Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Immunol ; 208(3): 753-761, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34996837

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), has seriously threatened global public health. Severe COVID-19 has been reported to be associated with an impaired IFN response. However, the mechanisms of how SARS-CoV-2 antagonizes the host IFN response are poorly understood. In this study, we report that SARS-CoV-2 helicase NSP13 inhibits type I IFN production by directly targeting TANK-binding kinase 1 (TBK1) for degradation. Interestingly, inhibition of autophagy by genetic knockout of Beclin1 or pharmacological inhibition can rescue NSP13-mediated TBK1 degradation in HEK-293T cells. Subsequent studies revealed that NSP13 recruits TBK1 to p62, and the absence of p62 can also inhibit TBK1 degradation in HEK-293T and HeLa cells. Finally, TBK1 and p62 degradation and p62 aggregation were observed during SARS-CoV-2 infection in HeLa-ACE2 and Calu3 cells. Overall, our study shows that NSP13 inhibits type I IFN production by recruiting TBK1 to p62 for autophagic degradation, enabling it to evade the host innate immune response, which provides new insights into the transmission and pathogenesis of SARS-CoV-2 infection.


Subject(s)
Autophagy , COVID-19/immunology , Coronavirus RNA-Dependent RNA Polymerase/physiology , Interferon Type I/biosynthesis , Methyltransferases/physiology , Protein Serine-Threonine Kinases/metabolism , RNA Helicases/physiology , SARS-CoV-2/physiology , Sequestosome-1 Protein/metabolism , Viral Nonstructural Proteins/physiology , Beclin-1/antagonists & inhibitors , Cell Line , Down-Regulation , Humans , Immune Evasion , Immunity, Innate , Immunoprecipitation , Interferon Type I/genetics , Multiprotein Complexes , Protein Aggregates , Protein Interaction Mapping
2.
Appl Opt ; 58(25): 6854-6864, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31503656

ABSTRACT

Optical interferometric techniques provide noncontact, full-field, and high-precision measurements that are very attractive in various research and application fields. Single fringe-pattern processing (SFPP) is often required when measuring fast phenomena, which contain multiple steps including noise removal, phase demodulation, and unwrapping. However, several difficulties are encountered during SFPP, among which the processing time is of interest due to the increasing computational load brought by the large amount and high-resolution fringe patterns in recent years. In this paper, we propose a general and complete graphics processing unit (GPU)-based SFPP framework to perform a systematic discussion on SFPP acceleration. Typical methods from the spatial domain, the transform-based, and the path-related are chosen to have a variety of methods in the framework for better parallelization demonstration, namely, coherence-enhancing diffusion for denoising, spiral phase quadrature transform for demodulation, and quality-guided phase unwrapping. To the best of our knowledge, this is the first time a complete GPU-based framework has been proposed for SFPP. The advantages of performing the analysis and parallelization in framework level are demonstrated, where processing redundancy can be identified and reduced. The proposed framework can be used as an example to demonstrate the GPU-based parallelization in SFPP. Methods in the framework can be replaced but the framework level analysis, the parallel design, and the involved functions are always good references. Experiments are performed on simulated and experimental fringe patterns to demonstrate the effectiveness of the proposed work and achieve at most 29.8 times speedup compared with CPU-based sequential processing.

3.
Biomolecules ; 14(4)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38672517

ABSTRACT

Obesity stands as a significant risk factor for type 2 diabetes, hyperlipidemia, and cardiovascular diseases, intertwining increased inflammation and decreased adipogenesis with metabolic disorders. Studies have highlighted the correlation between Caspase-1 and inflammation in obesity, elucidating its essential role in the biological functions of adipose tissue. However, the impact of Caspase-1 on adipogenesis and the underlying mechanisms remain largely elusive. In our study, we observed a positive correlation between Caspase-1 expression and obesity and its association with adipogenesis. In vivo experiments revealed that, under normal diet conditions, Caspase-1 deficiency improved glucose homeostasis, stimulated subcutaneous adipose tissue expansion, and enhanced adipogenesis. Furthermore, our findings indicate that Caspase-1 deficiency promotes the expression of autophagy-related proteins and inhibits autophagy with 3-MA or CQ blocked Caspase-1 deficiency-induced adipogenesis in vitro. Notably, Caspase-1 deficiency promotes adipogenesis via Atg7-mediated autophagy activation. In addition, Caspase-1 deficiency resisted against high-fat diet-induced obesity and glucose intolerance. Our study proposes the downregulation of Caspase-1 as a promising strategy for mitigating obesity and its associated metabolic disorders.


Subject(s)
Adipogenesis , Autophagy-Related Protein 7 , Autophagy , Caspase 1 , Inflammation , Obesity , Adipogenesis/genetics , Animals , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Mice , Caspase 1/metabolism , Caspase 1/genetics , Caspase 1/deficiency , Obesity/metabolism , Obesity/pathology , Obesity/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/genetics , Male , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , 3T3-L1 Cells , Mice, Knockout
4.
Infect Med (Beijing) ; 1(1): 7-16, 2022 Mar.
Article in English | MEDLINE | ID: mdl-38074973

ABSTRACT

Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 2019 and has led to a global coronavirus disease 2019 (COVID-19) pandemic. Currently, incomplete understanding of how SARS-CoV-2 arrogates the host cell to establish its life cycle has led to slow progress in the development of effective drugs. Results: In this study, we found that SARS-CoV-2 hijacks the host protein EWSR1 (Ewing Sarcoma breakpoint region 1/EWS RNA binding protein 1) to promote the activity of its helicase NSP13 to facilitate viral propagation. NSP13 is highly conserved among coronaviruses and is crucial for virus replication, providing chemical energy to unwind viral RNA replication intermediates. Treatment with different SARS-CoV-2 NSP13 inhibitors in multiple cell lines infected with SARS-CoV-2 effectively suppressed SARS-CoV-2 infection. Using affinity-purification mass spectrometry, the RNA binding protein EWSR1 was then identified as a potent host factor that physically associated with NSP13. Furthermore, silencing EWSR1 dramatically reduced virus replication at both viral RNA and protein levels. Mechanistically, EWSR1 was found to bind to the NTPase domain of NSP13 and potentially enhance its dsRNA unwinding ability. Conclusions: Our results pinpoint EWSR1 as a novel host factor for NSP13 that could potentially be used for drug repurposing as a therapeutic target for COVID-19.

5.
Genes Dis ; 9(2): 429-442, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35224158

ABSTRACT

Obesity-induced inflammation, characterized by augmented infiltration and altered balance of macrophages, is a critical component of systemic insulin resistance. Chemokine-chemokine receptor system plays a vital role in the macrophages accumulation. CC-Chemokine Receptor-like 2 (Ccrl2) is one of the receptors of Chemerin, which is a member of atypical chemokine receptors (ACKR) family, reported taking part in host immune responses and inflammation-related conditions. In our study, we found ccrl2 expression significantly elevated in visceral adipose tissue (VAT) of high fat diet (HFD) induced obese mice and ob/ob mice. Systemic deletion of Ccrl2 gene aggravated HFD induced obesity and insulin resistance and ccrl2 -/- mice showed aggravated VAT inflammation and increased M1/M2 macrophages ratio, which is due to the increase of macrophages chemotaxis in Ccrl2 deficiency mice. Cumulatively, these results indicate that Ccrl2 has a critical function in obesity and obesity-induced insulin resistance via mediating macrophages chemotaxis.

6.
Signal Transduct Target Ther ; 7(1): 312, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36071039

ABSTRACT

The Coronavirus Disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a global pandemic that seriously threatens health and socioeconomic development, but the existed antiviral drugs and vaccines still cannot yet halt the spread of the epidemic. Therefore, a comprehensive and profound understanding of the pathogenesis of SARS-CoV-2 is urgently needed to explore effective therapeutic targets. Here, we conducted a multiomics study of SARS-CoV-2-infected lung epithelial cells, including transcriptomic, proteomic, and ubiquitinomic. Multiomics analysis showed that SARS-CoV-2-infected lung epithelial cells activated strong innate immune response, including interferon and inflammatory responses. Ubiquitinomic further reveals the underlying mechanism of SARS-CoV-2 disrupting the host innate immune response. In addition, SARS-CoV-2 proteins were found to be ubiquitinated during infection despite the fact that SARS-CoV-2 itself didn't code any E3 ligase, and that ubiquitination at three sites on the Spike protein could significantly enhance viral infection. Further screening of the E3 ubiquitin ligases and deubiquitinating enzymes (DUBs) library revealed four E3 ligases influencing SARS-CoV-2 infection, thus providing several new antiviral targets. This multiomics combined with high-throughput screening study reveals that SARS-CoV-2 not only modulates innate immunity, but also promotes viral infection, by hijacking ubiquitination-specific processes, highlighting potential antiviral and anti-inflammation targets.


