Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Nat Cell Biol ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251719

ABSTRACT

The ten-eleven translocation (TET) family of dioxygenases maintain stable local DNA demethylation during cell division and lineage specification. As the major catalytic product of TET enzymes, 5-hydroxymethylcytosine is selectively enriched at specific genomic regions, such as enhancers, in a tissue-dependent manner. However, the mechanisms underlying this selectivity remain unresolved. Here we unveil a low-complexity insert domain within TET2 that facilitates its biomolecular condensation with epigenetic modulators, such as UTX and MLL4. This co-condensation fosters a permissive chromatin environment for precise DNA demethylation. Disrupting low-complexity insert-mediated condensation alters the genomic binding of TET2 to cause promiscuous DNA demethylation and genome reorganization. These changes influence the expression of key genes implicated in leukaemogenesis to curtail leukaemia cell proliferation. Collectively, this study establishes the pivotal role of TET2 condensation in orchestrating precise DNA demethylation and gene transcription to support tumour cell growth.

2.
Nat Commun ; 14(1): 5461, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37673917

ABSTRACT

The cGAS-STING signaling pathway has emerged as a promising target for immunotherapy development. Here, we introduce a light-sensitive optogenetic device for control of the cGAS/STING signaling to conditionally modulate innate immunity, called 'light-inducible SMOC-like repeats' (LiSmore). We demonstrate that photo-activated LiSmore boosts dendritic cell (DC) maturation and antigen presentation with high spatiotemporal precision. This non-invasive approach photo-sensitizes cytotoxic T lymphocytes to engage tumor antigens, leading to a sustained antitumor immune response. When combined with an immune checkpoint blocker (ICB), LiSmore improves antitumor efficacy in an immunosuppressive lung cancer model that is otherwise unresponsive to conventional ICB treatment. Additionally, LiSmore exhibits an abscopal effect by effectively suppressing tumor growth in a distal site in a bilateral mouse model of melanoma. Collectively, our findings establish the potential of targeted optogenetic activation of the STING signaling pathway for remote immunomodulation in mice.


Subject(s)
Neoplasms , Optogenetics , Animals , Mice , Immunotherapy , Immunomodulation , Antigen Presentation , Chromogranin A , Nucleotidyltransferases
3.
Sci Adv ; 9(13): eadg1123, 2023 03 31.
Article in English | MEDLINE | ID: mdl-37000871

ABSTRACT

Biomolecular condensates participate in the regulation of gene transcription, yet the relationship between nuclear condensation and transcriptional activation remains elusive. Here, we devised a biotinylated CRISPR-dCas9-based optogenetic method, light-activated macromolecular phase separation (LAMPS), to enable inducible formation, affinity purification, and multiomic dissection of nuclear condensates at the targeted genomic loci. LAMPS-induced condensation at enhancers and promoters activates endogenous gene transcription by chromatin reconfiguration, causing increased chromatin accessibility and de novo formation of long-range chromosomal loops. Proteomic profiling of light-induced condensates by dCas9-mediated affinity purification uncovers multivalent interaction-dependent remodeling of macromolecular composition, resulting in the selective enrichment of transcriptional coactivators and chromatin structure proteins. Our findings support a model whereby the formation of nuclear condensates at native genomic loci reconfigures chromatin architecture and multiprotein assemblies to modulate gene transcription. Hence, LAMPS facilitates mechanistic interrogation of the relationship between nuclear condensation, genome structure, and gene transcription in living cells.


Subject(s)
Chromatin , Proteomics , Chromatin/genetics , Cell Nucleus/genetics , Transcription Factors/genetics , Genome
4.
Biochem J ; 479(17): 1857-1875, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36111979

ABSTRACT

Membrane contact sites (MCSs) mediate crucial physiological processes in eukaryotic cells, including ion signaling, lipid metabolism, and autophagy. Dysregulation of MCSs is closely related to various diseases, such as type 2 diabetes mellitus (T2DM), neurodegenerative diseases, and cancers. Visualization, proteomic mapping and manipulation of MCSs may help the dissection of the physiology and pathology MCSs. Recent technical advances have enabled better understanding of the dynamics and functions of MCSs. Here we present a summary of currently known functions of MCSs, with a focus on optical approaches to visualize and manipulate MCSs, as well as proteomic mapping within MCSs.


Subject(s)
Diabetes Mellitus, Type 2 , Endoplasmic Reticulum , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mitochondrial Membranes/metabolism , Optogenetics , Proteomics
5.
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
6.
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
7.
Semin Cancer Biol ; 83: 487-502, 2022 08.
Article in English | MEDLINE | ID: mdl-33421619

ABSTRACT

Drug resistance, either intrinsic or acquired, represents a major hurdle to achieving optimal therapeutic outcomes during cancer treatment. In addition to acquisition of resistance-conferring genetic mutations, accumulating evidence suggests an intimate involvement of the epigenetic machinery in this process as well. Recent studies have revealed that epigenetic reprogramming, such as altered expression or relocation of DNA/histone modulators accompanied with chromatin structure remodeling, can lead to transcriptional plasticity in tumor cells, thereby driving their transformation towards a persistent state. These "persisters" represent a pool of slow-growing cells that can either re-expand when treatment is discontinued or acquire permanent resistance. Targeting epigenetic reprogramming or plasticity represents a new strategy to prevent the emergence of drug-refractory populations and to enable more consistent clinical responses. With the growing numbers of drugs or drug candidates developed to target epigenetic regulators, more and more epigenetic therapies are under preclinical evaluation, early clinical trials or approved by FDA as single agent or in combination with existing antitumor drugs. In this review, we highlight latest discoveries in the mechanistic understanding of epigenetically-induced drug resistance. In parallel, we discuss the potential of combining epigenetic drugs with existing anticancer regimens as a promising strategy for overcoming cancer drug resistance.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , DNA Methylation , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Epigenomics , Humans , Neoplasms/drug therapy , Neoplasms/genetics
8.
Methods Enzymol ; 654: 255-270, 2021.
Article in English | MEDLINE | ID: mdl-34120716

