Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.014
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 25(3): 525-536, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38356061

ABSTRACT

Regulatory T (Treg) cells are critical for immune tolerance but also form a barrier to antitumor immunity. As therapeutic strategies involving Treg cell depletion are limited by concurrent autoimmune disorders, identification of intratumoral Treg cell-specific regulatory mechanisms is needed for selective targeting. Epigenetic modulators can be targeted with small compounds, but intratumoral Treg cell-specific epigenetic regulators have been unexplored. Here, we show that JMJD1C, a histone demethylase upregulated by cytokines in the tumor microenvironment, is essential for tumor Treg cell fitness but dispensable for systemic immune homeostasis. JMJD1C deletion enhanced AKT signals in a manner dependent on histone H3 lysine 9 dimethylation (H3K9me2) demethylase and STAT3 signals independently of H3K9me2 demethylase, leading to robust interferon-γ production and tumor Treg cell fragility. We have also developed an oral JMJD1C inhibitor that suppresses tumor growth by targeting intratumoral Treg cells. Overall, this study identifies JMJD1C as an epigenetic hub that can integrate signals to establish tumor Treg cell fitness, and we present a specific JMJD1C inhibitor that can target tumor Treg cells without affecting systemic immune homeostasis.


Subject(s)
Autoimmune Diseases , Humans , Cytokines , Epigenomics , Histone Demethylases , Homeostasis , Oxidoreductases, N-Demethylating , Jumonji Domain-Containing Histone Demethylases/genetics
2.
Cell ; 182(1): 245-261.e17, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32649877

ABSTRACT

Genomic studies of lung adenocarcinoma (LUAD) have advanced our understanding of the disease's biology and accelerated targeted therapy. However, the proteomic characteristics of LUAD remain poorly understood. We carried out a comprehensive proteomics analysis of 103 cases of LUAD in Chinese patients. Integrative analysis of proteome, phosphoproteome, transcriptome, and whole-exome sequencing data revealed cancer-associated characteristics, such as tumor-associated protein variants, distinct proteomics features, and clinical outcomes in patients at an early stage or with EGFR and TP53 mutations. Proteome-based stratification of LUAD revealed three subtypes (S-I, S-II, and S-III) related to different clinical and molecular features. Further, we nominated potential drug targets and validated the plasma protein level of HSP 90ß as a potential prognostic biomarker for LUAD in an independent cohort. Our integrative proteomics analysis enables a more comprehensive understanding of the molecular landscape of LUAD and offers an opportunity for more precise diagnosis and treatment.


Subject(s)
Adenocarcinoma of Lung/metabolism , Lung Neoplasms/metabolism , Proteomics , Adenocarcinoma of Lung/genetics , Asian People/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Drug Delivery Systems , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Male , Middle Aged , Mutation/genetics , Neoplasm Staging , Phosphoproteins/metabolism , Principal Component Analysis , Prognosis , Proteome/metabolism , Treatment Outcome , Tumor Suppressor Protein p53/genetics
3.
Cell ; 167(6): 1525-1539.e17, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912060

ABSTRACT

Poorly immunogenic tumor cells evade host immunity and grow even in the presence of an intact immune system, but the complex mechanisms regulating tumor immunogenicity have not been elucidated. Here, we discovered an unexpected role of the Hippo pathway in suppressing anti-tumor immunity. We demonstrate that, in three different murine syngeneic tumor models (B16, SCC7, and 4T1), loss of the Hippo pathway kinases LATS1/2 (large tumor suppressor 1 and 2) in tumor cells inhibits tumor growth. Tumor regression by LATS1/2 deletion requires adaptive immune responses, and LATS1/2 deficiency enhances tumor vaccine efficacy. Mechanistically, LATS1/2-null tumor cells secrete nucleic-acid-rich extracellular vesicles, which induce a type I interferon response via the Toll-like receptors-MYD88/TRIF pathway. LATS1/2 deletion in tumors thus improves tumor immunogenicity, leading to tumor destruction by enhancing anti-tumor immune responses. Our observations uncover a key role of the Hippo pathway in modulating tumor immunogenicity and demonstrate a proof of concept for targeting LATS1/2 in cancer immunotherapy.