Subject(s)
COVID-19 , SARS-CoV-2 , Antiviral Agents , Humans , Proteomics , Ubiquitin-Protein Ligases , Ubiquitination/genetics
7.
Front Pharmacol ; 12: 697483, 2021.
Article in English | MEDLINE | ID: mdl-34393781

ABSTRACT

The chronic low-grade inflammation of adipose tissues, primarily mediated by adipose tissue macrophages (ATMs), is the key pathogenic link between obesity and metabolic disorders. Oleanolic acid (OA) is a natural triterpenoid possessing anti-diabetic and anti-inflammation effects, but the machinery is poorly understood. This study investigated the detailed mechanisms of OA on adipose tissue inflammation in obese mice. C57BL/6J mice were fed with high-fat diet (HFD) for 12 weeks, then daily intragastric administrated with vehicle, 25 and 50 mg/kg OA for 4 weeks. Comparing with vehicle, OA administration in obese mice greatly improved insulin resistance, and reduced adipose tissue hypertrophy, ATM infiltration as well as the M1/M2 ratio. The pro-inflammatory markers were significantly down-regulated by OA in both adipose tissue of obese mice and RAW264.7 macrophages treated with interferon gamma/lipopolysaccharide (IFN-γ/LPS). Furthermore, it was found that OA suppressed activation of mitogen-activated protein kinase (MAPK) signaling and NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome through decreasing voltage dependent anion channels (VDAC) expression and reactive oxygen species (ROS) production. This is the first report that oleanolic acid exerts its benefits by affecting mitochondrial function and macrophage activation.

8.
J Med Chem ; 64(18): 13893-13901, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34506138

ABSTRACT

Peptide drugs have the advantages of target specificity and good drugability and have become one of the most increasingly important hotspots in new drug research in biomedical sciences. However, peptide drugs generally have low bioavailability and metabolic stability, and therefore, the modification of existing peptide drugs for the purpose of improving stability and retaining activity is of viable importance. It is known that glucagon is an effective therapy for treating severe hypoglycemia, but its short half-life prevents its wide therapeutic use. Herein, we report that combined unnatural residues and long fatty acid conjugation afford potent α/sulfono-γ-AApeptide hybrid analogues of Glucagon with enhanced stability and prolonged in vivo activity. This strategy could be adopted to develop stabilized analogues of other short-acting bioactive peptides.


Subject(s)
Glucagon/analogs & derivatives , Glucagon/therapeutic use , Hypoglycemia/drug therapy , Amino Acid Sequence , Animals , Female , Glucagon/metabolism , Glucagon/pharmacokinetics , Humans , Male , Mice, Inbred C57BL , Protein Engineering , Protein Stability
9.
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
10.
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
11.
Nat Commun ; 12(1): 164, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431868

ABSTRACT

The current optogenetic toolkit lacks a robust single-component Ca2+-selective ion channel tailored for remote control of Ca2+ signaling in mammals. Existing tools are either derived from engineered channelrhodopsin variants without strict Ca2+ selectivity or based on the stromal interaction molecule 1 (STIM1) that might crosstalk with other targets. Here, we describe the design of a light-operated Ca2+ channel (designated LOCa) by inserting a plant-derived photosensory module into the intracellular loop of an engineered ORAI1 channel. LOCa displays biophysical features reminiscent of the ORAI1 channel, which enables precise optical control over Ca2+ signals and hallmark Ca2+-dependent physiological responses. Furthermore, we demonstrate the use of LOCa to modulate aberrant hematopoietic stem cell self-renewal, transcriptional programming, cell suicide, as well as neurodegeneration in a Drosophila model of amyloidosis.


