Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Sci Transl Med ; 16(741): eadj5705, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38569015

ABSTRACT

Cancer-associated fibroblasts (CAFs) are abundant stromal cells in the tumor microenvironment that promote cancer progression and relapse. However, the heterogeneity and regulatory roles of CAFs underlying chemoresistance remain largely unclear. Here, we performed a single-cell analysis using high-dimensional flow cytometry analysis and identified a distinct senescence-like tetraspanin-8 (TSPAN8)+ myofibroblastic CAF (myCAF) subset, which is correlated with therapeutic resistance and poor survival in multiple cohorts of patients with breast cancer (BC). TSPAN8+ myCAFs potentiate the stemness of the surrounding BC cells through secretion of senescence-associated secretory phenotype (SASP)-related factors IL-6 and IL-8 to counteract chemotherapy. NAD-dependent protein deacetylase sirtuin 6 (SIRT6) reduction was responsible for the senescence-like phenotype and tumor-promoting role of TSPAN8+ myCAFs. Mechanistically, TSPAN8 promoted the phosphorylation of ubiquitin E3 ligase retinoblastoma binding protein 6 (RBBP6) at Ser772 by recruiting MAPK11, thereby inducing SIRT6 protein destruction. In turn, SIRT6 down-regulation up-regulated GLS1 and PYCR1, which caused TSPAN8+ myCAFs to secrete aspartate and proline, and therefore proved a nutritional niche to support BC outgrowth. By demonstrating that TSPAN8+SIRT6low myCAFs were tightly associated with unfavorable disease outcomes, we proposed that the combined regimen of anti-TSPAN8 antibody and SIRT6 activator MDL-800 is a promising approach to overcome chemoresistance. These findings highlight that senescence contributes to CAF heterogeneity and chemoresistance and suggest that targeting TSPAN8+ myCAFs is a promising approach to circumvent chemoresistance.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Sirtuins , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/pathology , Fibroblasts/pathology , Tumor Microenvironment , DNA-Binding Proteins , Ubiquitin-Protein Ligases , Tetraspanins/genetics , Tetraspanins/metabolism
2.
Cell Insight ; 2(6): 100127, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37961047

ABSTRACT

Hypopharyngeal squamous cell carcinoma (HSCC) is a highly aggressive malignancy that constitutes approximately 95% of all hypopharyngeal carcinomas, and it carries a poor prognosis. The primary factor influencing the efficacy of anti-cancer drugs for this type of carcinoma is chemoresistance. Carnitine palmitoyltransferase 1A (CPT1A) has been associated with tumor progression in various cancers, including breast, gastric, lung, and prostate cancer. The inhibition or depletion of CPT1A can lead to apoptosis, curbing cancer cell proliferation and chemoresistance. However, the role of CPT1A in HSCC is not yet fully understood. In this study, we discovered that CPT1A is highly expressed in HSCC and is associated with an advanced T-stage and a poor 5-year survival rate among patients. Furthermore, the overexpression of CPT1A contributes to HSCC chemoresistance. Mechanistically, CPT1A can interact with the autophagy-related protein ATG16L1 and stimulate the succinylation of ATG16L1, which in turn drives autophagosome formation and autophagy. We also found that treatment with 3-methyladenine (3-MA) can reduce cisplatin resistance in HSCC cells that overexpress CPT1A. Our findings also showed that a CPT1A inhibitor significantly enhances cisplatin sensitivity both in vitro and in vivo. This study is the first to suggest that CPT1A has a regulatory role in autophagy and is linked to poor prognosis in HSCC patients. It presents novel insights into the roles of CPT1A in tumorigenesis and proposes that CPT1A could be a potential therapeutic target for HSCC treatment.