Subject(s)
Immune Tolerance , Neoplasms/immunology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cancer Vaccines/immunology , Gene Deletion , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Toll-Like Receptors/metabolism , Tumor Suppressor Proteins/genetics
4.
Nat Immunol ; 19(4): 354-365, 2018 04.
Article in English | MEDLINE | ID: mdl-29563620

ABSTRACT

Mechanisms that degrade inflammatory mRNAs are well known; however, stabilizing mechanisms are poorly understood. Here, we show that Act1, an interleukin-17 (IL-17)-receptor-complex adaptor, binds and stabilizes mRNAs encoding key inflammatory proteins. The Act1 SEFIR domain binds a stem-loop structure, the SEFIR-binding element (SBE), in the 3' untranslated region (UTR) of Cxcl1 mRNA, encoding an inflammatory chemokine. mRNA-bound Act1 directs formation of three compartmentally distinct RNA-protein complexes (RNPs) that regulate three disparate events in inflammatory-mRNA metabolism: preventing mRNA decay in the nucleus, inhibiting mRNA decapping in P bodies and promoting translation. SBE RNA aptamers decreased IL-17-mediated mRNA stabilization in vitro, IL-17-induced skin inflammation and airway inflammation in a mouse asthma model, thus providing a therapeutic strategy for autoimmune diseases. These results reveal a network in which Act1 assembles RNPs on the 3' UTRs of select mRNAs and consequently controls receptor-mediated mRNA stabilization and translation during inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Inflammation/immunology , Interleukin-17/metabolism , RNA Stability/physiology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Gene Expression Regulation/immunology , Inflammation/metabolism , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Receptors, Interleukin-17/metabolism
5.
Cell ; 163(4): 960-74, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26544941

ABSTRACT

Alterations in estrogen-mediated cellular signaling play an essential role in the pathogenesis of endometriosis. In addition to higher estrogen receptor (ER) ß levels, enhanced ERß activity was detected in endometriotic tissues, and the inhibition of enhanced ERß activity by an ERß-selective antagonist suppressed mouse ectopic lesion growth. Notably, gain of ERß function stimulated the progression of endometriosis. As a mechanism to evade endogenous immune surveillance for cell survival, ERß interacts with cellular apoptotic machinery in the cytoplasm to inhibit TNF-α-induced apoptosis. ERß also interacts with components of the cytoplasmic inflammasome to increase interleukin-1ß and thus enhance its cellular adhesion and proliferation properties. Furthermore, this gain of ERß function enhances epithelial-mesenchymal transition signaling, thereby increasing the invasion activity of endometriotic tissues for establishment of ectopic lesions. Collectively, we reveal how endometrial tissue generated by retrograde menstruation can escape immune surveillance and develop into sustained ectopic lesions via gain of ERß function.


Subject(s)
Endometriosis/pathology , Estrogen Receptor beta/metabolism , Inflammasomes/metabolism , Menstruation/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Proliferation , Endometriosis/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Immunologic Surveillance , Interleukin-1beta/metabolism , Mice , Tumor Necrosis Factor-alpha/metabolism
6.
Mol Cell ; 82(23): 4519-4536.e7, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36384137

ABSTRACT

Nutrient sensing and damage sensing are two fundamental processes in living organisms. While hyperglycemia is frequently linked to diabetes-related vulnerability to microbial infection, how body glucose levels affect innate immune responses to microbial invasion is not fully understood. Here, we surprisingly found that viral infection led to a rapid and dramatic decrease in blood glucose levels in rodents, leading to robust AMPK activation. AMPK, once activated, directly phosphorylates TBK1 at S511, which triggers IRF3 recruitment and the assembly of MAVS or STING signalosomes. Consistently, ablation or inhibition of AMPK, knockin of TBK1-S511A, or increased glucose levels compromised nucleic acid sensing, while boosting AMPK-TBK1 cascade by AICAR or TBK1-S511E knockin improves antiviral immunity substantially in various animal models. Thus, we identify TBK1 as an AMPK substrate, reveal the molecular mechanism coupling a dual sensing of glucose and nuclei acids, and report its physiological necessity in antiviral defense.


