Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters








Publication year range
1.
Adv Sci (Weinh) ; 11(18): e2303752, 2024 May.
Article in English | MEDLINE | ID: mdl-38311573

ABSTRACT

Loss of refreshment in nucleus pulposus (NP) cellularity leads to intervertebral disc (IVD) degeneration. Nevertheless, the cellular sequence of NP cell differentiation remains unclear, although an increasing body of literature has identified markers of NP progenitor cells (NPPCs). Notably, due to their fragility, the physical enrichment of NP-derived cells has limited conventional transcriptomic approaches in multiple studies. To overcome this limitation, a spatially resolved transcriptional atlas of the mouse IVD is generated via the 10x Genomics Visium platform dividing NP spots into two clusters. Based on this, most reported NPPC-markers, including Cathepsin K (Ctsk), are rare and predominantly located within the NP-outer subset. Cell lineage tracing further evidence that a small number of Ctsk-expressing cells generate the entire adult NP tissue. In contrast, Tie2, which has long suggested labeling NPPCs, is actually neither expressed in NP subsets nor labels NPPCs and their descendants in mouse models; consistent with this, an in situ sequencing (ISS) analysis validated the absence of Tie2 in NP tissue. Similarly, no Tie2-cre-mediated labeling of NPPCs is observed in an IVD degenerative mouse model. Altogether, in this study, the first spatial transcriptomic map of the IVD is established, thereby providing a public resource for bone biology.


Subject(s)
Nucleus Pulposus , Stem Cells , Transcriptome , Animals , Mice , Nucleus Pulposus/metabolism , Nucleus Pulposus/cytology , Stem Cells/metabolism , Transcriptome/genetics , Cell Differentiation/genetics , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Gene Expression Profiling/methods , Disease Models, Animal
2.
Nurse Educ Pract ; 73: 103842, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37984161

ABSTRACT

BACKGROUND: Humanistic practice ability serves as an indispensable skills that newly graduated nurses strive to develop. Yet, there is a dearth of knowledge regarding the role of organizational socialization in mediating the association between transition shock and humanistic practice ability in Chinese newly graduated nurses. AIM: To breakdown the association between humanistic practice ability and transition shock among Chinese newly graduated nurses, with a simultaneous concentration on the mediating effect of organizational socialization in the association. DESIGN: Utilizing a descriptive cross-sectional study design, this research aimed to provide a comprehensive overview of the variables being examined. METHODS: A web-based survey was completed by 417 newly graduated nurses from three general public hospitals in Shandong Province, eastern China from February to March 2023. Three questionnaires were administered: the Nurse Humanistic Practice Ability Scale (NHPAS), the Organizational Socialization Questionnaire(OSQ) and the Transition shock of Newly Graduated Nurses Scale (TSNGNS). The IBM SPSS 22.0, AMOS 22.0 and GraphPad Prism 9.0.0 was applied for figure preparation and statistical analyses. RESULTS: Findings indicated a significant statistical association among organizational socialization, transition shock and humanities practicing ability. A significant negative correlation was uncovered between OrS and TrS (r=-0.468, p<0.001), significant and negative correlation (r = -0.412, p < 0.001) was unmasked between the TrS and the HPA,whereas a significant positive correlation was observed between OrS and HPA (r=0.641, p<0.001). Moreover, in the mediation models, organizational socialization was identified as a partial mediating role in the association between transition shock and humanities practicing ability (indirect effect -0.324, p<0.001; direct effect -0.100, p =0.026; total effect -0.424, p =0.001). CONCLUSION/IMPLICATIONS FOR PRACTICE: TrS has a significantly negative influence on HPA among newly graduated nurses, and OrS partially mediating the negative effect of TrS on HPA. Nursing managers and nursing educators can reduce the adverse consequences resulted from TrS through enhancing the benefits of organizational socialization programs and eventually improve the level of HPA of newly graduated nurses.


Subject(s)
Nurses , Socialization , Humans , Cross-Sectional Studies , East Asian People , Surveys and Questionnaires , Workplace
3.
Prehosp Disaster Med ; 38(4): 485-494, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37485671

ABSTRACT

BACKGROUND: Decreasing symptom-to-door (S2D) delay is of vital importance for reducing morbidity and mortality in patients with ST-segment elevation myocardial infarction (STEMI). The factors associated with S2D delay in STEMI patients have not been well-characterized. OBJECTIVES: The aim of this study was to identify factors associated with S2D delay in patients with STEMI. METHODS: The PubMed, CINAHL, and Embase databases were searched for data. References from the selected articles and relevant background papers were also manually searched to identify additional eligible studies. The included articles were reviewed and assessed for risk of bias. The level of evidence for each identified factor was evaluated using a semiquantitative synthesis. RESULTS: Twelve (12) papers were included in the review. Factors associated with S2D delay were complex and could be divided into sociodemographic, clinical history, and onset characteristics. The level of evidence regarding female sex and diabetes was strong, and the evidence was moderate regarding older age, smoking, history of hypertension, self-transport, or referral. CONCLUSIONS: Female sex, older age, previous diabetes, previous hypertension, smoking, and self-transport are all strong or moderate risk factors for S2D time delay in patients with ST-segment myocardial infarction. More efforts should be made to educate at-risk populations concerning symptoms of STEMI and the importance of seeking early medical assistance.


Subject(s)
ST Elevation Myocardial Infarction , Time-to-Treatment , Delivery of Health Care , Patient Acceptance of Health Care , Risk Factors , ST Elevation Myocardial Infarction/therapy , Treatment Delay , Humans
4.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37328286

ABSTRACT

BACKGROUND: Immune exclusion (IE) where tumors deter the infiltration of immune cells into the tumor microenvironment has emerged as a key mechanism underlying immunotherapy resistance. We recently reported a novel role of discoidin domain-containing receptor 1 (DDR1) in promoting IE in breast cancer and validated its critical role in IE using neutralizing rabbit monoclonal antibodies (mAbs) in multiple mouse tumor models. METHODS: To develop a DDR1-targeting mAb as a potential cancer therapeutic, we humanized mAb9 with a complementarity-determining region grafting strategy. The humanized antibody named PRTH-101 is currently being tested in a Phase 1 clinical trial. We determined the binding epitope of PRTH-101 from the crystal structure of the complex between DDR1 extracellular domain (ECD) and the PRTH-101 Fab fragment with 3.15 Å resolution. We revealed the underlying mechanisms of action of PRTH-101 using both cell culture assays and in vivo study in a mouse tumor model. RESULTS: PRTH-101 has subnanomolar affinity to DDR1 and potent antitumor efficacy similar to the parental rabbit mAb after humanization. Structural information illustrated that PRTH-101 interacts with the discoidin (DS)-like domain, but not the collagen-binding DS domain of DDR1. Mechanistically, we showed that PRTH-101 inhibited DDR1 phosphorylation, decreased collagen-mediated cell attachment, and significantly blocked DDR1 shedding from the cell surface. Treatment of tumor-bearing mice with PRTH-101 in vivo disrupted collagen fiber alignment (a physical barrier) in the tumor extracellular matrix (ECM) and enhanced CD8+ T cell infiltration in tumors. CONCLUSIONS: This study not only paves a pathway for the development of PRTH-101 as a cancer therapeutic, but also sheds light on a new therapeutic strategy to modulate collagen alignment in the tumor ECM for enhancing antitumor immunity.


Subject(s)
Antibodies, Monoclonal , Discoidin Domain Receptor 1 , Neoplasms , Animals , Mice , Collagen/metabolism , Discoidin Domain Receptor 1/metabolism , Extracellular Matrix/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Microenvironment , Antibodies, Monoclonal/pharmacology
5.
Nature ; 599(7886): 673-678, 2021 11.
Article in English | MEDLINE | ID: mdl-34732895

ABSTRACT

Immune exclusion predicts poor patient outcomes in multiple malignancies, including triple-negative breast cancer (TNBC)1. The extracellular matrix (ECM) contributes to immune exclusion2. However, strategies to reduce ECM abundance are largely ineffective or generate undesired outcomes3,4. Here we show that discoidin domain receptor 1 (DDR1), a collagen receptor with tyrosine kinase activity5, instigates immune exclusion by promoting collagen fibre alignment. Ablation of Ddr1 in tumours promotes the intratumoral penetration of T cells and obliterates tumour growth in mouse models of TNBC. Supporting this finding, in human TNBC the expression of DDR1 negatively correlates with the intratumoral abundance of anti-tumour T cells. The DDR1 extracellular domain (DDR1-ECD), but not its intracellular kinase domain, is required for immune exclusion. Membrane-untethered DDR1-ECD is sufficient to rescue the growth of Ddr1-knockout tumours in immunocompetent hosts. Mechanistically, the binding of DDR1-ECD to collagen enforces aligned collagen fibres and obstructs immune infiltration. ECD-neutralizing antibodies disrupt collagen fibre alignment, mitigate immune exclusion and inhibit tumour growth in immunocompetent hosts. Together, our findings identify a mechanism for immune exclusion and suggest an immunotherapeutic target for increasing immune accessibility through reconfiguration of the tumour ECM.


Subject(s)
Collagen/metabolism , Discoidin Domain Receptor 1/metabolism , Extracellular Matrix/metabolism , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Tumor Escape , Animals , Cell Line, Tumor , Discoidin Domain Receptor 1/antagonists & inhibitors , Discoidin Domain Receptor 1/deficiency , Discoidin Domain Receptor 1/genetics , Disease Models, Animal , Extracellular Matrix/immunology , Female , Gene Deletion , Gene Knockout Techniques , Humans , Immunocompetence/immunology , Immunotherapy , Mice , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Triple Negative Breast Neoplasms/therapy
6.
Cancer Med ; 10(6): 2137-2152, 2021 03.
Article in English | MEDLINE | ID: mdl-33626233

ABSTRACT

Tumor cell-intrinsic programmed death-ligand 1 (PD-L1) signals mediate immunopathologic effects in breast, colon, and ovarian cancers and in melanomas, but bladder cancer (BC) effects are unreported. We show here that BC cell-intrinsic PD-L1 signals in mouse MB49 and human RT4, UM-UC3, and UM-UC-14 BC cells regulate important pathologic pathways and processes, including effects not reported in other cancers. α-PD-L1 antibodies reduced BC cell proliferation in vitro, demonstrating direct signaling effects. BC cell-intrinsic PD-L1 promoted mammalian target of rapamycin complex 1 (mTORC1) signals in vitro and augmented in vivo immune-independent cell growth and metastatic cancer spread, similar to effects we reported in melanoma and ovarian cancer. BC cell-intrinsic PD-L1 signals also promoted basal and stress-induced autophagy, whereas these signals inhibited autophagy in melanoma and ovarian cancer cells. BC cell-intrinsic PD-L1 also mediated chemotherapy resistance to the commonly used BC chemotherapy agents cis-platinum and gemcitabine and to the mTORC1 inhibitor, rapamycin. Thus, BC cell-intrinsic PD-L1 signals regulate important virulence and treatment resistance pathways that suggest novel, actionable treatment targets meriting additional studies. As a proof-of-concept, we showed that the autophagy inhibitor chloroquine improved cis-platinum treatment efficacy in vivo, with greater efficacy in PD-L1 null versus PD-L1-replete BC.


Subject(s)
Autophagy/physiology , B7-H1 Antigen/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Urinary Bladder Neoplasms/metabolism , Animals , Antibiotics, Antineoplastic/therapeutic use , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation , Chloroquine/pharmacology , Cisplatin/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Resistance, Neoplasm , Female , Gene Expression , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Melanoma/metabolism , Melanoma/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/physiopathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Gemcitabine
7.
Front Aging Neurosci ; 13: 772795, 2021.
Article in English | MEDLINE | ID: mdl-35069175

ABSTRACT

Objective: The aim of this study was to investigate whether progranulin (PGRN) levels in cerebrospinal fluid (CSF) were associated with postoperative delirium (POD) in geriatric patients undergoing knee replacement. Method: A total of 600 Han Chinese patients aged 65-90 years and who underwent unilateral total knee arthroplasty were included in the Perioperative Neurocognitive Disorder And Biomarker LifestylE (PNDABLE) study from June 2020 to November 2020. All participants were assessed using the Confusion Assessment Method and the Memorial Delirium Assessment Scale on postoperative days 1-7 (or before discharge) by an anesthesiologist. CSF PGRN and CSF biomarkers of POD were measured by ELISA. We analyzed the risk and protective factors of POD using the multivariate logistic regression, and the associations between CSF PGRN and CSF biomarkers of POD using multiple linear regression. We also explored whether the influence of CSF PGRN on POD was mediated by POD core pathology in linear regression models. Results: Postoperative delirium incidence was 9.7% (53/545). There were significant differences in preoperative CSF PGRN between patients with POD and non-POD (NPOD). As for CSF biomarkers, CSF Aß40, T-tau, and P-tau were risk factors for POD, while CSF PGRN, Aß42, and Aß42/Aß40 were protective factors for POD, as shown by the multivariate logistic regression analysis. CSF PGRN was positively associated with CSF Aß42 and was negatively associated with CSF Aß40, T-tau, and P-tau in patients with POD. We found that the AUC was 0.795 (95% CI = 0.706, 0.867) for PGRN between POD and NPOD groups. We found the influence of CSF PGRN on POD was mediated by POD core pathology. The effect was considered partial mediation with the proportion of mediation varying from 44.92 to 62.07%. Conclusion: Cerebrospinal fluid PGRN may be a reasonably good prognostic factor for POD development. Overall, amyloid pathology and tau protein might partially mediate the influence of PGRN on POD. Clinical Trial Registration: www.clinicaltrials.gov, identifier ChiCTR2000033439.

8.
J Immunother Cancer ; 8(2)2020 08.
Article in English | MEDLINE | ID: mdl-32817394

ABSTRACT

The programmed death-ligand 1 (PD-L1)-dependent immune checkpoint attenuates host immunity and maintains self-tolerance. Imbalance between protective immunity and immunopathology due to altered PD-L1 signaling can lead to autoimmunity or tumor immunosuppression. The role of the PD-L1-dependent checkpoint in non-immune system is less reported. We previously found that white adipocytes highly express PD-L1. Here we show that adipocyte-specific PD-L1 knockout mice exhibit enhanced host anti-tumor immunity against mammary tumors and melanoma with low or no tumor PD-L1. However, adipocyte PD-L1 ablation in tumor-free mice also exacerbates diet-induced body weight gain, pro-inflammatory macrophage infiltration into adipose tissue, and insulin resistance. Low PD-L1 mRNA levels in human adipose tissue correlate with high body mass index and presence of type 2 diabetes. Therefore, our mouse genetic approach unequivocally demonstrates a cell-autonomous function of adipocyte PD-L1 in promoting tumor growth and inhibiting antitumor immunity. In addition, our work uncovers a previously unrecognized role of adipocyte PD-L1 in mitigating obesity-related inflammation and metabolic dysfunction.


Subject(s)
Adipocytes/drug effects , Inflammation/physiopathology , Obesity/physiopathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Female , Humans , Mice
9.
Int J Biol Sci ; 16(9): 1526-1535, 2020.
Article in English | MEDLINE | ID: mdl-32226299

ABSTRACT

Immune checkpoint blockade-based immunotherapy has become standard of care for multiple cancer types. However, the overall response rates among various cancer types still remain unsatisfactory. There is a pressing clinical need to identify combination therapies to improve efficacy of anticancer immunotherapy. We previously showed that pharmacologic inhibition of PPARγ by GW9662 boosts αPD-L1 and αPD-1 antibody efficacy in treating murine mammary tumors. In addition, we defined sexually dimorphic αPD-L1 efficacy in B16 melanoma. Here, we show a sexually dimorphic response to the combination of GW9662 and αPD-L1 immunotherapy in B16 melanoma. Combination effects were observed in female, but not male hosts. Neither female oöphorectomy impairs, nor does male castration rescue the combination effects, suggesting a sex hormone-independent response to this combination therapy. In diet-induced obese females, melanoma growth remained responsive to the combination treatment, albeit less robustly than lean females. These findings are informative for future design and application of immunotherapy-related combination therapy for treating human melanoma patients by taking gender and obesity status into consideration.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/therapeutic use , Melanoma, Experimental/therapy , PPAR gamma/antagonists & inhibitors , Sex Characteristics , Anilides/therapeutic use , Animals , Diet, High-Fat/adverse effects , Female , Immunotherapy , Male , Melanoma, Experimental/complications , Mice, Inbred C57BL , Obesity/complications , Obesity/etiology
10.
Oncoimmunology ; 7(11): e1500107, 2018.
Article in English | MEDLINE | ID: mdl-30393583

ABSTRACT

Programmed death-ligand 1 (PD-L1) and its receptor programmed cell death protein 1 (PD-1) modulate antitumor immunity and are major targets of checkpoint blockade immunotherapy. However, clinical trials of anti-PD-L1 and anti-PD-1 antibodies in breast cancer demonstrate only modest efficacy. Furthermore, specific PD-L1 contributions in various tissue and cell compartments to antitumor immunity remain incompletely elucidated. Here we show that PD-L1 expression is markedly elevated in mature adipocytes versus preadipocytes. Adipocyte PD-L1 prevents anti-PD-L1 antibody from activating important antitumor functions of CD8+ T cells in vitro. Adipocyte PD-L1 ablation obliterates, whereas forced preadipocyte PD-L1 expression confers, these inhibitory effects. Pharmacologic inhibition of adipogenesis selectively reduces PD-L1 expression in mouse adipose tissue and enhances the antitumor efficacy of anti-PD-L1 or anti-PD-1 antibodies in syngeneic mammary tumor models. Our findings provide a previously unappreciated approach to bolster anticancer immunotherapy efficacy and suggest a mechanism for the role of adipose tissue in breast cancer progression.

11.
J Chromatogr A ; 1564: 76-84, 2018 Aug 24.
Article in English | MEDLINE | ID: mdl-29935814

ABSTRACT

Plasma proteome profiling by LC-MS based proteomics has drawn great attention recently for biomarker discovery from blood liquid biopsy. Due to standard multi-step sample preparation could potentially cause plasma protein degradation and analysis variation, integrated proteomics sample preparation technologies became promising solution towards this end. Here, we developed a fully integrated proteomics sample preparation technology for both fast and deep plasma proteome profiling under its native pH. All the sample preparation steps, including protein digestion and two-dimensional fractionation by both mixed-mode ion exchange and high-pH reversed phase mechanism were integrated into one spintip device for the first time. The mixed-mode ion exchange beads design achieved the sample loading at neutral pH and protein digestion within 30 min. Potential sample loss and protein degradation by pH changing could be voided. 1 µL of plasma sample with depletion of high abundant proteins was processed by the developed technology with 12 equally distributed fractions and analyzed with 12 h of LC-MS gradient time, resulting in the identification of 862 proteins. The combination of the Mixed-mode-SISPROT and data-independent MS method achieved fast plasma proteome profiling in 2 h with high identification overlap and quantification precision for a proof-of-concept study of plasma samples from 5 healthy donors. We expect that the Mixed-mode-SISPROT become a generally applicable sample preparation technology for clinical oriented plasma proteome profiling.


Subject(s)
Blood Proteins/analysis , Ion Exchange , Proteome/analysis , Proteomics/methods , Chemical Fractionation , Chromatography, Liquid , Gene Expression Profiling , Humans , Proteolysis , Tandem Mass Spectrometry
12.
Anal Chem ; 90(9): 5879-5886, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29641186

ABSTRACT

Increasing attention has been focused on cell type proteome profiling for understanding the heterogeneous multicellular microenvironment in tissue samples. However, current cell type proteome profiling methods need large amounts of starting materials which preclude their application to clinical tumor specimens with limited access. Here, by seamlessly combining laser capture microdissection and integrated proteomics sample preparation technology SISPROT, specific cell types in tumor samples could be precisely dissected with single cell resolution and processed for high-sensitivity proteome profiling. Sample loss and contamination due to the multiple transfer steps are significantly reduced by the full integration and noncontact design. H&E staining dyes which are necessary for cell type investigation could be selectively removed by the unique two-stage design of the spintip device. This easy-to-use proteome profiling technology achieved high sensitivity with the identification of more than 500 proteins from only 0.1 mm2 and 10 µm thickness colon cancer tissue section. The first cell type proteome profiling of four cell types from one colon tumor and surrounding normal tissue, including cancer cells, enterocytes, lymphocytes, and smooth muscle cells, was obtained. 5271, 4691, 4876, and 2140 protein groups were identified, respectively, from tissue section of only 5 mm2 and 10 µm thickness. Furthermore, spatially resolved proteome distribution profiles of enterocytes, lymphocytes, and smooth muscle cells on the same tissue slices and across four consecutive sections with micrometer distance were successfully achieved. This fully integrated proteomics technology, termed LCM-SISPROT, is therefore promising for spatial-resolution cell type proteome profiling of tumor microenvironment with a minute amount of clinical starting materials.


Subject(s)
Colonic Neoplasms/chemistry , Proteome/analysis , Proteomics , Colonic Neoplasms/pathology , Humans
13.
J Biol Chem ; 293(8): 2841-2849, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29298894

ABSTRACT

Discoidin domain receptor 1 (DDR1) is a collagen receptor that mediates cell communication with the extracellular matrix (ECM). Aberrant expression and activity of DDR1 in tumor cells are known to promote tumor growth. Although elevated DDR1 levels in the stroma of breast tumors are associated with poor patient outcome, a causal role for tumor-extrinsic DDR1 in cancer promotion remains unclear. Here we report that murine mammary tumor cells transplanted to syngeneic recipient mice in which Ddr1 has been knocked out (KO) grow less robustly than in WT mice. We also found that the tumor-associated stroma in Ddr1-KO mice exhibits reduced collagen deposition compared with the WT controls, supporting a role for stromal DDR1 in ECM remodeling of the tumor microenvironment. Furthermore, the stromal-vascular fraction (SVF) of Ddr1 knockout adipose tissue, which contains committed adipose stem/progenitor cells and preadipocytes, was impaired in its ability to stimulate tumor cell migration and invasion. Cytokine array-based screening identified interleukin 6 (IL-6) as a cytokine secreted by the SVF in a DDR1-dependent manner. SVF-produced IL-6 is important for SVF-stimulated tumor cell invasion in vitro, and, using antibody-based neutralization, we show that tumor promotion by IL-6 in vivo requires DDR1. In conclusion, our work demonstrates a previously unrecognized function of DDR1 in promoting tumor growth.


Subject(s)
Adipose Tissue/metabolism , Breast Neoplasms/metabolism , Discoidin Domain Receptor 1/metabolism , Interleukin-6/metabolism , Stromal Cells/metabolism , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/pathology , Animals , Antibodies, Neutralizing/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/metabolism , Discoidin Domain Receptor 1/genetics , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/pathology , Transplantation, Isogeneic , Tumor Burden/drug effects , Tumor Cells, Cultured , Tumor Microenvironment/drug effects
14.
Nat Commun ; 7: 12696, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27576939

ABSTRACT

Although food availability is a potent synchronizer of the peripheral circadian clock in mammals, the underlying mechanisms are unclear. Here, we show that hepatic Bmal1, a core transcription activator of the molecular clock, is post-transcriptionally regulated by signals from insulin, an important hormone that is temporally controlled by feeding. Insulin promotes postprandial Akt-mediated Ser42-phosphorylation of Bmal1 to induce its dissociation from DNA, interaction with 14-3-3 protein and subsequently nuclear exclusion, which results in the suppression of Bmal1 transcriptional activity. Inverted feeding cycles not only shift the phase of daily insulin oscillation, but also elevate the amplitude due to food overconsumption. This enhanced and reversed insulin signalling initiates the reset of clock gene rhythms by altering Bmal1 nuclear accumulation in mouse liver. These results reveal the molecular mechanism of insulin signalling in regulating peripheral circadian rhythms.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Clocks/physiology , Feeding Behavior/physiology , Insulin/metabolism , ARNTL Transcription Factors/genetics , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , Serine/metabolism , Signal Transduction/physiology
15.
Cell Rep ; 16(4): 1096-1105, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27425624

ABSTRACT

Type I interferons (IFNs) play both beneficial and harmful roles in antiviral responses. Precise regulation of host type I IFNs is thus needed to prevent immune dysregulation. Here, we find that the DNA demethylase TET3 is a negative regulator of IFN-ß in response to poly(I:C) stimulation or viral infection. Deletion of TET3 enhances antiviral responses, with elevated expression of IFN-ß and IFN-stimulated genes. The catalytic domain of TET3 was critical for the suppression of IFN-ß production, but TET3 enzymatic activity was dispensable. Instead, the catalytic domain of TET3 interacts with HDAC1 and SIN3A, thus enhancing their binding to the Ifnb1 promoter. Our study demonstrates that TET3 negatively regulates type I IFN production independent of DNA demethylation. This not only sheds light on TET3 as a signaling protein in immune cells for gene regulation but also will help to develop strategies to prevent type I IFN-related disease.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Interferon Type I/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Line , Demethylation , Dioxygenases , Gene Expression Regulation/genetics , HEK293 Cells , Histone Deacetylase 1/metabolism , Humans , Interferon-beta/metabolism , Interferon-gamma/metabolism , Mice , Poly I-C/metabolism , Promoter Regions, Genetic/genetics , Repressor Proteins/metabolism , Signal Transduction/physiology , Sin3 Histone Deacetylase and Corepressor Complex , Virus Diseases/metabolism
16.
Autophagy ; 12(9): 1447-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27304906

ABSTRACT

Macroautophagy/autophagy is a conserved catabolic process that recycles cytoplasmic material during low energy conditions. BECN1/Beclin1 (Beclin 1, autophagy related) is an essential protein for function of the class 3 phosphatidylinositol 3-kinase (PtdIns3K) complexes that play a key role in autophagy nucleation and elongation. Here, we show that AMP-activated protein kinase (AMPK) regulates autophagy by phosphorylating BECN1 at Thr388. Phosphorylation of BECN1 is required for autophagy upon glucose withdrawal. BECN1(T388A), a phosphorylation defective mutant, suppresses autophagy through decreasing the interaction between PIK3C3 (phosphatidylinositol 3-kinase catalytic subunit type 3) and ATG14 (autophagy-related 14). The BECN1(T388A) mutant has a higher affinity for BCL2 than its wild-type counterpart; the mutant is more prone to dimer formation. Conversely, a BECN1 phosphorylation mimic mutant, T388D, has stronger binding to PIK3C3 and ATG14, and promotes higher autophagy activity than the wild-type control. These findings uncover a novel mechanism of autophagy regulation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Beclin-1/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Catalysis , Class III Phosphatidylinositol 3-Kinases/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Glucose/chemistry , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Membrane Proteins/metabolism , Mice , Mutation , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Protein Multimerization , Threonine/chemistry
17.
J Biol Chem ; 290(4): 2189-97, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25480789

ABSTRACT

Energy metabolism follows a diurnal pattern responding to the cycles of light and food exposures. Although food availability is a potent synchronizer of peripheral circadian clock in mammals, the underlying mechanism remains elusive. Here, we found that the temporal signals of fasting and refeeding hormones regulate the transcription of Bmal1, a key transcription activator of molecular clock, in the liver. During fasting, glucagon, a major fasting hormone, activates CREB/CRTC2 transcriptional complex that is recruited to Bmal1 promoter to induce its expression. Furthermore, we showed that CRTC2 is required for basal transcriptional regulation of Bmal1 by experiments using either adenovirus-mediated CRTC2 RNAi knockdown or primary Crtc2 null hepatocytes. On the other hand, insulin suppresses fasting-induced Bmal1 expression by inhibiting CRTC2 activity after refeeding. Taken together, our results indicate CRTC2 as a key component of the circadian oscillator that integrates the mammalian clock and energy metabolism.


Subject(s)
ARNTL Transcription Factors/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Glucagon/metabolism , Liver/metabolism , Transcription Factors/physiology , Animals , Cell Line , Circadian Rhythm , Glucocorticoids/metabolism , Gluconeogenesis , HEK293 Cells , Hepatocytes/metabolism , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Oscillometry , Promoter Regions, Genetic , RNA Interference , Signal Transduction/genetics , Transcription, Genetic
18.
Nat Med ; 16(10): 1152-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20852621

ABSTRACT

During fasting, mammals maintain normal glucose homeostasis by stimulating hepatic gluconeogenesis. Elevations in circulating glucagon and epinephrine, two hormones that activate hepatic gluconeogenesis, trigger the cAMP-mediated phosphorylation of cAMP response element-binding protein (Creb) and dephosphorylation of the Creb-regulated transcription coactivator-2 (Crtc2)--two key transcriptional regulators of this process. Although the underlying mechanism is unclear, hepatic gluconeogenesis is also regulated by the circadian clock, which coordinates glucose metabolism with changes in the external environment. Circadian control of gene expression is achieved by two transcriptional activators, Clock and Bmal1, which stimulate cryptochrome (Cry1 and Cry2) and Period (Per1, Per2 and Per3) repressors that feed back on Clock-Bmal1 activity. Here we show that Creb activity during fasting is modulated by Cry1 and Cry2, which are rhythmically expressed in the liver. Cry1 expression was elevated during the night-day transition, when it reduced fasting gluconeogenic gene expression by blocking glucagon-mediated increases in intracellular cAMP concentrations and in the protein kinase A-mediated phosphorylation of Creb. In biochemical reconstitution studies, we found that Cry1 inhibited accumulation of cAMP in response to G protein-coupled receptor (GPCR) activation but not to forskolin, a direct activator of adenyl cyclase. Cry proteins seemed to modulate GPCR activity directly through interaction with G(s)α. As hepatic overexpression of Cry1 lowered blood glucose concentrations and improved insulin sensitivity in insulin-resistant db/db mice, our results suggest that compounds that enhance cryptochrome activity may provide therapeutic benefit to individuals with type 2 diabetes.


Subject(s)
Circadian Rhythm/physiology , Cryptochromes/physiology , Cyclic AMP/physiology , Gluconeogenesis , Liver/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/physiology , Male , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/physiology
SELECTION OF CITATIONS
SEARCH DETAIL