3.
Nat Commun ; 14(1): 6523, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37863889

ABSTRACT

Cuproptosis, caused by excessively high copper concentrations, is urgently exploited as a potential cancer therapeutic. However, the mechanisms underlying the initiation, propagation, and ultimate execution of cuproptosis in tumors remain unknown. Here, we show that copper content is significantly elevated in gastric cancer (GC), especially in malignant tumors. Screening reveals that METTL16, an atypical methyltransferase, is a critical mediator of cuproptosis through the m6A modification on FDX1 mRNA. Furthermore, copper stress promotes METTL16 lactylation at site K229 followed by cuproptosis. The process of METTL16 lactylation is inhibited by SIRT2. Elevated METTL16 lactylation significantly improves the therapeutic efficacy of the copper ionophore- elesclomol. Combining elesclomol with AGK2, a SIRT2-specific inhibitor, induce cuproptosis in gastric tumors in vitro and in vivo. These results reveal the significance of non-histone protein METTL16 lactylation on cuproptosis in tumors. Given the high copper and lactate concentrations in GC, cuproptosis induction becomes a promising therapeutic strategy for GC.


Subject(s)
Apoptosis , Stomach Neoplasms , Humans , Copper , Lactic Acid , Methyltransferases/genetics , RNA, Messenger/genetics , Sirtuin 2 , Stomach Neoplasms/genetics
4.
Free Radic Biol Med ; 195: 132-144, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36584797

ABSTRACT

Tubulointerstitial fibrosis (TIF) is essential during the development of end-stage kidney disease (ESKD) and is associated with the impairment of fatty acid oxidation (FAO). Kruppel-like factor 14 (KLF14) is an important gene in lipid metabolism, but its role in TIF remains unknown. TGF-ß-stimulated HK-2 cells and mouse unilateral ureteral obstruction (UUO) were used as renal fibrosis models. The role of KLF14 in the process of renal fibrosis was verified by gene knockout mice, genetic or pharmacological interference in animal model and cell model respectively. In the current study, we found that KLF14 expression increased after activation of the TGF-ß signaling pathway during TIF. In KLF14-/- mice, more severe fibrosis was observed after unilateral ureteral obstruction (UUO) was induced. In human HK2 cells, knockdown of KLF14 led to more severe fibrosis induced by TGF-ß1, while overexpression of KLF14 partially attenuated this process. Specifically, KLF14 deficiency decreased mitochondrial FAO activity, resulting in lipid accumulation. Thus, the energy supply to the cells was insufficient, finally resulting in TIF. We further proved that KLF14 could target peroxisome proliferator activated receptor alpha (PPARα) as a transcriptional activator. This study identified the upregulation of KLF14 expression in response to kidney stress during the process of fibrosis. Upon TIF, the activated TGF-ß signaling pathway can enhance KLF14 expression, while the upregulation of KLF14 expression can decrease the degree of TIF by improving FAO activity in tubular epithelial cells and recovering the energy supply mediated by PPARα.


Subject(s)
Kidney Diseases , Kruppel-Like Transcription Factors , PPAR alpha , Ureteral Obstruction , Animals , Humans , Mice , Fatty Acids/metabolism , Fibrosis , Kidney/metabolism , Kidney Diseases/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Lipid Metabolism/genetics , PPAR alpha/genetics , PPAR alpha/metabolism , Transforming Growth Factor beta1/genetics , Up-Regulation , Ureteral Obstruction/genetics , Mice, Knockout
5.
Microsc Res Tech ; 85(11): 3589-3599, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35869784

ABSTRACT

The pollution of lead (Pb (II)) to water resources is becoming more and more serious. It is always a difficult problem to find efficient and low-cost adsorbents. Chicken manure (CM) and Chinese medicine residue (CMR) were modified with potassium dihydrogen phosphate (KH2 PO4 ) and pyrolyzed to obtain a modified material (PBC) for the treatment of Pb(II) in an aqueous solution. A variety of characterization analysis results could prove that KH2 PO4 was successfully introduced to PBC. By adjusting the initial pH, the zeta potential of PBC varies from -3.2 to -43.1 mV, it could be seen that PBC had excellent applicability in the broad range pH value (1.0-6.0). Experimental and model results showed that R2 of the pseudo-first order kinetic model and the pseudo-second order kinetic model are greater than 0.99, indicating that that physical and chemical adsorption played a significant role in Pb(II) removal by PBC. An adsorption isotherm analysis showed that the adsorption capacity n in this study is greater than 1, confirming that PBC has a good adsorption effect on Pb (II). R2 of the Langmuir model of PBC is 0.981, and its maximum adsorption capacity of Pb(II) could reach 599.4 mg/g. Environmentally friendly PBC could be used as an effective adsorbent to remove Pb(II) from aqueous systems. RESEARCH HIGHLIGHTS: Chicken manure and Chinese medicine residue were converted into biochar to improve utilization. The modified biochar exhibited extraordinary Pb(II) adsorption capacity. Adsorption mechanisms: Surface complexation, ions exchange, coprecipitation, and so on. Remained great Pb(II) removal efficiency at different pH and Pb(II) concentration.


Subject(s)
Pyrolysis , Water Pollutants, Chemical , Adsorption , Animals , Charcoal , Chickens , Hydrogen-Ion Concentration , Ions , Kinetics , Lead , Manure/analysis , Medicine, Chinese Traditional , Phosphorus , Water/chemistry , Water Pollutants, Chemical/analysis
6.
Cell Res ; 32(4): 359-374, 2022 04.
Article in English | MEDLINE | ID: mdl-35197608

ABSTRACT

TSPAN family of proteins are generally considered to assemble as multimeric complexes on the plasma membrane. Our previous work uncovered that TSPAN8 can translocate into the nucleus as a membrane-free form, a process that requires TSPAN8 palmitoylation and association with cholesterol to promote its extraction from the plasma membrane and subsequent binding with 14-3-3θ and importin-ß. However, what upstream signal(s) regulate(s) the nuclear translocation of TSPAN8, the potential function of TSPAN8 in the nucleus, and the underlying molecular mechanisms all remain unclear. Here, we demonstrate that, epidermal growth factor receptor (EGFR) signaling induces TSPAN8 nuclear translocation by activating the kinase AKT, which in turn directly phosphorylates TSPAN8 at Ser129, an event essential for its binding with 14-3-3θ and importin ß1. In the nucleus, phosphorylated TSPAN8 interacts with STAT3 to enhance its chromatin occupancy and therefore regulates transcription of downstream cancer-promoting genes, such as MYC, BCL2, MMP9, etc. The EGFR-AKT-TSPAN8-STAT3 axis was found to be hyperactivated in multiple human cancers, and associated with aggressive phenotype and dismal prognosis. We further developed a humanized monoclonal antibody hT8Ab4 that specifically recognizes the large extracellular loop of TSPAN8 (TSPAN8-LEL), thus being able to block the extraction of TSPAN8 from the plasma membrane and consequently its nuclear localization. Importantly, both in vitro and in vivo studies demonstrated an antitumor effect of hT8Ab4. Collectively, we discovered an unconventional function of TSPAN8 and dissected the underlying molecular mechanisms, which not only showcase a new layer of biological complexity of traditional membrane proteins, but also shed light on TSPAN8 as a novel therapeutic target for refractory cancers.


Subject(s)
ErbB Receptors , Neoplasms , STAT3 Transcription Factor , Tetraspanins , Cell Line, Tumor , ErbB Receptors/metabolism , Humans , Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Tetraspanins/genetics , Tetraspanins/metabolism
8.
Cell Mol Immunol ; 19(4): 504-515, 2022 04.
Article in English | MEDLINE | ID: mdl-34983946