Subject(s)
AMP-Activated Protein Kinases , Nucleic Acids , Animals , AMP-Activated Protein Kinases/genetics , Immunity, Innate , Antiviral Agents , Glucose
7.
Mol Cell ; 81(13): 2736-2751.e8, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33932349

ABSTRACT

Cholesterol metabolism is tightly associated with colorectal cancer (CRC). Nevertheless, the clinical benefit of statins, the inhibitor of cholesterol biogenesis mevalonate (MVA) pathway, is inconclusive, possibly because of a lack of patient stratification criteria. Here, we describe that YAP-mediated zinc finger MYND-type containing 8 (ZMYND8) expression sensitizes intestinal tumors to the inhibition of the MVA pathway. We show that the oncogenic activity of YAP relies largely on ZMYND8 to enhance intracellular de novo cholesterol biogenesis. Disruption of the ZMYND8-dependent MVA pathway greatly restricts the self-renewal capacity of Lgr5+ intestinal stem cells (ISCs) and intestinal tumorigenesis. Mechanistically, ZMYND8 and SREBP2 drive the enhancer-promoter interaction to facilitate the recruitment of Mediator complex, thus upregulating MVA pathway genes. Together, our results establish that the epigenetic reader ZMYND8 endows YAP-high intestinal cancer with metabolic vulnerability.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colorectal Neoplasms/metabolism , Mevalonic Acid/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Mice , Mice, Transgenic , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
8.
Mol Cell ; 81(20): 4147-4164.e7, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34453890

ABSTRACT

Missense mutations of the tumor suppressor Neurofibromin 2 (NF2/Merlin/schwannomin) result in sporadic to frequent occurrences of tumorigenesis in multiple organs. However, the underlying pathogenicity of NF2-related tumorigenesis remains mostly unknown. Here we found that NF2 facilitated innate immunity by regulating YAP/TAZ-mediated TBK1 inhibition. Unexpectedly, patient-derived individual mutations in the FERM domain of NF2 (NF2m) converted NF2 into a potent suppressor of cGAS-STING signaling. Mechanistically, NF2m gained extreme associations with IRF3 and TBK1 and, upon innate nucleic acid sensing, was directly induced by the activated IRF3 to form cellular condensates, which contained the PP2A complex, to eliminate TBK1 activation. Accordingly, NF2m robustly suppressed STING-initiated antitumor immunity in cancer cell-autonomous and -nonautonomous murine models, and NF2m-IRF3 condensates were evident in human vestibular schwannomas. Our study reports phase separation-mediated quiescence of cGAS-STING signaling by a mutant tumor suppressor and reveals gain-of-function pathogenesis for NF2-related tumors by regulating antitumor immunity.


Subject(s)
Immunity, Innate , Membrane Proteins/metabolism , Mutation, Missense , Neoplasms/metabolism , Neurofibromin 2/metabolism , Nucleotidyltransferases/metabolism , Tumor Escape , Animals , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neurofibromin 2/genetics , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
9.
Nature ; 605(7910): 457-463, 2022 05.
Article in English | MEDLINE | ID: mdl-35585341

ABSTRACT

Microcombs have sparked a surge of applications over the past decade, ranging from optical communications to metrology1-4. Despite their diverse deployment, most microcomb-based systems rely on a large amount of bulky elements and equipment to fulfil their desired functions, which is complicated, expensive and power consuming. By contrast, foundry-based silicon photonics (SiPh) has had remarkable success in providing versatile functionality in a scalable and low-cost manner5-7, but its available chip-based light sources lack the capacity for parallelization, which limits the scope of SiPh applications. Here we combine these two technologies by using a power-efficient and operationally simple aluminium-gallium-arsenide-on-insulator microcomb source to drive complementary metal-oxide-semiconductor SiPh engines. We present two important chip-scale photonic systems for optical data transmission and microwave photonics, respectively. A microcomb-based integrated photonic data link is demonstrated, based on a pulse-amplitude four-level modulation scheme with a two-terabit-per-second aggregate rate, and a highly reconfigurable microwave photonic filter with a high level of integration is constructed using a time-stretch approach. Such synergy of a microcomb and SiPh integrated components is an essential step towards the next generation of fully integrated photonic systems.

