Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Aging ; 3(11): 1387-1400, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37884767

ABSTRACT

DNA methylation deregulation at partially methylated domains (PMDs) represents an epigenetic signature of aging and cancer, yet the underlying molecular basis and resulting biological consequences remain unresolved. We report herein a mechanistic link between disrupted DNA methylation at PMDs and the spatial relocalization of H3K9me3-marked heterochromatin in aged hematopoietic stem and progenitor cells (HSPCs) or those with impaired DNA methylation. We uncover that TET2 modulates the spatial redistribution of H3K9me3-marked heterochromatin to mediate the upregulation of endogenous retroviruses (ERVs) and interferon-stimulated genes (ISGs), hence contributing to functional decline of aged HSPCs. TET2-deficient HSPCs retain perinuclear distribution of heterochromatin and exhibit age-related clonal expansion. Reverse transcriptase inhibitors suppress ERVs and ISGs expression, thereby restoring age-related defects in aged HSPCs. Collectively, our findings deepen the understanding of the functional interplay between DNA methylation and histone modifications, which is vital for maintaining heterochromatin function and safeguarding genome stability in stem cells.


Subject(s)
Hematopoietic Stem Cells , Heterochromatin , Heterochromatin/genetics , Hematopoietic Stem Cells/metabolism , DNA Methylation/genetics
2.
Stem Cells ; 40(3): 260-272, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35296897

ABSTRACT

Ten-eleven Translocation (TET) dioxygenases mediated DNA methylation oxidation plays an important role in regulating the embryonic stem cells (ESCs) differentiation. Herein, we utilized a CRISPR/Cas9 based genome editing method to generate single, double, and triple Tet-deficient mouse ESCs (mESCs) and differentiated these cells toward cardiac progenitors. By using emerald green fluorescent protein (GFP; emGFP) expression under the control of Nkx2.5 promoter as marker for cardiac progenitor cells, we discovered that Tet1 and Tet2 depletion significantly impaired mESC-to-cardiac progenitor differentiation. Single-cell RNA-seq analysis further revealed that Tet deletion resulted in the accumulation of mesoderm progenitors to hamper cardiac differentiation. Re-expression of the Tet1 catalytic domain (Tet1CD) rescued the differentiation defect in Tet-triple knockout mESCs. Dead Cas9 (dCas9)-Tet1CD mediated loci-specific epigenome editing at the Hand1 loci validated the direct involvement of Tet-mediated epigenetic modifications in transcriptional regulation during cardiac differentiation. Our study establishes that Tet-mediated epigenetic remodeling is essential for maintaining proper transcriptional outputs to safeguard mESC-to-cardiac progenitor differentiation.


Subject(s)
Mouse Embryonic Stem Cells , Proto-Oncogene Proteins , Animals , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
3.
Adv Sci (Weinh) ; 9(11): e2103940, 2022 04.
Article in English | MEDLINE | ID: mdl-35076181

ABSTRACT

Deregulated store-operated calcium entry (SOCE) mediated by aberrant STIM1-ORAI1 signaling is closely implicated in cancer initiation and progression. Here the authors report the identification of an alternatively spliced variant of STIM1, designated STIM1ß, that harbors an extra exon to encode 31 additional amino acids in the cytoplasmic domain. STIM1ß, highly conserved in mammals, is aberrantly upregulated in glioma tissues to perturb Ca2+ signaling. At the molecular level, the 31-residue insertion destabilizes STIM1ß by perturbing its cytosolic inhibitory domain and accelerating its activation kinetics to efficiently engage and gate ORAI calcium channels. Functionally, STIM1ß depletion affects SOCE in glioblastoma cells, suppresses tumor cell proliferation and growth both in vitro and in vivo. Collectively, their study establishes a splicing variant-specific tumor-promoting role of STIM1ß that can be potentially targeted for glioblastoma intervention.


Subject(s)
Glioblastoma , Animals , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling/physiology , Glioblastoma/genetics , Mammals/metabolism , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
4.
Nat Nanotechnol ; 16(12): 1424-1434, 2021 12.
Article in English | MEDLINE | ID: mdl-34697491

ABSTRACT

Chimeric antigen receptor (CAR) T cell-based immunotherapy, approved by the US Food and Drug Administration, has shown curative potential in patients with haematological malignancies. However, owing to the lack of control over the location and duration of the anti-tumour immune response, CAR T cell therapy still faces safety challenges arising from cytokine release syndrome and on-target, off-tumour toxicity. Herein, we present the design of light-switchable CAR (designated LiCAR) T cells that allow real-time phototunable activation of therapeutic T cells to precisely induce tumour cell killing. When coupled with imaging-guided, surgically removable upconversion nanoplates that have enhanced near-infrared-to-blue upconversion luminescence as miniature deep-tissue photon transducers, LiCAR T cells enable both spatial and temporal control over T cell-mediated anti-tumour therapeutic activity in vivo with greatly mitigated side effects. Our nano-optogenetic immunomodulation platform not only provides a unique approach to interrogate CAR-mediated anti-tumour immunity, but also sets the stage for developing precision medicine to deliver personalized anticancer therapy.


Subject(s)
Immunotherapy, Adoptive , Nanotechnology , Optogenetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Animals , Cell Death , Female , Humans , Immunity , Jurkat Cells , Lymphocyte Activation/immunology , Lymphoma/immunology , Lymphoma/pathology , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL
5.
Cancer Res ; 81(8): 1965-1976, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33589517

ABSTRACT

Inactivation of tumor-infiltrating lymphocytes (TIL) is one of the mechanisms mitigating antitumor immunity during tumor onset and progression. Epigenetic abnormalities are regarded as a major culprit contributing to the dysfunction of TILs within tumor microenvironments. In this study, we used a murine model of melanoma to discover that Tet2 inactivation significantly enhances the antitumor activity of TILs with an efficacy comparable to immune checkpoint inhibition imposed by anti-PD-L1 treatment. Single-cell RNA-sequencing analysis suggested that Tet2-deficient TILs exhibit effector-like features. Transcriptomic and ATAC-sequencing analysis showed that Tet2 ablation reshapes chromatin accessibility and favors binding of transcription factors geared toward CD8+ T-cell activation. Furthermore, the ETS family of transcription factors contributed to augmented CD8+ T-cell function following Tet2 depletion. Overall, our study establishes that Tet2 constitutes one of the epigenetic barriers that account for dysfunction of TILs and that Tet2 inactivation could promote antitumor immunity to suppress tumor growth. SIGNIFICANCE: This study suggests that ablation of TET2+ from TILs could promote their antitumor function by reshaping chromatin accessibility for key transcription factors and enhancing the transcription of genes essential for antitumor activity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/deficiency , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/immunology , Proto-Oncogene Proteins/deficiency , Adoptive Transfer/methods , Animals , Chromatin/metabolism , DNA Demethylation , DNA-Binding Proteins/genetics , Dioxygenases , Disease Models, Animal , Epigenesis, Genetic , Gene Deletion , Gene Silencing , Immune Checkpoint Inhibitors/therapeutic use , MAP Kinase Kinase Kinases , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Perforin/metabolism , Proto-Oncogene Proteins/genetics , Sequence Analysis, RNA , Transcription Factors/metabolism , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/metabolism
6.
Nat Commun ; 10(1): 4297, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541101

ABSTRACT

Tet-mediated DNA demethylation plays an important role in shaping the epigenetic landscape and chromatin accessibility to control gene expression. While several studies demonstrated pivotal roles of Tet in regulating embryonic development, little is known about their functions in heart development. Here we analyze DNA methylation and hydroxymethylation dynamics during early cardiac development in both human and mice. We find that cardiac-specific deletion of Tet2 and Tet3 in mice (Tet2/3-DKO) leads to ventricular non-compaction cardiomyopathy (NCC) with embryonic lethality. Single-cell RNA-seq analyses reveal a reduction in cardiomyocyte numbers and transcriptional reprogramming in cardiac tissues upon Tet2/3 depletion. Impaired DNA demethylation and reduced chromatin accessibility in Tet2/3-DKO mice further compromised Ying-yang1 (YY1) binding to its genomic targets, and perturbed high-order chromatin organization at key genes involved in heart development. Our studies provide evidence of the physiological role of Tet in regulating DNA methylation dynamics and chromatin organization during early heart development.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Embryonic Development/physiology , Organogenesis/physiology , Proto-Oncogene Proteins/metabolism , YY1 Transcription Factor/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Catalytic Domain , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA Demethylation , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Embryonic Development/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Heart/embryology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Proto-Oncogene Proteins/genetics
7.
Oncogene ; 38(11): 1905-1919, 2019 03.
Article in English | MEDLINE | ID: mdl-30390073

ABSTRACT

Tumor cells with p53 inactivation frequently exhibit chemotherapy resistance, which poses a long-standing challenge to cancer treatment. Here we unveiled a previously unrecognized role of TET2 in mediating p53-loss induced chemotherapy resistance in colon cancer. Deletion of TET2 in p53-null colon cancer cells enhanced DNA damage and restored chemotherapy sensitivity. By taking a two-pronged approach that combined pharmacological inhibition with genetic depletion, we discovered that p53 destabilized TET2 at the protein level by promoting its autophagic degradation. At the molecular level, we further revealed a physical association between TET2 and p53 that facilitated the nucleoplasmic shuttling of TET2, as well as its recruitment to the autophagosome for degradation. Our study has unveiled a functional interplay between TET2 and p53 during anti-cancer therapy. Our findings establish the rationale for targeting TET2 to overcome chemotherapy resistance associated with mutant p53 tumors.


Subject(s)
Autophagy , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/physiology , Animals , Autophagy/genetics , Dioxygenases , Drug Resistance, Neoplasm/genetics , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Proteolysis , Tumor Cells, Cultured
8.
Sci Rep ; 7: 44165, 2017 03 08.
Article in English | MEDLINE | ID: mdl-28272434

ABSTRACT

Transcription factor TBX1 plays a pivotal role in heart development and has been implicated in 22q11.2 deletion syndrome. The structure of this protein has been elucidated, and several mutations have been identified that disrupt TBX1 localization, DNA/protein-binding, or mRNA expression. This study reports a mutation in the TBX1 gene that leads to significantly reduced expression of the mutant protein. A total of 773 conotruncal heart defect patients and 516 unrelated healthy control individuals were enrolled, none of which harbored a 22q11.2 deletion or duplication. We identified a mutation, c.303-305delGAA, located in the third exon of TBX1 that does not disrupt TBX1 mRNA expression or DNA binding activity, but results in decreased TBX1 protein levels and transcriptional activity. Through protein degradation studies we demonstrated that TBX1 is degraded primarily in proteasomes. Although the c.303-305delGAA mutation leads to low levels of the mutant protein, we found that increased protein degradation was not the cause, and we hypothesize that an alternate mechanism, such as translational inhibition, may be the cause.


Subject(s)
Base Sequence , Gene Expression Regulation , Heart Defects, Congenital , Proteolysis , Sequence Deletion , T-Box Domain Proteins , Transcription, Genetic , Child, Preschool , Female , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Humans , Male , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics
9.
Elife ; 42015 Dec 08.
Article in English | MEDLINE | ID: mdl-26646180

ABSTRACT

The application of current channelrhodopsin-based optogenetic tools is limited by the lack of strict ion selectivity and the inability to extend the spectra sensitivity into the near-infrared (NIR) tissue transmissible range. Here we present an NIR-stimulable optogenetic platform (termed 'Opto-CRAC') that selectively and remotely controls Ca(2+) oscillations and Ca(2+)-responsive gene expression to regulate the function of non-excitable cells, including T lymphocytes, macrophages and dendritic cells. When coupled to upconversion nanoparticles, the optogenetic operation window is shifted from the visible range to NIR wavelengths to enable wireless photoactivation of Ca(2+)-dependent signaling and optogenetic modulation of immunoinflammatory responses. In a mouse model of melanoma by using ovalbumin as surrogate tumor antigen, Opto-CRAC has been shown to act as a genetically-encoded 'photoactivatable adjuvant' to improve antigen-specific immune responses to specifically destruct tumor cells. Our study represents a solid step forward towards the goal of achieving remote and wireless control of Ca(2+)-modulated activities with tailored function.


Subject(s)
Calcium Signaling/radiation effects , Immunomodulation , Infrared Rays , Optogenetics/methods , Animals , Dendritic Cells/physiology , Dendritic Cells/radiation effects , Disease Models, Animal , Macrophages/physiology , Macrophages/radiation effects , Melanoma/immunology , Melanoma/therapy , Mice , T-Lymphocytes/physiology , T-Lymphocytes/radiation effects
10.
Cell Physiol Biochem ; 35(1): 270-80, 2015.
Article in English | MEDLINE | ID: mdl-25591769

ABSTRACT

BACKGROUND: Cornelia de Lange Syndrome (CdLS) is a rare but severe clinically heterogeneous developmental disorder characterized by facial dysmorphia, growth and cognitive retardation, and abnormalities of limb development. OBJECTIVES: To determine the pathogenesis of a patient with CdLS. METHODS: We studied a patient with CdLS by whole exome sequencing, karyotyping and Agilent CGH Array. The results were confirmed by quantitative real-time PCR analysis of the patient and her parents. Further comparison of our patient and cases with partially overlapping deletions retrieved from the literature and databases was undertaken. RESULTS: Whole exome sequencing had excluded the mutation of cohesion genes such as NIPBL,SMC1A and SMC3. The result of karyotyping showed a deletion of chromosome 9q31.1-q32 and the result of Agilent CGH Array further displayed a 12.01-Mb region of deletion at chromosome bands 9q31.1-q32. Reported cases with the deletion of 9q31.1-q32 share similar features with our CdLS patient. One of the genes in the deleted region, SMC2, belongs to the Structural Maintenance of Chromosomes (SMC) family and regulates gene expression and DNA repair. CONCLUSIONS: Patients carrying the deletion of 9q31.1-q32 showed similar phenotypes with CdLS.


Subject(s)
Chromosomes, Human, Pair 9 , De Lange Syndrome/genetics , Comparative Genomic Hybridization , De Lange Syndrome/pathology , Echocardiography , Female , High-Throughput Nucleotide Sequencing , Humans , Infant , Karyotyping , Phenotype , Sequence Analysis, DNA
11.
BMC Med Genet ; 15: 78, 2014 Jul 06.
Article in English | MEDLINE | ID: mdl-24998776

ABSTRACT

BACKGROUND: TBX1 and CRKL haploinsufficiency is thought to cause the cardiac phenotype of the 22q11.2 deletion syndrome. However, few unequivocal mutations of TBX1 and CRKL have been discovered in isolated conotrucal heart defects (CTDs) patients. The aim of the study was to screen the mutation of TBX1 and CRKL in isolated CTDs Chinese patients without 22q11.2 deletion and identify the pathomechanism of the missense mutations. METHODS: We enrolled 199 non-22q11.2 deletion patients with CTDs and 139 unrelated healthy controls. Gene sequencing were performed for all of them. The functional data of mutations were obtained by in vitro transfection and luciferase experiments and computer modelling. RESULTS: Screening of the TBX1 coding sequence identified a de novo missense mutation (c.385G → A; p.E129K) and a known polymorphism (c.928G → A; p.G310S). In vitro experiments demonstrate that the TBX1E129K variant almost lost transactivation activity. The TBX1G310S variant seems to affect the interaction of TBX1 with other factors. Computer molecular dynamics simulations showed the de novo missense mutation is likely to affect TBX1-DNA interaction. No mutation of CRKL gene was found. CONCLUSIONS: These observations suggest that the TBX1 loss-of-function mutation may be involved in the pathogenesis of isolated CTDs. This is the first human missense mutation showing that TBX1 is a candidate causing isolated CTDs in Chinese patients without 22q11.2 deletion.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DiGeorge Syndrome/genetics , Heart Defects, Congenital/genetics , Nuclear Proteins/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Animals , Asian People/genetics , COS Cells , Case-Control Studies , Chlorocebus aethiops , DNA/chemistry , DNA/metabolism , DiGeorge Syndrome/pathology , Exons , Female , HEK293 Cells , Heart Defects, Congenital/pathology , Humans , Male , Molecular Dynamics Simulation , Mutation, Missense , Phylogeny , Protein Conformation , Protein Structure, Secondary , Sequence Analysis, DNA , T-Box Domain Proteins/chemistry
12.
PLoS One ; 8(8): e72515, 2013.
Article in English | MEDLINE | ID: mdl-24009689

ABSTRACT

OBJECTIVE: To determine the pathogenesis of a patient born with congenital heart defects, who had appeared normal in prenatal screening. METHODS: In routine prenatal screening, G-banding was performed to analyse the karyotypes of the family and fluorescence in situ hybridization was used to investigate the 22q11.2 deletion in the fetus. After birth, the child was found to be suffering from heart defects by transthoracic echocardiography. In the following study, sequencing was used to search for potential mutations in pivotal genes. SNP-array was employed for fine mapping of the aberrant region and quantitative real-time PCR was used to confirm the results. Furthermore, other patients with a similar phenotype were screened for the same genetic variations. To compare with a control, these variations were also assessed in the general population. RESULTS: The child and his mother each had a region that was deleted in the beta-defensin repeats, which are usually duplicated in the general population. Besides, the child carried a SOX7-gene duplication. While this duplication was not detected in his mother, it was found in two other patients with cardiac defects who also had the similar deletion in the beta-defensin repeats. CONCLUSION: The congenital heart defects of the child were probably caused by a SOX7-gene duplication, which may be a consequence of the partial haplotype of beta-defensin regions at 8p23.1. To our knowledge, this is the first congenital heart defect case found to have the haplotype of beta-defensin and the duplication of SOX7.


Subject(s)
Gene Duplication , Haplotypes , Heart Defects, Congenital/genetics , SOXF Transcription Factors/genetics , beta-Defensins/genetics , Adult , Child , Chromosome Deletion , Chromosomes, Human, Pair 8 , Comparative Genomic Hybridization , Echocardiography , Female , Genetic Association Studies , Heart Defects, Congenital/diagnosis , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...