ABSTRACT

Sepsis is a heterogeneous syndrome induced by a dysregulated host response to infection. Glycolysis plays a role in maintaining the immune function of macrophages, which is crucial for severely septic patients. However, how the pathways that link glycolysis and macrophages are regulated is still largely unknown. Here, we provide evidence to support the function of KLF14, a novel Krüppel-like transcription factor, in the regulation of glycolysis and the immune function of macrophages during sepsis. KLF14 deletion led to significantly increased mortality in lethal models of murine endotoxemia and sepsis. Mechanistically, KLF14 decreased glycolysis and the secretion of inflammatory cytokines by macrophages by inhibiting the transcription of HK2. In addition, we confirmed that the expression of KLF14 was upregulated in septic patients. Furthermore, pharmacological activation of KLF14 conferred protection against sepsis in mice. These findings uncover a key role of KLF14 in modulating the inflammatory signaling pathway and shed light on the development of KLF14-targeted therapeutics for sepsis.


Subject(s)
Sepsis , Transcription Factors , Animals , Glycolysis , Hexokinase , Humans , Immunity , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Transcription Factors/metabolism
9.
Environ Sci Pollut Res Int ; 29(3): 3853-3863, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34402012

ABSTRACT

Biochar (BC) obtained by the co-pyrolysis of municipal sewage sludge (MSS) and sunflower seed shells (SSS) was utilized to support nanoscale zero-valent iron particles (nZVI) for the synthesis of a composite material (nZVI-BC) for Cr(VI) removal from aqueous systems. A series of characterization methods confirmed successful immobilization of nZVI on the surface of biochar with no aggregation. Batch experiments showed that the initial pH, initial Cr(VI) concentration, and nZVI-BC dose all significantly affected the Cr(VI) removal using nZVI-BC. The kinetics for Cr(VI) removal via nZVI-BC could be better explained by the pseudo-second-order (PSO) adsorption model. Adsorption isotherms analysis demonstrated the superior Cr(VI) removal capability of nZVI-BC in comparison to bare nZVI and BC. nZVI-BC can be reused after the regeneration process by applying 0.1 M H2SO4 and 0.1 M NaBH4 solutions. The reaction mechanism for Cr(VI) removal might involve its chemical reduction on the nZVI-BC surface. Overall, environmentally friendly nZVI-BC was highly efficient in Cr(VI) removal from aqueous systems.


Subject(s)
Iron , Water Pollutants, Chemical , Adsorption , Charcoal , Chromium , Sewage , Water Pollutants, Chemical/analysis
10.
Hepatology ; 75(5): 1218-1234, 2022 05.
Article in English | MEDLINE | ID: mdl-34591986

ABSTRACT

BACKGROUND AND AIMS: NAFLD is considered as the hepatic manifestation of the metabolic syndrome, which includes insulin resistance, obesity and hyperlipidemia. NASH is a progressive stage of NAFLD with severe hepatic steatosis, hepatocyte death, inflammation, and fibrosis. Currently, no pharmacological interventions specifically tailored for NASH are approved. Ovarian tumor domain, ubiquitin aldehyde binding 1 (OTUB1), the founding member of deubiquitinases, regulates many metabolism-associated signaling pathways. However, the role of OTUB1 in NASH is unclarified. METHODS AND RESULTS: We demonstrated that mice with Otub1 deficiency exhibited aggravated high-fat diet-induced and high-fat high-cholesterol (HFHC) diet-induced hyperinsulinemia and liver steatosis. Notably, hepatocyte-specific overexpression of Otub1 markedly alleviated HFHC diet-induced hepatic steatosis, inflammatory responses, and liver fibrosis. Mechanistically, we identified apoptosis signal-regulating kinase 1 (ASK1) as a key candidate target of OTUB1 through RNA-sequencing analysis and immunoblot analysis. Through immunoprecipitation-mass spectrometry analysis, we further found that OTUB1 directly bound to tumor necrosis factor receptor-associated factor 6 (TRAF6) and suppressed its lysine 63-linked polyubiquitination, thus inhibiting the activation of ASK1 and its downstream pathway. CONCLUSIONS: OTUB1 is a key suppressor of NASH that inhibits polyubiquitinations of TRAF6 and attenuated TRAF6-mediated ASK1 activation. Targeting the OTUB1-TRAF6-ASK1 axis may be a promising therapeutic strategy for NASH.


Subject(s)
Cysteine Endopeptidases/metabolism , Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat , Disease Models, Animal , Liver , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Signal Transduction , TNF Receptor-Associated Factor 6
11.
Oncogene ; 40(30): 4884-4893, 2021 07.
Article in English | MEDLINE | ID: mdl-34163029

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is the deadliest cancer mainly owing to its proclivity to early metastasis and the lack of effective targeted therapeutic drugs. Hence, understanding the molecular mechanisms underlying early invasion and metastasis by PDAC is imperative for improving patient outcomes. The present study identified that upregulation of TSPAN8 expression in PDAC facilitates metastasis in vivo and in vitro. We found SOX9 as a key transcriptional regulator of TSPAN8 expression in response to EGF stimulation. SOX9 modulation was sufficient to positively regulate endogenous expression of TSPAN8, with concomitant in vitro phenotypic changes such as loss of cell-matrix adherence and increased invasion. Moreover, increased SOX9 and TSPAN8 levels were shown to correlate in human pancreatic cancer specimens and downregulated in vitro by EGFR tyrosine kinase inhibitors. High expression of SOX9 and TSPAN8 has been associated with tumor stage, poor prognosis and poor patient survival in PDAC. In conclusion, this study highlights the importance of the EGF-SOX9-TSPAN8 signaling cascade in the control of PDAC invasion and implies that TSPAN8 may be a promising novel therapeutic target for the treatment of PDAC.


Subject(s)
Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , SOX9 Transcription Factor/metabolism , Tetraspanins/genetics , Biomarkers, Tumor , Cell Movement/genetics , Disease Progression , Disease Susceptibility , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Metastasis , Neoplasm Staging , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Promoter Regions, Genetic , Protein Binding , Tetraspanins/metabolism
12.
Oncogene ; 40(23): 3974-3988, 2021 06.
Article in English | MEDLINE | ID: mdl-33990700

ABSTRACT

5-Fluorouracil (5-FU)-based chemotherapy is the first-line treatment for colorectal cancer (CRC) but is hampered by chemoresistance. Despite its impact on patient survival, the mechanism underlying chemoresistance against 5-FU remains poorly understood. Here, we identified serine hydroxymethyltransferase-2 (SHMT2) as a critical regulator of 5-FU chemoresistance in CRC. SHMT2 inhibits autophagy by binding cytosolic p53 instead of metabolism. SHMT2 prevents cytosolic p53 degradation by inhibiting the binding of p53 and HDM2. Under 5-FU treatment, SHMT2 depletion promotes autophagy and inhibits apoptosis. Autophagy inhibitors decrease low SHMT2-induced 5-FU resistance in vitro and in vivo. Finally, the lethality of 5-FU treatment to CRC cells was enhanced by treatment with the autophagy inhibitor chloroquine in patient-derived and CRC cell xenograft models. Taken together, our findings indicate that autophagy induced by low SHMT2 levels mediates 5-FU resistance in CRC. These results reveal the SHMT2-p53 interaction as a novel therapeutic target and provide a potential opportunity to reduce chemoresistance.


Subject(s)
Chloroquine/pharmacology , Colorectal Neoplasms/drug therapy , Fluorouracil/pharmacology , Glycine Hydroxymethyltransferase/metabolism , Animals , Antimalarials/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Female , Glycine Hydroxymethyltransferase/deficiency , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Survival Rate , Xenograft Model Antitumor Assays
13.
Sci Adv ; 7(9)2021 02.
Article in English | MEDLINE | ID: mdl-33627431