10.
Mol Cell ; 80(5): 810-827.e7, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33171123

ABSTRACT

Mitochondrial morphology shifts rapidly to manage cellular metabolism, organelle integrity, and cell fate. It remains unknown whether innate nucleic acid sensing, the central and general mechanisms of monitoring both microbial invasion and cellular damage, can reprogram and govern mitochondrial dynamics and function. Here, we unexpectedly observed that upon activation of RIG-I-like receptor (RLR)-MAVS signaling, TBK1 directly phosphorylated DRP1/DNM1L, which disabled DRP1, preventing its high-order oligomerization and mitochondrial fragmentation function. The TBK1-DRP1 axis was essential for assembly of large MAVS aggregates and healthy antiviral immunity and underlay nutrient-triggered mitochondrial dynamics and cell fate determination. Knockin (KI) strategies mimicking TBK1-DRP1 signaling produced dominant-negative phenotypes reminiscent of human DRP1 inborn mutations, while interrupting the TBK1-DRP1 connection compromised antiviral responses. Thus, our findings establish an unrecognized function of innate immunity governing both morphology and physiology of a major organelle, identify a lacking loop during innate RNA sensing, and report an elegant mechanism of shaping mitochondrial dynamics.


Subject(s)
Dynamins/metabolism , Mitochondria/physiology , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , Zebrafish/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Dynamins/genetics , HCT116 Cells , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Mutation , Protein Serine-Threonine Kinases/genetics , RNA/genetics , Signal Transduction/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
11.
Mol Cell ; 80(6): 1013-1024.e6, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33338401

ABSTRACT

Impaired DNA crosslink repair leads to Fanconi anemia (FA), characterized by a unique manifestation of bone marrow failure and pancytopenia among diseases caused by DNA damage response defects. As a germline disorder, why the hematopoietic hierarchy is specifically affected is not fully understood. We find that reprogramming transcription during hematopoietic differentiation results in an overload of genotoxic stress, which causes aborted differentiation and depletion of FA mutant progenitor cells. DNA damage onset most likely arises from formaldehyde, an obligate by-product of oxidative protein demethylation during transcription regulation. Our results demonstrate that rapid and extensive transcription reprogramming associated with hematopoietic differentiation poses a major threat to genome stability and cell viability in the absence of the FA pathway. The connection between differentiation and DNA damage accumulation reveals a novel mechanism of genome scarring and is critical to exploring therapies to counteract the aplastic anemia for the treatment of FA patients.


Subject(s)
Cell Differentiation/drug effects , Cellular Reprogramming/genetics , Fanconi Anemia/genetics , Formaldehyde/toxicity , DNA Damage/drug effects , DNA Repair/genetics , Fanconi Anemia/blood , Fanconi Anemia/pathology , Formaldehyde/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Genomic Instability/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Humans , K562 Cells , Transcription, Genetic
12.
Cell ; 145(5): 787-99, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21620140

ABSTRACT

Elucidation of endogenous cellular protein-protein interactions and their networks is most desirable for biological studies. Here we report our study of endogenous human coregulator protein complex networks obtained from integrative mass spectrometry-based analysis of 3290 affinity purifications. By preserving weak protein interactions during complex isolation and utilizing high levels of reciprocity in the large dataset, we identified many unreported protein associations, such as a transcriptional network formed by ZMYND8, ZNF687, and ZNF592. Furthermore, our work revealed a tiered interplay within networks that share common proteins, providing a conceptual organization of a cellular proteome composed of minimal endogenous modules (MEMOs), complex isoforms (uniCOREs), and regulatory complex-complex interaction networks (CCIs). This resource will effectively fill a void in linking correlative genomic studies with an understanding of transcriptional regulatory protein functions within the proteome for formulation and testing of future hypotheses.


Subject(s)
Proteins/metabolism , Proteome/analysis , Amino Acid Sequence , BRCA1 Protein/metabolism , Genome-Wide Association Study , Humans , Immunoprecipitation , Mass Spectrometry , Molecular Sequence Data , Protein Interaction Mapping , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic
13.
Mol Cell Proteomics ; 23(5): 100756, 2024 May.
Article in English | MEDLINE | ID: mdl-38554776

ABSTRACT

In orthotopic mouse tumor models, tumor progression is a complex process, involving interactions among tumor cells, host cell-derived stromal cells, and immune cells. Much attention has been focused on the tumor and its tumor microenvironment, while the host's macroenvironment including immune organs in response to tumorigenesis is poorly understood. Here, we report a temporal proteomic analysis on a subcutaneous tumor and three immune organs (LN, MLN, and spleen) collected on Days 0, 3, 7, 10, 14, and 21 after inoculation of mouse forestomach cancer cells in a syngeneic mouse model. Bioinformatics analysis identified key biological processes during distinct tumor development phases, including an initial acute immune response, the attack by the host immune system, followed by the adaptive immune activation, and the build-up of extracellular matrix. Proteomic changes in LN and spleen largely recapitulated the dynamics of the immune response in the tumor, consistent with an acute defense response on D3, adaptive immune response on D10, and immune evasion by D21. In contrast, the immune response in MLN showed a gradual and sustained activation, suggesting a delayed response from a distal immune organ. Combined analyses of tumors and host immune organs allowed the identification of potential therapeutic targets. A proof-of-concept experiment demonstrated that significant growth reduction can be achieved by dual inhibition of MEK and DDR2. Together, our temporal proteomic dataset of tumors and immune organs provides a useful resource for understanding the interaction between tumors and the immune system and has the potential for identifying new therapeutic targets for cancer treatment.


Subject(s)
Proteomics , Spleen , Animals , Proteomics/methods , Mice , Spleen/metabolism , Tumor Microenvironment , Cell Line, Tumor , Mice, Inbred C57BL , Lymph Nodes/metabolism , Proteome/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Female
14.
Proc Natl Acad Sci U S A ; 120(30): e2221809120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37459541

ABSTRACT

Early in the COVID-19 pandemic, data suggested that males had a higher risk of developing severe disease and that androgen deprivation therapy might be associated with protection. Combined with the fact that TMPRSS2 (transmembrane serine protease 2), a host entry factor for the SARS-CoV-2 virus, was a well-known androgen-regulated gene, this led to an upsurge of research investigating androgen receptor (AR)-targeting drugs. Proxalutamide, an AR antagonist, was shown in initial clinical studies to benefit COVID-19 patients; however, further validation is needed as one study was retracted. Due to continued interest in proxalutamide, which is in phase 3 trials, we examined its ability to impact SARS-CoV-2 infection and downstream inflammatory responses. Proxalutamide exerted similar effects as enzalutamide, an AR antagonist prescribed for advanced prostate cancer, in decreasing AR signaling and expression of TMPRSS2 and angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 receptor. However, proxalutamide led to degradation of AR protein, which was not observed with enzalutamide. Proxalutamide inhibited SARS-CoV-2 infection with an IC50 value of 97 nM, compared to 281 nM for enzalutamide. Importantly, proxalutamide inhibited infection by multiple SARS-CoV-2 variants and synergized with remdesivir. Proxalutamide protected against cell death in response to tumor necrosis factor alpha and interferon gamma, and overall survival of mice was increased with proxalutamide treatment prior to cytokine exposure. Mechanistically, we found that proxalutamide increased levels of NRF2, an essential transcription factor that mediates antioxidant responses, and decreased lung inflammation. These data provide compelling evidence that proxalutamide can prevent SARS-CoV-2 infection and cytokine-induced lung damage, suggesting that promising clinical data may emerge from ongoing phase 3 trials.