ABSTRACT

Optogenetics combines optics and genetics to enable non-invasive interrogation of cell physiology at an unprecedented high spatiotemporal resolution. Here, we introduce Opto-CRAC as a set of genetically-encoded calcium actuators (GECAs) engineered from the calcium release-activated calcium (CRAC) channel, which has been tailored for optical control of calcium entry and calcium-dependent physiological responses in non-excitable cells and tissues. We describe a detailed protocol for applying Opto-CRAC as an optogenetic tool to achieve photo-tunable control over intracellular calcium signals and calcium-dependent gene expression in mammalian cells.


Subject(s)
Calcium Release Activated Calcium Channels , Calcium , Animals , Calcium/metabolism , Calcium Release Activated Calcium Channels/metabolism , Calcium Signaling , Optogenetics
9.
J Mol Biol ; 432(10): 3127-3136, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32277990

ABSTRACT

To expand the repertoire of chemogenetic tools tailored for molecular and cellular engineering, we describe herein the design of cpRAPID as a circularly permuted rapamycin-inducible dimerization system composed of the canonical FK506-binding protein (FKBP) and circular permutants of FKBP12-rapamycin binding domain (cpFRB). By permuting the topology of the four helices within FRB, we have created cpFRB-FKBP pairs that respond to ligand with varying activation kinetics and dynamics. The cpRAPID system enables chemical-controllable subcellular redistribution of proteins, as well as inducible transcriptional activation when coupled with the CRISPR activation (CRISPRa) technology to induce a GFP reporter and endogenous gene expression. We have further demonstrated the use of cpRAPID to generate chemically switchable split nanobody (designated Chessbody) for ligand-gated antigen recognition in living cells. Collectively, the circular permutation approach offers a powerful means for diversifying the chemogenetics toolbox to benefit the burgeoning synthetic biology field.


Subject(s)
Protein Engineering/methods , Recombinant Proteins/metabolism , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Proteins/genetics , Cloning, Molecular , HEK293 Cells , HeLa Cells , Humans , Ligands , Protein Domains , Protein Structure, Secondary , Recombinant Proteins/chemistry , Sirolimus/pharmacology , Synthetic Biology , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Proteins/metabolism
10.
Nat Commun ; 11(1): 1039, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32098964

ABSTRACT

Genetically encoded photoswitches have enabled spatial and temporal control of cellular events to achieve tailored functions in living cells, but their applications to probe the structure-function relations of signaling proteins are still underexplored. We illustrate herein the incorporation of various blue light-responsive photoreceptors into modular domains of the stromal interaction molecule 1 (STIM1) to manipulate protein activity and faithfully recapitulate STIM1-mediated signaling events. Capitalizing on these optogenetic tools, we identify the molecular determinants required to mediate protein oligomerization, intramolecular conformational switch, and protein-target interactions. In parallel, we have applied these synthetic devices to enable light-inducible gating of calcium channels, conformational switch, dynamic protein-microtubule interactions and assembly of membrane contact sites in a reversible manner. Our optogenetic engineering approach can be broadly applied to aid the mechanistic dissection of cell signaling, as well as non-invasive interrogation of physiological processes with high precision.


Subject(s)
Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Optogenetics/methods , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Arabidopsis Proteins/genetics , Bacterial Proteins/genetics , Calcium/metabolism , Cryptochromes/genetics , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Ion Channel Gating , Luminescent Proteins/genetics , Mutation , Neoplasm Proteins/chemistry , Neoplasms/genetics , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Protein Engineering/methods , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Stromal Interaction Molecule 1/chemistry , Structure-Activity Relationship
11.
Cell Calcium ; 87: 102165, 2020 05.
Article in English | MEDLINE | ID: mdl-32004817

ABSTRACT

Genetically Encoded Calcium Indicators (GECIs) are powerful molecular tools for monitoring calcium (Ca2+) signaling in the cytosol and organellar compartments. However, currently available ratiometric indicators that allow measurements of resting Ca2+ levels have limitations in long-term Ca2+ imaging. They either are ultraviolet (UV)-excited ones with strong photo-toxicity, or have poor performance. To overcome this hurdle, we developed a set of visible light excited ratiometric-GECIs (VR-GECIs) based on existing mono-colored GECIs. With performance comparable to their corresponding mono-color prototypes, this set of VR-GECIs enables long-term measurements of intra-cellular or intra-organellar Ca2+ signals. Using these VR-GECIs together with a newly developed off-line analysis tool, we achieved long-term measurements of Ca2+ homeostasis of moving or dividing cells. Our tools may find broad applications in decoding Ca2+-modulated physiological or pathological processes.


Subject(s)
Calcium Signaling/radiation effects , Calcium/metabolism , Light , Cell Shape , Endoplasmic Reticulum/metabolism , Fluorescence , HEK293 Cells , HeLa Cells , Humans , Mitosis
SELECTION OF CITATIONS
SEARCH DETAIL