ABSTRACT

Improper distribution of chromosomes during mitosis can contribute to malignant transformation. Higher eukaryotes have evolved a mitotic catastrophe mechanism for eliminating mitosis-incompetent cells; however, the signaling cascade and its epigenetic regulation are poorly understood. Our analyses of human cancerous tissue revealed that the NAD-dependent deacetylase SIRT2 is up-regulated in early-stage carcinomas of various organs. Mass spectrometry analysis revealed that SIRT2 interacts with and deacetylates the structural maintenance of chromosomes protein 1 (SMC1A), which then promotes SMC1A phosphorylation to properly drive mitosis. We have further demonstrated that inhibition of SIRT2 activity or continuously increasing SMC1A-K579 acetylation causes abnormal chromosome segregation, which, in turn, induces mitotic catastrophe in cancer cells and enhances their vulnerability to chemotherapeutic agents. These findings suggest that regulation of the SIRT2-SMC1A axis through deacetylation-phosphorylation permits escape from mitotic catastrophe, thus allowing early precursor lesions to overcome oncogenic stress.


Subject(s)
Antimitotic Agents , Sirtuin 2 , Acetylation , Carcinogenesis/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Humans , Phosphorylation , Sirtuin 2/genetics , Sirtuin 2/metabolism
14.
Nat Commun ; 12(1): 20, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397932

ABSTRACT

Drug resistance and tumor recurrence are major challenges in cancer treatment. Cancer cells often display centrosome amplification. To maintain survival, cancer cells achieve bipolar division by clustering supernumerary centrosomes. Targeting centrosome clustering is therefore considered a promising therapeutic strategy. However, the regulatory mechanisms of centrosome clustering remain unclear. Here we report that KIFC1, a centrosome clustering regulator, is positively associated with tumor recurrence. Under DNA damaging treatments, the ATM and ATR kinases phosphorylate KIFC1 at Ser26 to selectively maintain the survival of cancer cells with amplified centrosomes via centrosome clustering, leading to drug resistance and tumor recurrence. Inhibition of KIFC1 phosphorylation represses centrosome clustering and tumor recurrence. This study identified KIFC1 as a prognostic tumor recurrence marker, and revealed that tumors can acquire therapeutic resistance and recurrence via triggering centrosome clustering under DNA damage stresses, suggesting that blocking KIFC1 phosphorylation may open a new vista for cancer therapy.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Centrosome/metabolism , Kinesins/metabolism , Neoplasm Recurrence, Local/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Chromosomal Instability , DNA Damage , Drug Resistance, Neoplasm , Humans , Kinesins/chemistry , Mice , Neoplasm Recurrence, Local/pathology , Phosphorylation , Phosphoserine/metabolism
15.
Aging (Albany NY) ; 12(8): 7380-7396, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32327611

ABSTRACT

Cardiac fibrosis is a primary phenotype of cardiac remodeling that contributes to cardiac dysfunction and heart failure. The expansion and activation of CD4+ T cells in the heart has been identified to facilitate pathological cardiac remodeling and dysfunction; however, the underlying mechanisms remained not well clarified. Herein, we found that exosomes derived from activated CD4+ T cells (CD4-activated Exos) evoked pro-fibrotic effects of cardiac fibroblasts, and their delivery into the heart aggravated cardiac fibrosis and dysfunction post-infarction. Mechanistically, miR-142-3p that was enriched in CD4-activated Exos recapitulated the pro-fibrotic effects of CD4-activated Exos in cardiac fibroblasts, and vice versa. Furthermore, miR-142-3p directly targeted and inhibited the expression of Adenomatous Polyposis Coli (APC), a negative WNT signaling pathway regulator, contributing to the activation of WNT signaling pathway and cardiac fibroblast activation. Thus, CD4-activated Exos promote post-ischemic cardiac fibrosis through exosomal miR-142-3p-WNT signaling cascade-mediated activation of myofibroblasts. Targeting miR-142-3p in CD4-activated Exos may hold promise for treating cardiac remodeling post-MI.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Myocardial Infarction/genetics , Myofibroblasts/metabolism , T-Lymphocytes/metabolism , Ventricular Remodeling , Animals , Disease Models, Animal , Exosomes , Fibroblasts/metabolism , Fibroblasts/pathology , MicroRNAs/biosynthesis , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , RNA/genetics , Wnt Signaling Pathway
16.
EMBO J ; 39(10): e103111, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32187724

ABSTRACT

The homeostatic link between oxidative stress and autophagy plays an important role in cellular responses to a wide variety of physiological and pathological conditions. However, the regulatory pathway and outcomes remain incompletely understood. Here, we show that reactive oxygen species (ROS) function as signaling molecules that regulate autophagy through ataxia-telangiectasia mutated (ATM) and cell cycle checkpoint kinase 2 (CHK2), a DNA damage response (DDR) pathway activated during metabolic and hypoxic stress. We report that CHK2 binds to and phosphorylates Beclin 1 at Ser90/Ser93, thereby impairing Beclin 1-Bcl-2 autophagy-regulatory complex formation in a ROS-dependent fashion. We further demonstrate that CHK2-mediated autophagy has an unexpected role in reducing ROS levels via the removal of damaged mitochondria, which is required for cell survival under stress conditions. Finally, CHK2-/- mice display aggravated infarct phenotypes and reduced Beclin 1 p-Ser90/Ser93 in a cerebral stroke model, suggesting an in vivo role of CHK2-induced autophagy in cell survival. Taken together, these results indicate that the ROS-ATM-CHK2-Beclin 1-autophagy axis serves as a physiological adaptation pathway that protects cells exposed to pathological conditions from stress-induced tissue damage.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Beclin-1/metabolism , Checkpoint Kinase 2/metabolism , Ischemic Stroke/metabolism , Reactive Oxygen Species/metabolism , Animals , Autophagy , Cell Line , Disease Models, Animal , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Mice , Oxidative Stress , Phosphorylation
17.
Curr Biol ; 27(15): 2271-2284.e5, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28756945

ABSTRACT

Most eukaryotic mRNAs are polyadenylated in the nucleus, and the poly(A)-tail is required for efficient mRNA export and translation. However, mechanisms governing mRNA transport remain unclear. Here, we report that the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase SIRT1 acts as an energy sensor and negatively regulates poly(A)RNA transport via deacetylating a poly(A)-binding protein, PABP1. Upon energy starvation, SIRT1 interacts with and deacetylates PABP1 and deactivates its poly(A)RNA binding, leading to nuclear accumulation of PABP1 and poly(A)RNA and thus facilitating eukaryotic cells to attenuate protein synthesis and energy consumption to adapt to energy stress. Moreover, AMPK-directed SIRT1 phosphorylation is required for energy starvation-induced PABP1-SIRT1 association, PABP1 deacetylation, and poly(A)RNA nuclear retention. In addition, the SIRT1-PABP1 association is not specific to energy starvation but represents a common stress response. These observations provide insights into dynamic modulation of eukaryotic mRNA transport and translation, suggesting that the poly(A)-tail also provides a basis for eukaryotes to effectively shut down mature mRNA transport and thereby tailor protein synthesis to maintain energy homeostasis under stress conditions.


Subject(s)
Poly(A)-Binding Protein I/genetics , RNA Transport , Sirtuin 1/genetics , Animals , HeLa Cells , Humans , Mice , Mice, Knockout , Poly(A)-Binding Protein I/metabolism , Sirtuin 1/metabolism
18.
Nat Commun ; 7: 12497, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27511885

ABSTRACT

Maintenance of energy homeostasis is essential for cell survival. Here, we report that the ATP- and ubiquitin-independent REGγ-proteasome system plays a role in maintaining energy homeostasis and cell survival during energy starvation via repressing rDNA transcription, a major intracellular energy-consuming process. Mechanistically, REGγ-proteasome limits cellular rDNA transcription and energy consumption by targeting the rDNA transcription activator SirT7 for ubiquitin-independent degradation under normal conditions. Moreover, energy starvation induces an AMPK-directed SirT7 phosphorylation and subsequent REGγ-dependent SirT7 subcellular redistribution and degradation, thereby further reducing rDNA transcription to save energy to overcome cell death. Energy starvation is a promising strategy for cancer therapy. Our report also shows that REGγ knockdown markedly improves the anti-tumour activity of energy metabolism inhibitors in mice. Our results underscore a control mechanism for an ubiquitin-independent process in maintaining energy homeostasis and cell viability under starvation conditions, suggesting that REGγ-proteasome inhibition has a potential to provide tumour-starving benefits.


Subject(s)
Autoantigens/metabolism , Homeostasis , Neoplasms/therapy , Proteasome Endopeptidase Complex/metabolism , Animals , Cell Survival , Cytoplasm/metabolism , DNA, Ribosomal/metabolism , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Phosphorylation , Ubiquitin/metabolism
19.
Nat Commun ; 6: 8450, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26439168

ABSTRACT

Centrosome amplification is frequent in cancer, but the underlying mechanisms remain unclear. Here we report that disruption of the Kruppel-like factor 14 (KLF14) gene in mice causes centrosome amplification, aneuploidy and spontaneous tumorigenesis. Molecularly, KLF14 functions as a transcriptional repressor of Plk4, a polo-like kinase whose overexpression induces centrosome overduplication. Transient knockdown of KLF14 is sufficient to induce Plk4-directed centrosome amplification. Clinically, KLF14 transcription is significantly downregulated, whereas Plk4 transcription is upregulated in multiple types of cancers, and there exists an inverse correlation between KLF14 and Plk4 protein expression in human breast and colon cancers. Moreover, KLF14 depletion promotes AOM/DSS-induced colon tumorigenesis. Our findings reveal that KLF14 reduction serves as a mechanism leading to centrosome amplification and tumorigenesis. On the other hand, forced expression of KLF14 leads to mitotic catastrophe. Collectively, our findings identify KLF14 as a tumour suppressor and highlight its potential as biomarker and therapeutic target for cancer.


Subject(s)
Carcinogenesis/genetics , Centrosome/metabolism , Kruppel-Like Transcription Factors/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Sp Transcription Factors/genetics , Aneuploidy , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Gene Knockdown Techniques , HCT116 Cells , HeLa Cells , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Mitosis/genetics , Protein Serine-Threonine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction
20.
Biosens Bioelectron ; 56: 258-63, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24508817

ABSTRACT

Calmodulin (CaM) is a ubiquitous protein in eukaryotic cells, and it plays an important role in cancer progression. In this paper, a highly sensitive immunosensor adopting a dual-layered enzyme strategy was proposed for electrochemical detection of CaM. This immunosensor was constructed by introducing honeycomb-like mesoporous carbon (HMPC) as a sensor platform to sequentially immobilize antibody (Ab1), CaM and a multi-functionalized label. The label (HRP-PAupc-Ab1) was synthesized by covalently binding Ab1 and horseradish peroxidase (HRP) to poly(acrylic acid)-functionalized Au popcorn (PAupc) nanoparticles. A novel dual-layered enzyme strategy was employed by incubating HRP-secondary antibody (HRP-Ab2) onto the label surface and the enhanced biocatalyzed precipitation was therefore induced. This immunosensor exhibited satisfactory analytical performances for CaM detection with a linear response ranging from 5.0 pg mL(-1) to 100 ng mL(-1) and a detection limit of 1.5 pg mL(-1). The immunosensor has also been successfully applied to the CaM analysis in two cancer cells (HepG2 and MCF-7) with high sensitivity, which has shown great potency for cancer study.


Subject(s)
Antibodies, Immobilized/chemistry , Biosensing Techniques/methods , Calmodulin/analysis , Biomarkers, Tumor , Carbon/chemistry , Cell Line, Tumor , Gold/chemistry , Horseradish Peroxidase/chemistry , Humans , Immunoenzyme Techniques/methods , Limit of Detection , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Porosity
SELECTION OF CITATIONS
SEARCH DETAIL
...