Subject(s)
COVID-19 , Prostatic Neoplasms , Male , Humans , Animals , Mice , SARS-CoV-2/metabolism , Androgens , Androgen Antagonists/therapeutic use , Pandemics , Peptidyl-Dipeptidase A/metabolism , Prostatic Neoplasms/drug therapy , Interferon-gamma/therapeutic use
15.
Plant Physiol ; 195(2): 1347-1364, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38488068

ABSTRACT

Potato (Solanum tuberosum L.) is cultivated worldwide for its underground tubers, which provide an important part of human nutrition and serve as a model system for belowground storage organ formation. Similar to flowering, stolon-expressed FLOWERING LOCUS T-like (FT-like) protein SELF-PRUNING 6A (StSP6A) plays an instrumental role in tuberization by binding to the bZIP transcription factors StABI5-like 1 (StABL1) and StFD-like 1 (StFDL1), causing transcriptional reprogramming at the stolon subapical apices. However, the molecular mechanism regulating the widely conserved FT-bZIP interactions remains largely unexplored. Here, we identified a TCP transcription factor StAST1 (StABL1 and StSP6A-associated TCP protein 1) binding to both StSP6A and StABL1. StAST1 is specifically expressed in the vascular tissue of leaves and developing stolons. Silencing of StAST1 leads to accelerated tuberization and a shortened life cycle. Molecular dissection reveals that the interaction of StAST1 with StSP6A and StABL1 attenuates the formation of the alternative tuberigen activation complex (aTAC). We also observed StAST1 directly activates the expression of potato GA 20-oxidase gene (StGA20ox1) to regulate GA responses. These results demonstrate StAST1 functions as a tuberization repressor by regulating plant hormone levels; our findings also suggest a mechanism by which the widely conserved FT-FD genetic module is fine-tuned.


Subject(s)
Gene Expression Regulation, Plant , Plant Proteins , Plant Tubers , Solanum tuberosum , Transcription Factors , Solanum tuberosum/genetics , Solanum tuberosum/metabolism , Solanum tuberosum/physiology , Solanum tuberosum/growth & development , Plant Tubers/genetics , Plant Tubers/growth & development , Plant Tubers/metabolism , Plant Tubers/physiology , Plant Proteins/metabolism , Plant Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
16.
Blood ; 141(21): 2629-2641, 2023 05 25.
Article in English | MEDLINE | ID: mdl-36867840

ABSTRACT

The communication of talin-activated integrin αIIbß3 with the cytoskeleton (integrin outside-in signaling) is essential for platelet aggregation, wound healing, and hemostasis. Filamin, a large actin crosslinker and integrin binding partner critical for cell spreading and migration, is implicated as a key regulator of integrin outside-in signaling. However, the current dogma is that filamin, which stabilizes inactive αIIbß3, is displaced from αIIbß3 by talin to promote the integrin activation (inside-out signaling), and how filamin further functions remains unresolved. Here, we show that while associating with the inactive αIIbß3, filamin also associates with the talin-bound active αIIbß3 to mediate platelet spreading. Fluorescence resonance energy transfer-based analysis reveals that while associating with both αIIb and ß3 cytoplasmic tails (CTs) to maintain the inactive αIIbß3, filamin is spatiotemporally rearranged to associate with αIIb CT alone on activated αIIbß3. Consistently, confocal cell imaging indicates that integrin α CT-linked filamin gradually delocalizes from the ß CT-linked focal adhesion marker-vinculin likely because of the separation of integrin α/ß CTs occurring during integrin activation. High-resolution crystal and nuclear magnetic resonance structure determinations unravel that the activated integrin αIIb CT binds to filamin via a striking α-helix→ß-strand transition with a strengthened affinity that is dependent on the integrin-activating membrane environment containing enriched phosphatidylinositol 4,5-bisphosphate. These data suggest a novel integrin αIIb CT-filamin-actin linkage that promotes integrin outside-in signaling. Consistently, disruption of such linkage impairs the activation state of αIIbß3, phosphorylation of focal adhesion kinase/proto-oncogene tyrosine kinase Src, and cell migration. Together, our findings advance the fundamental understanding of integrin outside-in signaling with broad implications in blood physiology and pathology.