Subject(s)
Calcium Channels/metabolism , Light , Protein Engineering , Animals , Biophysical Phenomena , Calcium/metabolism , Drosophila/metabolism , HEK293 Cells , HeLa Cells , Humans , Nerve Degeneration/pathology , Optogenetics
12.
Nat Commun ; 11(1): 740, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029739

ABSTRACT

Primary and acquired drug resistance imposes a major threat to achieving optimized clinical outcomes during cancer treatment. Aberrant changes in epigenetic modifications are closely involved in drug resistance of tumor cells. Using BET inhibitor (BETi) resistant leukemia cells as a model system, we demonstrated herein that genome-wide enhancer remodeling played a pivotal role in driving therapeutic resistance via compensational re-expression of pro-survival genes. Capitalizing on the CRISPR interference technology, we identified the second intron of IncRNA, PVT1, as a unique bona fide gained enhancer that restored MYC transcription independent of BRD4 recruitment in leukemia. A combined BETi and CDK7 inhibitor treatment abolished MYC transcription by impeding RNAPII loading without affecting PVT1-mediated chromatin looping at the MYC locus in BETi-resistant leukemia cells. Together, our findings have established the feasibility of targeting enhancer plasticity to overcome drug resistance associated with epigenetic therapies.


Subject(s)
Leukemia, Experimental/drug therapy , Leukemia, Experimental/genetics , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Drug Synergism , Enhancer Elements, Genetic , Female , Genes, myc/drug effects , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Humans , Jurkat Cells , K562 Cells , Leukemia, Experimental/metabolism , Mice , Models, Genetic , Phenylenediamines/administration & dosage , Pyrimidines/administration & dosage , RNA Polymerase II/metabolism , RNA, Long Noncoding/genetics , Cyclin-Dependent Kinase-Activating Kinase
13.
Mol Metab ; 28: 36-47, 2019 10.
Article in English | MEDLINE | ID: mdl-31327757

ABSTRACT

OBJECTIVE: Brown and beige adipocytes in humans and rodents are specialized to burn lipids for heat generation as a natural defense against cold and obesity, which is advantageous to metabolic homeostasis. MicroRNAs as another regulatory layer to regulate metabolic homeostasis attracted a lot of attentions. Our previous work revealed microRNA (miR)-203 as a brown adipocyte-enriched microRNA involved in brown adipocytes development. However, the potential role of miR-203 in adipose tissue metabolic homeostasis has not been determined in vivo. In this study, we investigate the potential role of miR-203 in subcutaneous white adipose tissue (sub-WAT) browning and metabolic homeostasis. METHODS: We investigated the relationship between miR-203 and energy homeostasis in adipose tissue from cold exposed, high fat diet (HFD) fed, ob/ob and db/db mice. The functions of miR-203 on sub-WAT browning were validated through miR-203 knockdown or overexpression. The miR-203 targeted signal pathway was screened by RNAseq analysis. Luciferase report assay, western blot, and qPCR were performed to establish the miR-203 related upstream and downstream signal pathway in vivo and in vitro. The functions of miR-203 on obesity and metabolic homeostasis were validated through GTT/ITT and western blot on high fat diet-induced obesity in C57 mice. ELISA was used to determine the concentration of IFN-γ. Flow cytometry analysis was performed to determine the infiltration of macrophages in adipose tissue. RESULTS: MiR-203 expression positively correlates with energy expenditure, and overexpression of miR-203 could enhance sub-WAT browning in normal diet (ND) condition. Mechanistically, the expression of miR-203 is activated by cAMP-dependent C/EBPß up-regulation. Subsequently, miR-203 inhibits IFN-γ signal pathway activation by directly targeting Lyn, which is an activator of Jak1-Stat1. Moreover, the forced expression of miR-203 could improve insulin sensitivity and resist high fat diet-induced obesity by inhibiting IFN-γ. CONCLUSIONS: MicroRNA-203 (miR-203) promotes white adipose tissue browning in cold exposed mice and improves glucose tolerance in HFD fed mice by repressing IFN-γ. Since miR-203 is activated by cAMP-dependent C/EBPß up-regulation and directly represses IFN-γ signal pathway, we declare that miR-203 acts as a messenger between cAMP signal pathway and IFN-γ signal pathway.


