decreased cell survival and increased apoptosis upon genotoxic stress. Mechanistic studies revealed that PELP1 depletion contributes to increased stability of E2F1, a transcription factor that regulates both cell cycle and apoptosis in a context-dependent manner. Further, PELP1 regulates E2F1 stability in a KDM1A-dependent manner, and PELP1 phosphorylation at the S1033 residue plays an important role in mediating its oncogenic functions in TNBC cells. Accordingly, depletion of PELP1 increased the expression of E2F1 target genes and reduced TNBC cell survival in response to genotoxic agents. PELP1 phosphorylation was significantly greater in the TNBC tumors than in the other subtypes of breast cancer and in the normal tissues. These findings suggest that PELP1 is an important molecular target in TNBC, and that PELP1-targeted therapies may enhance response to chemotherapies.


Subject(s)
Co-Repressor Proteins/metabolism , E2F1 Transcription Factor/metabolism , Mutation , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Drug Resistance, Neoplasm/drug effects , Female , Gene Knockdown Techniques , Humans , Phosphorylation , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Triple Negative Breast Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism
19.
Exp Cell Res ; 326(2): 326-35, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24803390

ABSTRACT

Transcription factor Foxa1 plays a critical role during neural differentiation and is induced immediately after retinoic acid (RA)-initiated differentiation of pluripotent P19 embryonal carcinoma cells, correlated with the downregulated expression of pluripotency-related genes such as Nanog. To study whether Foxa1 participates in the repression of pluripotency factors, we expressed Foxa1 ectopically in P19 cells and identified that Nanog was repressed directly by Foxa1. We confirmed that Foxa1 was able to interact with Grg3, which is a transcriptional corepressor that expresses in P19 cells as well as during RA-induced P19 cell differentiation. Knockdown of Foxa1 or Grg3 delayed the downregulation of Nanog expression during RA-induced P19 cell differentiation. Furthermore, we found that Foxa1 recruited Grg3 to the Nanog promoter -2kb upstream region and switched the promoter to an inactive chromatin status represented by typical modifications in histone H3. Together, our results suggested a critical involvement of Foxa1 in the negative regulation of Nanog expression during the differentiation of pluripotent stem cells.


Subject(s)
Co-Repressor Proteins/metabolism , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/pathology , Hepatocyte Nuclear Factor 3-alpha/metabolism , Homeodomain Proteins/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Embryonal Carcinoma Stem Cells/drug effects , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , HEK293 Cells , Hepatocyte Nuclear Factor 3-alpha/antagonists & inhibitors , Hepatocyte Nuclear Factor 3-alpha/genetics , Histones/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Mice , Nanog Homeobox Protein , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Promoter Regions, Genetic , Tretinoin/pharmacology
20.
Protein Cell ; 4(8): 620-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23836152

ABSTRACT

The differentiation of periodontal ligament (PDL) progenitor cells is important for maintaining the homeostasis of PDL tissue and alveolar bone. Vitamin C (VC), a water-soluble nutrient that cannot be biosynthesized by humans, is vital for mesenchymal stem cells differentiation and plays an important role in bone remodeling. Therefore, the objective of this study was to determine the function and mechanism of VC in PDL progenitor cells osteogenic differentiation at the molecular level. We demonstrated that VC could induce the osteogenic differentiation and maturation of PDL progenitor cell without other osteogenic agents. During the process, VC preferentially activated ERK1/2 but did not affect JNK or p38. Co-treatment with ERK inhibitor effectively decreased the Vitamin C-induced expression of Runx2. ERK inhibitor also abrogated Vitamin C-induced the minimized nodules formation. PELP1, a nuclear receptor co-regulator, was up-regulated under VC treatment. PELP1 knockdown inhibited ERK phosphorylation. The overexpression of PELP1 had a positive relationship with Runx2 expression. Taken together, we could make a conclude that VC induces the osteogenic differentiation of PDL progenitor cells via PELP1-ERK axis. Our finding implies that VC may have a potential in the regeneration medicine and application to periodontitis treatment.


Subject(s)
Ascorbic Acid/pharmacology , Cell Differentiation/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Periodontal Ligament/cytology , Stem Cells/cytology , Butadienes/pharmacology , Cells, Cultured , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nitriles/pharmacology , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...