Subject(s)
Platelet Glycoprotein GPIIb-IIIa Complex , Platelet Membrane Glycoprotein IIb , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoprotein IIb/metabolism , Actins/metabolism , Filamins/metabolism , Talin/metabolism , Blood Platelets/metabolism
17.
Plant Cell ; 34(10): 4088-4104, 2022 09 27.
Article in English | MEDLINE | ID: mdl-35863056

ABSTRACT

Plants utilize localized cell-surface and intracellular receptors to sense microbes and activate the influx of calcium, which serves as an important second messenger in eukaryotes to regulate cellular responses. However, the mechanisms through which plants decipher calcium influx to activate immune responses remain largely unknown. Here, we show that pathogen-associated molecular patterns (PAMPs) trigger calcium-dependent phosphorylation of CAM-BINDING PROTEIN 60-LIKE G (CBP60g) in Arabidopsis (Arabidopsis thaliana). CALCIUM-DEPENDENT PROTEIN KINASE5 (CPK5) phosphorylates CBP60g directly, thereby enhancing its transcription factor activity. TOUCH 3 (TCH3) and its homologs CALMODULIN (CAM) 1/4/6 and CPK4/5/6/11 are required for PAMP-induced CBP60g phosphorylation. TCH3 interferes with the auto-inhibitory region of CPK5 and promotes CPK5-mediated CBP60g phosphorylation. Furthermore, CPKs-mediated CBP60g phosphorylation positively regulates plant resistance to soil-borne fungal pathogens. These lines of evidence uncover a novel calcium signal decoding mechanism during plant immunity through which TCH3 relieves auto-inhibition of CPK5 to phosphorylate and activate CBP60g. The findings reveal cooperative interconnections between different types of calcium sensors in eukaryotes.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Calcium/metabolism , Calcium-Binding Proteins , Calmodulin/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Phosphorylation , Protein Kinases , Soil , Transcription Factors/metabolism
18.
Circ Res ; 133(5): 400-411, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37492967

ABSTRACT

BACKGROUND: FLNC (filamin C), a member of the filamin family predominantly expressed in striated muscles, plays a crucial role in bridging the cytoskeleton and ECM (extracellular matrix) in cardiomyocytes, thereby maintaining heart integrity and function. Although genetic variants within the N-terminal ABD (actin-binding domain) of FLNC have been identified in patients with cardiomyopathy, the precise contribution of the actin-binding capability to FLNC's function in mammalian hearts remains poorly understood. METHODS: We conducted in silico analysis of the 3-dimensional structure of mouse FLNC to identify key amino acid residues within the ABD that are essential for FLNC's actin-binding capacity. Subsequently, we performed coimmunoprecipitation and immunofluorescent assays to validate the in silico findings and assess the impact of these mutations on the interactions with other binding partners and the subcellular localization of FLNC. Additionally, we generated and analyzed knock-in mouse models in which the FLNC-actin interaction was completely disrupted by these mutations. RESULTS: Our findings revealed that F93A/L98E mutations completely disrupted FLNC-actin interaction while preserving FLNC's ability to interact with other binding partners ITGB1 (ß1 integrin) and γ-SAG (γ-sarcoglycan), as well as maintaining FLNC subcellular localization. Loss of FLNC-actin interaction in embryonic cardiomyocytes resulted in embryonic lethality and cardiac developmental defects, including ventricular wall malformation and reduced cardiomyocyte proliferation. Moreover, disruption of FLNC-actin interaction in adult cardiomyocytes led to severe dilated cardiomyopathy, enhanced lethality and dysregulation of key cytoskeleton components. CONCLUSIONS: Our data strongly support the crucial role of FLNC as a bridge between actin filaments and ECM through its interactions with actin, ITGB1, γ-SAG, and other associated proteins in cardiomyocytes. Disruption of FLN-actin interaction may result in detachment of actin filaments from the extracellular matrix, ultimately impairing normal cardiac development and function. These findings also provide insights into mechanisms underlying cardiomyopathy associated with genetic variants in FLNC ABD and other regions.