Subject(s)
Adipose Tissue, White/metabolism , Cyclic AMP/metabolism , Interferon-gamma/metabolism , MicroRNAs/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Glucose Tolerance Test , Homeostasis , Injections, Subcutaneous , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Obesity/metabolism
14.
Cancer Lett ; 467: 1-8, 2019 12 28.
Article in English | MEDLINE | ID: mdl-31563562

ABSTRACT

TET2 is among the most frequently mutated genes in hematological malignancies, as well as in healthy individuals with clonal hematopoiesis. Inflammatory stress is known to promote the expansion of Tet2-deficient hematopoietic stem cells, as well as the initiation of pre-leukemic conditions. Infection is one of the most frequent complications in hematological malignancies and antibiotics are commonly used to suppress infection-induced inflammation, but their application in TET2 mutation-associated cancers remained underexplored. In this study, we discovered that Tet2 depletion led to aberrant expansion of myeloid cells, which was correlated with elevated serum levels of pro-inflammatory cytokines at the pre-malignant stage. Antibiotics treatment suppressed the growth of Tet2-deficient myeloid and lymphoid tumor cells in vivo. Transcriptomic profiling further revealed significant changes in the expression of genes involved in the TNF-α signaling and other immunomodulatory pathways in antibiotics-treated tumor cells. Pharmacological inhibition of TNF-α signaling partially attenuated Tet2-deficient tumor cell growth in vivo. Therefore, our findings establish the feasibility of targeting pro-inflammatory pathways to curtail TET2 inactivation-associated hematological malignancies.


Subject(s)
Anti-Bacterial Agents/therapeutic use , DNA-Binding Proteins/genetics , Hematologic Neoplasms/drug therapy , Loss of Function Mutation , Proto-Oncogene Proteins/genetics , Animals , Anti-Bacterial Agents/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cytokines/blood , Dioxygenases , Disease Models, Animal , Female , Gene Expression Profiling , Gene Knockout Techniques , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Mice , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics
15.
J Clin Invest ; 127(8): 2998-3012, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28691928

ABSTRACT

Angioimmunoblastic T cell lymphoma (AITL) represents a distinct, aggressive form of peripheral T cell lymphoma with a dismal prognosis. Recent exome sequencing in patients with AITL has revealed the frequent coexistence of somatic mutations in the Rho GTPase RhoA (RhoAG17V) and loss-of-function mutations in the 5-methylcytosine oxidase TET2. Here, we have demonstrated that TET2 loss and RhoAG17V expression in mature murine T cells cooperatively cause abnormal CD4+ T cell proliferation and differentiation by perturbing FoxO1 gene expression, phosphorylation, and subcellular localization, an abnormality that is also detected in human primary AITL tumor samples. Reexpression of FoxO1 attenuated aberrant immune responses induced in mouse models adoptively transferred with T cells and bearing genetic lesions in both TET2 and RhoA. Our findings suggest a mutational cooperativity between epigenetic factors and GTPases in adult CD4+ T cells that may account for immunoinflammatory responses associated with AITL patients.


Subject(s)
DNA-Binding Proteins/genetics , Mutation , Oxidoreductases/genetics , Proto-Oncogene Proteins/genetics , T-Lymphocytes/immunology , rhoA GTP-Binding Protein/genetics , 5-Methylcytosine , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Cycle , Cell Proliferation , DNA Methylation , Dioxygenases , Epigenesis, Genetic , Female , GTP Phosphohydrolases/metabolism , Homeostasis , Humans , Male , Mice , Phosphorylation , Signal Transduction , rho GTP-Binding Proteins/genetics
16.
Oncotarget ; 7(25): 38347-38358, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27224916

ABSTRACT

Targeting leukemia-initiating cells (LICs) is the key to eradicating leukemia and preventing its relapse. Recent studies have indicated that metabolic regulation may play a critical role in the maintenance of stemness in LICs, although the detailed mechanisms are poorly understood. Herein, we provide intriguing evidence showing that a glucose-responsive transcription factor, carbohydrate responsive element binding protein (ChREBP), served as a tumor suppressor rather than an oncogene, as previously described, to inhibit the development of acute myeloid leukemia by promoting the differentiation of LICs. Using an MLL-AF9-induced murine leukemia model, we demonstrated that the deletion of ChREBP resulted in the blockage of the differentiation of LICs and significantly reduced survival in ChREBP-null leukemic mice. However, ChREBP was not required for the normal repopulation abilities of hematopoietic stem cells. ChREBP promoted leukemia cell differentiation through the direct inhibition of RUNX1 or the transactivation of TXNIP to downregulate the RUNX1 level and ROS generation. Moreover, knockdown of ChREBP in human leukemia THP1 cells led to markedly enhanced proliferation and decreased differentiation upon PMA treatment. Collectively, we unraveled an unexpected role of ChREBP in leukemogenesis, which may provide valuable clues for developing novel metabolic strategies for leukemia treatment.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Thioredoxins/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carcinogenesis/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Thioredoxins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection , Up-Regulation
17.
J Clin Invest ; 126(12): 4537-4553, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27797340

ABSTRACT

Certain secretory proteins are known to be critical for maintaining the stemness of stem cells through autocrine signaling. However, the processes underlying the biogenesis, maturation, and secretion of these proteins remain largely unknown. Here we demonstrate that many secretory proteins produced by hematopoietic stem cells (HSCs) undergo exosomal maturation and release that is controlled by vacuolar protein sorting protein 33b (VPS33B). Deletion of VPS33B in either mouse or human HSCs resulted in impaired exosome maturation and secretion as well as loss of stemness. Additionally, VPS33B deficiency led to a dramatic delay in leukemogenesis. Exosomes purified from either conditioned medium or human plasma could partially rescue the defects of HSCs and leukemia-initiating cells (LICs). VPS33B co-existed in exosomes with GDI2, VPS16B, FLOT1, and other known exosome markers. Mechanistically, VPS33B interacted with the GDI2/RAB11A/RAB27A pathway to regulate the trafficking of secretory proteins as exosomes. These findings reveal an essential role for VPS33B in exosome pathways in HSCs and LICs. Moreover, they shed light on the understanding of vesicle trafficking in other stem cells and on the development of improved strategies for cancer treatment.


Subject(s)
Autocrine Communication , Cell Transformation, Neoplastic/metabolism , Exosomes/metabolism , Hematopoiesis , Leukemia/metabolism , Signal Transduction , Vesicular Transport Proteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Exosomes/genetics , Guanine Nucleotide Dissociation Inhibitors/genetics , Guanine Nucleotide Dissociation Inhibitors/metabolism , HEK293 Cells , Humans , Leukemia/genetics , Leukemia/pathology , Mice , Mice, Knockout , Protein Transport/genetics , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab27 GTP-Binding Proteins
18.
Oncotarget ; 6(25): 21004-15, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26056041

ABSTRACT

Immune inhibitory receptors expressed on various types of immune cells deliver inhibitory signals that maintain the homeostasis of the immune system. Recently we demonstrated that leukocyte immunoglobulin-like receptor subfamily B member 2 (LILRB2) and its murine homolog, paired immunoglobulin-like receptor B (PIRB), are expressed on hematopoietic stem cells and acute myeloid leukemia stem cells and function in maintenance of stemness. Herein, we determined that both LILRB2 and its soluble ligand ANGPTL2 are highly expressed in non-small cell lung cancer (NSCLC) samples, and levels are adversely related to patient prognosis. Inhibition of LILRB2 expression in NSCLC cell lines, such as A549 cells, resulted in a dramatic decrease in proliferation, colony formation, and migration. Mechanistic analyses indicated that ANGPTL2 binds LILRB2 to support the growth of lung cancer cells and that the SHP2/CaMK1/CREB axis controls the proliferation of lung cancer cell lines. Our results suggest that signaling involving ANGPTL2 and LILRB2 is important for lung cancer development and represents a novel target for treatment of this type of cancer.


Subject(s)
Angiopoietins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Ligands , Neoplasm Metastasis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL