Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Cancer Res ; 84(4): 598-615, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38095539

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is the most aggressive pediatric brain tumor, and the oncohistone H3.3K27M mutation is associated with significantly worse clinical outcomes. Despite extensive research efforts, effective approaches for treating DIPG are lacking. Through drug screening, we identified the combination of gemcitabine and fimepinostat as a potent therapeutic intervention for H3.3K27M DIPG. H3.3K27M facilitated gemcitabine-induced apoptosis in DIPG, and gemcitabine stabilized and activated p53, including increasing chromatin accessibility for p53 at apoptosis-related loci. Gemcitabine simultaneously induced a prosurvival program in DIPG through activation of RELB-mediated NF-κB signaling. Specifically, gemcitabine induced the transcription of long terminal repeat elements, activated cGAS-STING signaling, and stimulated noncanonical NF-κB signaling. A drug screen in gemcitabine-treated DIPG cells revealed that fimepinostat, a dual inhibitor of HDAC and PI3K, effectively suppressed the gemcitabine-induced NF-κB signaling in addition to blocking PI3K/AKT activation. Combination therapy comprising gemcitabine and fimepinostat elicited synergistic antitumor effects in vitro and in orthotopic H3.3K27M DIPG xenograft models. Collectively, p53 activation using gemcitabine and suppression of RELB-mediated NF-κB activation and PI3K/AKT signaling using fimepinostat is a potential therapeutic strategy for treating H3.3K27M DIPG. SIGNIFICANCE: Gemcitabine activates p53 and induces apoptosis to elicit antitumor effects in H3.3K27M DIPG, which can be enhanced by blocking NF-κB and PI3K/AKT signaling with fimepinostat, providing a synergistic combination therapy for DIPG.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Morpholines , Pyrimidines , Sulfur Compounds , Child , Humans , Diffuse Intrinsic Pontine Glioma/genetics , Gemcitabine , NF-kappa B , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Proto-Oncogene Proteins c-akt , Phosphatidylinositol 3-Kinases , Tumor Suppressor Protein p53
2.
Cell Regen ; 12(1): 37, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38049682

ABSTRACT

The BMP (Bone morphogenetic protein) signaling pathway plays a central role in metazoan biology, intricately shaping embryonic development, maintaining tissue homeostasis, and influencing disease progression. In the context of cancer, BMP signaling exhibits context-dependent dynamics, spanning from tumor suppression to promotion. Cancer stem cells (CSCs), a modest subset of neoplastic cells with stem-like attributes, exert substantial influence by steering tumor growth, orchestrating therapy resistance, and contributing to relapse. A comprehensive grasp of the intricate interplay between CSCs and their microenvironment is pivotal for effective therapeutic strategies. Among the web of signaling pathways orchestrating cellular dynamics within CSCs, BMP signaling emerges as a vital conductor, overseeing CSC self-renewal, differentiation dynamics, and the intricate symphony within the tumor microenvironment. Moreover, BMP signaling's influence in cancer extends beyond CSCs, intricately regulating cellular migration, invasion, and metastasis. This multifaceted role underscores the imperative of comprehending BMP signaling's contributions to cancer, serving as the foundation for crafting precise therapies to navigate multifaceted challenges posed not only by CSCs but also by various dimensions of cancer progression. This article succinctly encapsulates the diverse roles of the BMP signaling pathway across different cancers, spanning glioblastoma multiforme (GBM), diffuse intrinsic pontine glioma (DIPG), colorectal cancer, acute myeloid leukemia (AML), lung cancer, prostate cancer, and osteosarcoma. It underscores the necessity of unraveling underlying mechanisms and molecular interactions. By delving into the intricate tapestry of BMP signaling's engagement in cancers, researchers pave the way for meticulously tailored therapies, adroitly leveraging its dualistic aspects-whether as a suppressor or promoter-to effectively counter the relentless march of tumor progression.

3.
Cell Rep Med ; 4(11): 101281, 2023 11 21.
Article in English | MEDLINE | ID: mdl-37992683

ABSTRACT

During cancer progression, tumorigenic and immune signals are spread through circulating molecules, such as cell-free DNA (cfDNA) and cell-free RNA (cfRNA) in the blood. So far, they have not been comprehensively investigated in gastrointestinal cancers. Here, we profile 4 categories of cell-free omics data from patients with colorectal cancer and patients with stomach adenocarcinoma and then assay 15 types of genomic, epigenomic, and transcriptomic variations. We find that multi-omics data are more appropriate for detection of cancer genes compared with single-omics data. In particular, cfRNAs are more sensitive and informative than cfDNAs in terms of detection rate, enriched functional pathways, etc. Moreover, we identify several peripheral immune signatures that are suppressed in patients with cancer. Specifically, we establish a γδ-T cell score and a cancer-associated-fibroblast (CAF) score, providing insights into clinical statuses like cancer stage and survival. Overall, we reveal a cell-free multi-molecular landscape that is useful for blood monitoring in personalized cancer treatment.


Subject(s)
Cell-Free Nucleic Acids , Gastrointestinal Neoplasms , Humans , Multiomics , Biomarkers, Tumor/genetics , Cell-Free Nucleic Acids/genetics , Neoplasm Staging , Gastrointestinal Neoplasms/diagnosis , Gastrointestinal Neoplasms/genetics
4.
EMBO J ; 42(3): e112058, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36524443

ABSTRACT

TRIM33 is a chromatin reader required for mammalian mesendoderm differentiation after activation of Nodal signaling, while its role in mESCs is still elusive. Here, we report that TRIM33 co-localizes with promyelocytic leukemia nuclear bodies (PML-NBs) specifically in mESCs, to mediate Nodal signaling-directed transcription of Lefty1/2. We show that TRIM33 puncta formation in mESCs depends on PML and on specific assembly of PML-NBs. Moreover, TRIM33 and PML co-regulate Lefty1/2 expression in mESCs, with both PML protein and formation of mESCs-specific PML-NBs being required for TRIM33 recruitment to these loci, and PML-NBs directly associating with the Lefty1/2 loci. Finally, a TurboID proximity-labeling experiment confirmed that TRIM33 is highly enriched only in mESCs-specific PML-NBs. Thus, our study supports a model in which TRIM33 condensates regulate Nodal signaling-directed transcription in mESCs and shows that PML-NBs can recruit distinct sets of client proteins in a cell-context-dependent manner.


Subject(s)
Mouse Embryonic Stem Cells , Promyelocytic Leukemia Nuclear Bodies , Animals , Humans , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Mouse Embryonic Stem Cells/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction , Cell Nucleus/metabolism , Mammals , Transcription Factors/genetics
5.
Nucleic Acids Res ; 50(18): 10526-10543, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36134711

ABSTRACT

Transforming growth factor ß (TGF-ß) superfamily proteins are potent regulators of cellular development and differentiation. Nodal/Activin/TGF-ß and BMP ligands are both present in the intra- and extracellular milieu during early development, and cross-talk between these two branches of developmental signaling is currently the subject of intense research focus. Here, we show that the Nodal induced lncRNA-Smad7 regulates cell fate determination via repression of BMP signaling in mouse embryonic stem cells (mESCs). Depletion of lncRNA-Smad7 dramatically impairs cardiomyocyte differentiation in mESCs. Moreover, lncRNA-Smad7 represses Bmp2 expression through binding with the Bmp2 promoter region via (CA)12-repeats that forms an R-loop. Importantly, Bmp2 knockdown rescues defects in cardiomyocyte differentiation induced by lncRNA-Smad7 knockdown. Hence, lncRNA-Smad7 antagonizes BMP signaling in mESCs, and similarly regulates cell fate determination between osteocyte and myocyte formation in C2C12 mouse myoblasts. Moreover, lncRNA-Smad7 associates with hnRNPK in mESCs and hnRNPK binds at the Bmp2 promoter, potentially contributing to Bmp2 expression repression. The antagonistic effects between Nodal/TGF-ß and BMP signaling via lncRNA-Smad7 described in this work provides a framework for understanding cell fate determination in early development.


Subject(s)
RNA, Long Noncoding , Smad7 Protein/metabolism , Activins/metabolism , Activins/pharmacology , Animals , Cell Differentiation , Ligands , Mice , RNA, Long Noncoding/metabolism , Smad7 Protein/genetics , Smad7 Protein/pharmacology , Transforming Growth Factor beta/metabolism
6.
Nat Cancer ; 3(9): 1105-1122, 2022 09.
Article in English | MEDLINE | ID: mdl-35915262

ABSTRACT

The most lethal subtype of diffuse intrinsic pontine glioma (DIPG) is H3K27M. Although ACVR1 mutations have been implicated in the pathogenesis of this currently incurable disease, the impacts of bone morphogenetic protein (BMP) signaling on more than 60% of H3K27M DIPG carrying ACVR1 wild-type remain unknown. Here we show that BMP ligands exert potent tumor-suppressive effects against H3.3K27M and ACVR1 WT DIPG in a SMAD-dependent manner. Specifically, clinical data revealed that many DIPG tumors have exploited the capacity of CHRDL1 to hijack BMP ligands. We discovered that activation of BMP signaling promotes the exit of DIPG tumor cells from 'prolonged stem-cell-like' state to differentiation by epigenetically regulating CXXC5, which acts as a tumor suppressor and positive regulator of BMP signaling. Beyond showing how BMP signaling impacts DIPG, our study also identified the potent antitumor efficacy of Dacinostat for DIPG. Thus, our study delineates context-dependent features of the BMP signaling pathway in a DIPG subtype.


Subject(s)
Astrocytoma , Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Astrocytoma/genetics , Bone Morphogenetic Proteins/genetics , Brain Stem Neoplasms/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Diffuse Intrinsic Pontine Glioma/genetics , Epigenesis, Genetic , Humans , Ligands , Signal Transduction/genetics , Transcription Factors/genetics
7.
Nat Commun ; 13(1): 3984, 2022 07 09.
Article in English | MEDLINE | ID: mdl-35810171

ABSTRACT

TGF-ß family proteins including Nodal are known as central regulators of early development in metazoans, yet our understanding of the scope of Nodal signaling's downstream targets and associated physiological mechanisms in specifying developmentally appropriate cell fates is far from complete. Here, we identified a highly conserved, transmembrane micropeptide-NEMEP-as a direct target of Nodal signaling in mesendoderm differentiation of mouse embryonic stem cells (mESCs), and this micropeptide is essential for mesendoderm differentiation. We showed that NEMEP interacts with the glucose transporters GLUT1/GLUT3 and promotes glucose uptake likely through these interactions. Thus, beyond expanding the scope of known Nodal signaling targets in early development and showing that this target micropeptide augments the glucose uptake during mesendoderm differentiation, our study provides a clear example for the direct functional impact of altered glucose metabolism on cell fate determination.


Subject(s)
Mesoderm , Nodal Protein , Animals , Cell Differentiation , Embryonic Stem Cells/metabolism , Glucose/metabolism , Mesoderm/metabolism , Mice , Nodal Protein/metabolism , Transforming Growth Factor beta/metabolism
8.
Cell Rep ; 40(2): 111038, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35830803

ABSTRACT

Despite the fundamental roles of TGF-ß family signaling in cell fate determination in all metazoans, the mechanism by which these signals are spatially and temporally interpreted remains elusive. The cell-context-dependent function of TGF-ß signaling largely relies on transcriptional regulation by SMAD proteins. Here, we discover that the DNA repair-related protein, HMCES, contributes to early development by maintaining nodal/activin- or BMP-signaling-regulated transcriptional network. HMCES binds with R-SMAD proteins, co-localizing at active histone marks. However, HMCES chromatin occupancy is independent on nodal/activin or BMP signaling. Mechanistically, HMCES competitively binds chromatin to limit binding by R-SMAD proteins, thereby forcing their dissociation and resulting in repression of their regulatory effects. In Xenopus laevis embryo, hmces KD causes dramatic development defects with abnormal left-right axis asymmetry along with increasing expression of lefty1. These findings reveal HMCES transcriptional regulatory function in the context of TGF-ß family signaling.


Subject(s)
Activins , Bone Morphogenetic Proteins , Activins/metabolism , Bone Morphogenetic Proteins/metabolism , Chromatin , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
9.
Cell Stem Cell ; 29(6): 948-961.e6, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35659877

ABSTRACT

2-cell-like cells (2CLCs)-which comprise only ∼1% of murine embryonic stem cells (mESCs)-resemble blastomeres of 2-cell-stage embryos and are used to investigate zygotic genome activation (ZGA). Here, we discovered that TRIM66 and DAX1 function together as negative regulators of the 2C-like state in mESCs. Chimeric assays confirmed that mESCs lacking TRIM66 or DAX1 function have bidirectional embryonic and extraembryonic differentiation potential. TRIM66 functions by recruiting the co-repressor DAX1 to the Dux promoter, and TRIM66's repressive effect on Dux is dependent on DAX1. A solved crystal structural shows that TRIM66's PHD finger recognizes H3K4-K9me3, and mutational evidence confirmed that TRIM66's PHD finger is essential for its repression of Dux. Thus, beyond expanding the scope of known 2CLC regulators, our study demonstrates that interventions disrupting TRIM66 or DAX1 function in mESCs yield 2CLCs with expanded bidirectional differentiation potential, opening doors for the practical application of these totipotent-like cells.


Subject(s)
Gene Expression Regulation, Developmental , Zygote , Animals , Embryonic Stem Cells , Genome , Mice , Promoter Regions, Genetic
10.
EMBO Rep ; 21(12): e49684, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33073493

ABSTRACT

mascRNA is a small cytoplasmic RNA derived from the lncRNA MALAT1. After being processed by the tRNA processing enzymes RNase P and RNase Z, mascRNA undergoes CCA addition like tRNAs and folds into a tRNA-like cloverleaf structure. While MALAT1 functions in multiple cellular processes, the role of mascRNA was largely unknown. Here, we show that mascRNA binds directly to the multi-tRNA synthetase complex (MSC) component glutaminyl-tRNA synthetase (QARS). mascRNA promotes global protein translation and cell proliferation by positively regulating QARS protein levels. Our results uncover a role of mascRNA that is independent of MALAT1, but could be part of the molecular mechanism of MALAT1's function in cancer, and provide a paradigm for understanding tRNA-like structures in mammalian cells.


Subject(s)
RNA, Long Noncoding , RNA, Small Untranslated , Animals , Protein Biosynthesis , RNA Processing, Post-Transcriptional , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism
11.
Stem Cell Reports ; 13(4): 642-656, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31564646

ABSTRACT

Cellular responses to transforming growth factor ß (TGF-ß) depend on cell context. Here, we explored how TGF-ß/nodal signaling crosstalks with the epigenome to promote mesendodermal differentiation. We find that expression of a group of mesendodermal genes depends on both TRIM33 and nodal signaling in embryoid bodies (EBs) but not in embryonic stem cells (ESCs). Only in EBs, TRIM33 binds these genes in the presence of expanded H3K18ac marks. Furthermore, the H3K18ac landscape at mesendodermal genes promotes TRIM33 recruitment. We reveal that HDAC1 binds to active gene promoters and interferes with TRIM33 recruitment to mesendodermal gene promoters. However, the TRIM33-interacting protein p300 deposits H3K18ac and further enhances TRIM33 recruitment. ATAC-seq data demonstrate that TRIM33 primes mesendodermal genes for activation by maintaining chromatin accessibility at their regulatory regions. Altogether, our study suggests that HDAC1 and p300 are key factors linking the epigenome through TRIM33 to the cell context-dependent nodal response during mesendodermal differentiation.


Subject(s)
Cell Differentiation , Histones/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Nodal Protein/metabolism , Signal Transduction , Acetylation , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Promoter Regions, Genetic , Protein Binding , Protein Transport , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism
13.
Nucleic Acids Res ; 47(10): 5436-5448, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31162607

ABSTRACT

HDGF-related protein 3 (HRP3, also known as HDGFL3) belongs to the family of HDGF-related proteins (HRPs) and plays an essential role in hepatocellular carcinoma pathogenesis. All HRPs have a PWWP domain at the N-terminus that binds both histone and DNA substrates. Despite previous advances in PWWP domains, the molecular basis by which HRP3 interacts with chromatin is unclear. In this study, we solved the crystal structures of the HRP3 PWWP domain in complex with various double-stranded DNAs with/without bound histone peptides. We found that HRP3 PWWP bound to the phosphate backbone of the DNA minor groove and showed a preference for DNA molecules bearing a narrow minor groove width. In addition, HRP3 PWWP preferentially bound to histone peptides bearing the H3K36me3/2 modification. HRP3 PWWP uses two adjacent surfaces to bind both DNA and histone substrates simultaneously, enabling us to generate a model illustrating the recruitment of PWWP to H3K36me3-containing nucleosomes. Cell-based analysis indicated that both DNA and histone binding by the HRP3 PWWP domain is important for HRP3 recruitment to chromatin in vivo. Our work establishes that HRP3 PWWP is a new family of minor groove-specific DNA-binding proteins, which improves our understanding of HRP3 and other PWWP domain-containing proteins.


Subject(s)
Chromatin/chemistry , DNA/chemistry , Nuclear Proteins/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Cytoskeletal Proteins , HEK293 Cells , Hep G2 Cells , Histones/chemistry , Humans , Intracellular Signaling Peptides and Proteins , Magnetic Resonance Spectroscopy , Nucleic Acid Conformation , Nucleosomes/chemistry , Peptides/chemistry , Protein Binding , Protein Domains , Static Electricity , Subcellular Fractions , Xenopus laevis
14.
EBioMedicine ; 43: 171-179, 2019 May.
Article in English | MEDLINE | ID: mdl-31060906

ABSTRACT

BACKGROUND: Diffuse intrinsic pontine glioma (DIPG) is a rare and fatal pediatric brain cancer without cure. Seeking therapeutic strategies is still a major challenge in DIPG research. Previous study has shown that dysregulation of G1/S cell cycle checkpoint was common in DIPG and this dysregulation is even more enriched in the H3.3K27 M mutant subgroup. Here we assess potential anti-tumor efficacy of palbociclib, a specific and cytostatic inhibitor of CDK4/6, on high grade H3.3-K27 M-mutant DIPGs in vitro and in vivo. METHODS: We established patient-derived cell lines from treatment-naïve specimens. All the lines have H3.3K27 M mutation. We used a range of biological in vitro assays to assess the effect of palbociclib on growth of DIPGs. Palbociclib activity was also assayed in vivo against three independent DIPG orthotropic xenografts model. FINDINGS: Dysregulation of G1/S cell cycle checkpoint is enriched in these DIPGs. Then, we showed that depletion of CDK4 or CDK6 inhibits DIPG cells growth and blocks G1/S transition. Furthermore, palbociclib effectively repressed DIPG growth in vitro. Transcriptome analysis showed that palbociclib not only blocks G1/S transition, it also blocks other oncogenic targets such as MYC. Finally, palbociclib activity was assayed in vivo against DIPG orthotropic xenografts to demonstrate the high efficiency of blocking tumor growth. INTERPRETATION: Our findings thus revealed that palbociclib could be the therapeutic strategy for treatment-naïve DIPG with H3.3K27 M mutation. FUND: Beijing Municipal Administration of Hospitals Clinical Medicine Development of Special Funding Support, Beijing Municipal Natural Science Foundation, Ministry of Science and Technology of China, and National Natural Science Foundation of China.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Stem Neoplasms/genetics , Glioma/genetics , Histones/genetics , Mutation , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Brain Stem Neoplasms/diagnosis , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Disease Models, Animal , Gene Expression Profiling , Glioma/diagnosis , Glioma/drug therapy , Glioma/metabolism , Humans , Treatment Outcome , Xenograft Model Antitumor Assays
15.
Neurosci Bull ; 35(2): 216-224, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30607770

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is the main cause of brain tumor-related death among children. Until now, there is still a lack of effective therapy with prolonged overall survival for this disease. A typical strategy for preclinical cancer research is to find out the molecular differences between tumor tissue and para-tumor normal tissue, in order to identify potential therapeutic targets. Unfortunately, it is impossible to obtain normal tissue for DIPG because of the vital functions of the pons. Here we report the human fetal hindbrain-derived neural progenitor cells (pontine progenitor cells, PPCs) as normal control cells for DIPG. The PPCs not only harbored similar cell biological and molecular signatures as DIPG glioma stem cells, but also had the potential to be immortalized by the DIPG-specific mutation H3K27M in vitro. These findings provide researchers with a candidate normal control and a potential medicine carrier for preclinical research on DIPG.


Subject(s)
Brain Stem Neoplasms/metabolism , Glioma/metabolism , Neural Stem Cells/metabolism , Pons/metabolism , Animals , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology , Cell Line, Tumor , Cellular Senescence , Female , Glioma/genetics , Glioma/pathology , Histones/genetics , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Pons/embryology , Pons/pathology , Primary Cell Culture
16.
Acta Biochim Biophys Sin (Shanghai) ; 50(3): 322, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29390114
17.
Arch Toxicol ; 92(1): 469-485, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28871336

ABSTRACT

Zoledronate is a bisphosphonate that is widely used in the treatment of metabolic bone diseases. However, zoledronate induces significant nephrotoxicity associated with acute tubular necrosis and renal fibrosis when administered intravenously. There is speculation that zoledronate-induced nephrotoxicity may result from its pharmacological activity as an inhibitor of the mevalonate pathway but the molecular mechanisms are not fully understood. In this report, human proximal tubular HK-2 cells and mouse models were combined to dissect the molecular pathways underlying nephropathy caused by zoledronate treatments. Metabolomic and proteomic assays revealed that multiple cellular processes were significantly disrupted, including the TGFß pathway, fatty acid metabolism and small GTPase signaling in zoledronate-treated HK-2 cells (50 µM) as compared with those in controls. Zoledronate treatments in cells (50 µM) and mice (3 mg/kg) increased TGFß/Smad3 pathway activation to induce fibrosis and kidney injury, and specifically elevated lipid accumulation and expression of fibrotic proteins. Conversely, fatty acid transport protein Slc27a2 deficiency or co-administration of PPARA agonist fenofibrate (20 mg/kg) prevented zoledronate-induced lipid accumulation and kidney fibrosis in mice, indicating that over-expression of fatty acid transporter SLC27A2 and defective fatty acid ß-oxidation following zoledronate treatments were significant factors contributing to its nephrotoxicity. These pharmacological and genetic studies provide an important mechanistic insight into zoledronate-associated kidney toxicity that will aid in development of therapeutic prevention and treatment options for this nephropathy.


Subject(s)
Fatty Acids/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Zoledronic Acid/adverse effects , Animals , Benzamides/pharmacology , Cell Line , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Dioxoles/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fenofibrate/pharmacology , Fibrosis/chemically induced , Humans , Kidney Diseases/pathology , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Lipid Metabolism/drug effects , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Oxidation-Reduction/drug effects , Transforming Growth Factor beta/metabolism
19.
Acta Biochim Biophys Sin (Shanghai) ; 50(1): 60-67, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29190318

ABSTRACT

The transforming growth factor beta (TGF-ß) family of ligands plays major roles in embryonic development, tissue homeostasis, adult immunity, and wound repair. Dysregulation of TGF-ß signaling pathway leads to severe diseases. Its key components have been revealed over the past two decades. This family of cytokines acts by activating receptor activated SMAD (R-SMAD) transcription factors, which in turn modulate the expression of specific sets of target genes. Cells of a multicellular organism have the same genetic information, yet they show structural and functional differences owing to differential expression of their genes. Studies have demonstrated that epigenetic regulation, an integral part of the TGF-ß signaling, enables cells to sense and respond to TGF-ß signaling in a cell context-dependent manner. R-SMAD, as the central transcription factor of TGF-ß signaling, can recruit various epigenetic regulators to shape the transcriptome. In this review, we focus on epigenetic regulatory mechanisms in the TGF-ß signaling during mammalian development and diseases and discuss the central role of the interaction between R-SMAD and various epigenetic regulators in this epigenetic regulation. The crosstalk between TGF-ß signaling and the epigenome could serve as a versatile fine-tuning mechanism for transcriptional regulation during embryonic development and progression of diseases, particularly cancer.


Subject(s)
Epigenesis, Genetic , Epigenomics/methods , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Gene Expression Regulation , Humans , Models, Genetic , Smad Proteins, Receptor-Regulated/metabolism
20.
Nat Commun ; 8(1): 2070, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29234012

ABSTRACT

Smad transcription factors activated by TGF-ß or by BMP receptors form trimeric complexes with Smad4 to target specific genes for cell fate regulation. The CAGAC motif has been considered as the main binding element for Smad2/3/4, whereas Smad1/5/8 have been thought to preferentially bind GC-rich elements. However, chromatin immunoprecipitation analysis in embryonic stem cells showed extensive binding of Smad2/3/4 to GC-rich cis-regulatory elements. Here, we present the structural basis for specific binding of Smad3 and Smad4 to GC-rich motifs in the goosecoid promoter, a nodal-regulated differentiation gene. The structures revealed a 5-bp consensus sequence GGC(GC)|(CG) as the binding site for both TGF-ß and BMP-activated Smads and for Smad4. These 5GC motifs are highly represented as clusters in Smad-bound regions genome-wide. Our results provide a basis for understanding the functional adaptability of Smads in different cellular contexts, and their dependence on lineage-determining transcription factors to target specific genes in TGF-ß and BMP pathways.


Subject(s)
Amino Acid Motifs , Goosecoid Protein/genetics , Smad3 Protein/chemistry , Smad4 Protein/chemistry , Allosteric Regulation/genetics , Animals , Binding Sites/genetics , Bone Morphogenetic Proteins/metabolism , CRISPR-Cas Systems , Cell Lineage/genetics , Crystallography, X-Ray , Gene Expression Regulation, Developmental , Magnetic Resonance Spectroscopy , Mice , Mouse Embryonic Stem Cells , Promoter Regions, Genetic , Protein Binding , Smad3 Protein/genetics , Smad4 Protein/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...