Subject(s)
Actins , Cardiomyopathies , Mice , Animals , Filamins/genetics , Filamins/metabolism , Actins/genetics , Actins/metabolism , Muscle, Skeletal/metabolism , Cardiomyopathies/genetics , Myocytes, Cardiac/metabolism , Mutation , Mammals
19.
J Pathol ; 264(1): 68-79, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39022843

ABSTRACT

Metastasis is the primary culprit behind cancer-related fatalities in multiple cancer types, including prostate cancer. Despite great advances, the precise mechanisms underlying prostate cancer metastasis are far from complete. By using a transgenic mouse prostate cancer model (TRAMP) with and without Phf8 knockout, we have identified a crucial role of PHF8 in prostate cancer metastasis. By complexing with E2F1, PHF8 transcriptionally upregulates SNAI1 in a demethylation-dependent manner. The upregulated SNAI1 subsequently enhances epithelial-to-mesenchymal transition (EMT) and metastasis. Given the role of the abnormally activated PHF8/E2F1-SNAI1 axis in prostate cancer metastasis and poor prognosis, the levels of PHF8 or the activity of this axis could serve as biomarkers for prostate cancer metastasis. Moreover, targeting this axis could become a potential therapeutic strategy for prostate cancer treatment. © 2024 The Pathological Society of Great Britain and Ireland.


Subject(s)
E2F1 Transcription Factor , Epithelial-Mesenchymal Transition , Histone Demethylases , Prostatic Neoplasms , Snail Family Transcription Factors , Transcription Factors , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/enzymology , Animals , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Mice , Histone Demethylases/metabolism , Histone Demethylases/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Mice, Knockout , Signal Transduction , Neoplasm Metastasis , Mice, Transgenic , Cell Movement
20.
Nature ; 567(7747): 257-261, 2019 03.
Article in English | MEDLINE | ID: mdl-30814741

ABSTRACT

Hepatocellular carcinoma is the third leading cause of deaths from cancer worldwide. Infection with the hepatitis B virus is one of the leading risk factors for developing hepatocellular carcinoma, particularly in East Asia1. Although surgical treatment may be effective in the early stages, the five-year overall rate of survival after developing this cancer is only 50-70%2. Here, using proteomic and phospho-proteomic profiling, we characterize 110 paired tumour and non-tumour tissues of clinical early-stage hepatocellular carcinoma related to hepatitis B virus infection. Our quantitative proteomic data highlight heterogeneity in early-stage hepatocellular carcinoma: we used this to stratify the cohort into the subtypes S-I, S-II and S-III, each of which has a different clinical outcome. S-III, which is characterized by disrupted cholesterol homeostasis, is associated with the lowest overall rate of survival and the greatest risk of a poor prognosis after first-line surgery. The knockdown of sterol O-acyltransferase 1 (SOAT1)-high expression of which is a signature specific to the S-III subtype-alters the distribution of cellular cholesterol, and effectively suppresses the proliferation and migration of hepatocellular carcinoma. Finally, on the basis of a patient-derived tumour xenograft mouse model of hepatocellular carcinoma, we found that treatment with avasimibe, an inhibitor of SOAT1, markedly reduced the size of tumours that had high levels of SOAT1 expression. The proteomic stratification of early-stage hepatocellular carcinoma presented in this study provides insight into the tumour biology of this cancer, and suggests opportunities for personalized therapies that target it.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Molecular Targeted Therapy/trends , Proteomics , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Growth Processes , Cell Movement , Hepatitis B virus/pathogenicity , Humans , Liver Neoplasms/pathology , Liver Neoplasms/virology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Staging , Prognosis , Sterol O-Acyltransferase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL