Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Exp Hematol Oncol ; 13(1): 14, 2024 Feb 07.
Article En | MEDLINE | ID: mdl-38326887

Brexucabtagene autoleucel CAR-T therapy is highly efficacious in overcoming resistance to Bruton's tyrosine kinase inhibitors (BTKi) in mantle cell lymphoma. However, many patients relapse post CAR-T therapy with dismal outcomes. To dissect the underlying mechanisms of sequential resistance to BTKi and CAR-T therapy, we performed single-cell RNA sequencing analysis for 66 samples from 25 patients treated with BTKi and/or CAR-T therapy and conducted in-depth bioinformatics™ analysis. Our analysis revealed that MYC activity progressively increased with sequential resistance. HSP90AB1 (Heat shock protein 90 alpha family class B member 1), a MYC target, was identified as early driver of CAR-T resistance. CDK9 (Cyclin-dependent kinase 9), another MYC target, was significantly upregulated in Dual-R samples. Both HSP90AB1 and CDK9 expression were correlated with MYC activity levels. Pharmaceutical co-targeting of HSP90 and CDK9 synergistically diminished MYC activity, leading to potent anti-MCL activity. Collectively, our study revealed that HSP90-MYC-CDK9 network is the primary driving force of therapeutic resistance.

2.
Nanoscale ; 15(47): 19322-19329, 2023 Dec 07.
Article En | MEDLINE | ID: mdl-37999717

Fe-based LDHs have been proven to be an excellent class of catalysts for the oxygen evolution reaction (OER). To achieve industrial applications of water splitting, it is critical to develop a cost-effective and simple strategy to achieve large-area catalytic electrodes. Herein, we present a moderate in situ method for growing Fe-based layered double hydroxide nanosheets on a Ni foam (LDH@NF) substrate at room temperature. Through systematic experimental design characterization, it is found that this in situ growth process is mainly driven by moderate oxidation of Fe2+ in an O2-dissolved solution, the consequent local alkaline environment, and abundant TM2+ ions (Ni2+, Co2+, Ni2+/Co2+). Compared with other in situ methods, this method is not accompanied by violent redox reactions and is favorable for the uniform growth of LDHs, and the composition of the catalyst can be easily regulated. Specifically, the optimized NiFe-LDH@NF catalyst demonstrates excellent catalytic performance in the alkaline water oxidation reaction with a low overpotential of 206/239 mV at a current density of 10/100 mA cm-2, respectively.

3.
Molecules ; 28(5)2023 Mar 02.
Article En | MEDLINE | ID: mdl-36903566

A batch of Fe-modified biochars MS (for soybean straw), MR (for rape straw), and MP (for peanut shell) were prepared by impregnating biochars pyrolyzed from three different raw biomass materials, i.e., peanut shell, soybean straw, and rape straw, with FeCl3 solution in different Fe/C impregnation ratios (0, 0.112, 0.224, 0.448, 0.560, 0.672, and 0.896) in this research. Their characteristics (pH, porosities, surface morphologies, crystal structures, and interfacial chemical behaviors) and phosphate adsorption capacities and mechanisms were evaluated. The optimization of their phosphate removal efficiency (Y%) was analyzed using the response surface method. Our results indicated that MR, MP, and MS showed their best phosphate adsorption capacity at Fe/C ratios of 0.672, 0.672, and 0.560, respectively. Rapid phosphate removal was observed within the first few minutes and the equilibrium was attained by 12 h in all treatment. The optimal conditions for phosphorus removal were pH = 7.0, initial phosphate concentration = 132.64 mg L-1, and ambient temperature = 25 °C, where the Y% values were 97.76, 90.23, and 86.23% of MS, MP, and MR, respectively. Among the three biochars, the maximum phosphate removal efficiency determined was 97.80%. The phosphate adsorption process of three modified biochars followed a pseudo-second-order adsorption kinetic model, indicating monolayer adsorption based on electrostatic adsorption or ion exchange. Thus, this study clarified the mechanism of phosphate adsorption by three Fe-modified biochar composites, which present as low-cost soil conditioners for rapid and sustainable phosphate removal.


Phosphates , Water Pollutants, Chemical , Adsorption , Charcoal/chemistry , Phosphorus , Water Pollutants, Chemical/chemistry
4.
Blood Cancer J ; 13(1): 27, 2023 02 17.
Article En | MEDLINE | ID: mdl-36797243

Constant challenges for the treatment of mantle cell lymphoma (MCL) remain to be recurrent relapses and therapy resistance, especially in patients harboring somatic mutations in the tumor suppressors ATM and TP53, which are accumulated as therapy resistance emerges and the disease progresses, consistent with our OncoPrint results that ATM and TP53 alterations were most frequent in relapsed/refractory (R/R) MCL. We demonstrated that protein arginine methyltransferase-5 (PRMT5) was upregulated in R/R MCL, which predicted a poor prognosis. PRMT5 inhibitors displayed profound antitumor effects in the mouse models of MCL with mutated ATM and/or TP53, or refractory to CD19-targeted CAR T-cell therapy. Genetic knockout of PRMT5 robustly inhibited tumor growth in vivo. Co-targeting PRMT5, and ATR or CDK4 by using their inhibitors showed synergistic antitumor effects both in vitro and in vivo. Our results have provided a rational combination therapeutic strategy targeting multiple PRMT5-coordinated tumor-promoting processes for the treatment of R/R MCL with high mutation burdens.


Lymphoma, Mantle-Cell , Animals , Mice , Enzyme Inhibitors/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Mutation , Neoplasm Recurrence, Local , Tumor Suppressor Protein p53/metabolism
5.
J Clin Invest ; 133(3)2023 02 01.
Article En | MEDLINE | ID: mdl-36719376

Bruton's tyrosine kinase (BTK) is a proven target in mantle cell lymphoma (MCL), an aggressive subtype of non-Hodgkin lymphoma. However, resistance to BTK inhibitors is a major clinical challenge. We here report that MALT1 is one of the top overexpressed genes in ibrutinib-resistant MCL cells, while expression of CARD11, which is upstream of MALT1, is decreased. MALT1 genetic knockout or inhibition produced dramatic defects in MCL cell growth regardless of ibrutinib sensitivity. Conversely, CARD11-knockout cells showed antitumor effects only in ibrutinib-sensitive cells, suggesting that MALT1 overexpression could drive ibrutinib resistance via bypassing BTK/CARD11 signaling. Additionally, BTK knockdown and MALT1 knockout markedly impaired MCL tumor migration and dissemination, and MALT1 pharmacological inhibition decreased MCL cell viability, adhesion, and migration by suppressing NF-κB, PI3K/AKT/mTOR, and integrin signaling. Importantly, cotargeting MALT1 with safimaltib and BTK with pirtobrutinib induced potent anti-MCL activity in ibrutinib-resistant MCL cell lines and patient-derived xenografts. Therefore, we conclude that MALT1 overexpression associates with resistance to BTK inhibitors in MCL, targeting abnormal MALT1 activity could be a promising therapeutic strategy to overcome BTK inhibitor resistance, and cotargeting of MALT1 and BTK should improve MCL treatment efficacy and durability as well as patient outcomes.


Lymphoma, Mantle-Cell , Protein-Tyrosine Kinases , Humans , Adult , Agammaglobulinaemia Tyrosine Kinase/genetics , Protein-Tyrosine Kinases/metabolism , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Cell Line, Tumor , Phosphatidylinositol 3-Kinases , Drug Resistance, Neoplasm/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics
6.
Haematologica ; 108(6): 1616-1627, 2023 06 01.
Article En | MEDLINE | ID: mdl-36420799

Mantle cell lymphoma (MCL) is an incurable B-cell non-Hodgkin lymphoma characterized by frequent relapses. The development of resistance to ibrutinib therapy remains a major challenge in MCL. We previously showed that glutaminolysis is associated with resistance to ibrutinib. In this study, we confirmed that glutaminase (GLS), the first enzyme in glutaminolysis, is overexpressed in ibrutinib-resistant MCL cells, and that its expression correlates well with elevated glutamine dependency and glutaminolysis. Furthermore, we discovered that GLS expression correlates with MYC expression and the functioning of the glutamine transporter ASCT2. Depletion of glutamine or GLS significantly reduced cell growth, while GLS overexpression enhanced glutamine dependency and ibrutinib resistance. Consistent with this, GLS inhibition by its specific inhibitor telaglenastat suppressed MCL cell growth both in vitro and in vivo. Moreover, telaglenastat showed anti-MCL synergy when combined with ibrutinib or venetoclax in vitro, which was confirmed using an MCL patient-derived xenograft model. Our study provides the first evidence that targeting GLS with telaglenastat, alone or in combination with ibrutinib or venetoclax, is a promising strategy to overcome ibrutinib resistance in MCL.


Drug Resistance, Neoplasm , Lymphoma, Mantle-Cell , Humans , Adult , Cell Line, Tumor , Glutaminase/pharmacology , Lymphoma, Mantle-Cell/pathology , Glutamine , Neoplasm Recurrence, Local , Enzyme Inhibitors/pharmacology
7.
Mol Cancer ; 21(1): 185, 2022 09 26.
Article En | MEDLINE | ID: mdl-36163179

BACKGROUND: Chimeric antigen receptor (CAR) T-cell therapy using brexucabtagene autoleucel (BA) induces remission in many patients with mantle cell lymphoma (MCL), and BA is the only CAR T-cell therapy approved by the FDA for MCL. However, development of relapses to BA is recognized with poor patient outcomes. Multiple CAR T-cell therapies have been approved for other lymphomas and the resistance mechanisms have been investigated. However, the mechanisms underlying BA relapse in MCL have not been investigated and whether any previously reported resistance mechanisms apply to BA-relapsed patients with MCL is unknown. METHODS: To interrogate BA resistance mechanisms in MCL, we performed single-cell RNA sequencing on 39 longitudinally collected samples from 15 BA-treated patients, and multiplex cytokine profiling on 80 serial samples from 20 patients. RESULTS: We demonstrate that after BA relapse, the proportion of T cells, especially cytotoxic T cells (CTLs), decreased among non-tumor cells, while the proportion of myeloid cells correspondingly increased. TIGIT, LAG3, and CD96 were the predominant checkpoint molecules expressed on exhausted T cells and CTLs; only TIGIT was significantly increased after relapse. CTLs expanded during remission, and then contracted during relapse with upregulated TIGIT expression. Tumor cells also acquired TIGIT expression after relapse, leading to the enhanced interaction of tumor cell TIGIT with monocyte CD155/PVR. In myeloid cells, post-relapse HLA-II expression was reduced relative to pretreatment and during remission. Myeloid-derived suppressor cells (MDSCs) were enriched after relapse with elevated expression of activation markers, including CLU (clusterin) and VCAN (versican). Extracellular chemokines (CCL4, CXCL9, CXCL13), soluble checkpoint inhibitors (sPD-L1, sTIM3, s4-1BB), and soluble receptors (sIL-2R, sTNFRII) were decreased during remission but elevated after relapse. CONCLUSIONS: Our data demonstrate that multiple tumor-intrinsic and -extrinsic factors are associated with T-cell suppression and BA relapse. Among these, TIGIT appears to be the central player given its elevated expression after BA relapse in not only CTLs but also MCL cells. The acquisition of TIGIT expression on tumor cells is MCL-specific and has not been reported in other CAR T-treated diseases. Together, our data suggest that co-targeting TIGIT may prevent CAR T relapses and thus promote long-term progression-free survival in MCL patients.


Lymphoma, Mantle-Cell , Receptors, Chimeric Antigen , Adult , Antigens, CD , Clusterin , Cytokines/metabolism , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/therapy , Neoplasm Recurrence, Local , Receptors, Immunologic/genetics , T-Lymphocytes , Versicans
8.
J Hematol Oncol ; 15(1): 42, 2022 04 11.
Article En | MEDLINE | ID: mdl-35410313

Inevitable relapses remain as the major therapeutic challenge in patients with mantle cell lymphoma (MCL) despite FDA approval of multiple targeted therapies and immunotherapies. Fc gamma receptors (FcγRs) play important roles in regulating antibody-mediated immunity. FcγRIIB, the unique immune-checkpoint inhibitory member of the FcγR family, has been implicated in immune cell desensitization and tumor cell resistance to the anti-CD20 antibody rituximab and other antibody-mediated immunotherapies; however, little is known about its expression and its immune-modulatory function in patients with aggressive MCL, especially those with multi-resistance. In this study, we found that FcγRIIB was ubiquitously expressed in both MCL cell lines and primary patient samples. FcγRIIB expression is significantly higher in CAR T-relapsed patient samples (p < 0.0001) compared to ibrutinib/rituximab-naïve, sensitive or resistant samples. Rituximab-induced CD20 internalization in JeKo-1 cells was completely blocked by concurrent treatment with BI-1206, a recombinant human monoclonal antibody targeting FcγRIIB. Combinational therapies with rituximab-ibrutinib, rituximab-venetoclax and rituximab-CHOP also induced CD20 internalization which was again effectively blocked by BI-1206. BI-1206 significantly enhanced the in vivo anti-MCL efficacy of rituximab-ibrutinib (p = 0.05) and rituximab-venetoclax (p = 0.02), but not the rituximab-CHOP combination in JeKo-1 cell line-derived xenograft models. In patient-derived xenograft (PDX) models, BI-1206, as a single agent, showed high potency (p < 0.0001, compared to vehicle control) in one aggressive PDX model that is resistant to both ibrutinib and venetoclax but sensitive to the combination of rituximab and lenalidomide (the preclinical mimetic of R2 therapy). BI-1206 sensitized the efficacy of rituximab monotherapy in a PDX model with triple resistance to rituximab, ibrutinib and CAR T-therapies (p = 0.030). Moreover, BI-1206 significantly enhanced the efficacy of the rituximab-venetoclax combination (p < 0.05), which led to long-term tumor remission in 25% of mice. Altogether, these data support that targeting this new immune-checkpoint blockade enhances the therapeutic activity of rituximab-based regimens in aggressive MCL models with multi-resistance.


Antineoplastic Agents , Lymphoma, Mantle-Cell , Receptors, Chimeric Antigen , Adult , Animals , Antibodies, Monoclonal, Murine-Derived , Antigens, CD20 , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Lymphoma, Mantle-Cell/drug therapy , Mice , Neoplasm Recurrence, Local/drug therapy , Receptors, Chimeric Antigen/therapeutic use , Rituximab/pharmacology , Rituximab/therapeutic use
9.
Methods Mol Biol ; 2326: 301-313, 2021.
Article En | MEDLINE | ID: mdl-34097278

Despite the fact that pesticides help increase food production, widespread application of pesticides has resulted in negative impact in the environment and human health. Routine and comprehensive screening of pesticides in food and water is important for regulatory agencies to ensure that concentrations of toxic pesticides are below maximum allowable levels. Regardless of the pesticides that are not GC amenable, GC-MS still dominates the analysis of pesticides. The focus of the current chapter is a step-by-step method for GC-MS approaches in analysis of several classes of pesticides, including organochlorines, organophosphates, and triazines. GC-MS is superior or at least equivalent to LC-MS method and derivatization is not required prior to instrumental analysis.


Gas Chromatography-Mass Spectrometry/methods , Hydrocarbons, Chlorinated/analysis , Organophosphates/analysis , Pesticides/analysis , Triazines/analysis , Food Contamination/analysis , Humans , Pesticide Residues/analysis , Water/analysis , Water Pollutants, Chemical/analysis
10.
Mol Cell ; 76(3): 516-527.e7, 2019 11 07.
Article En | MEDLINE | ID: mdl-31492635

The PTEN tumor suppressor is frequently mutated or deleted in cancer and regulates glucose metabolism through the PI3K-AKT pathway. However, whether PTEN directly regulates glycolysis in tumor cells is unclear. We demonstrate here that PTEN directly interacts with phosphoglycerate kinase 1 (PGK1). PGK1 functions not only as a glycolytic enzyme but also as a protein kinase intermolecularly autophosphorylating itself at Y324 for activation. The protein phosphatase activity of PTEN dephosphorylates and inhibits autophosphorylated PGK1, thereby inhibiting glycolysis, ATP production, and brain tumor cell proliferation. In addition, knockin expression of a PGK1 Y324F mutant inhibits brain tumor formation. Analyses of human glioblastoma specimens reveals that PGK1 Y324 phosphorylation levels inversely correlate with PTEN expression status and are positively associated with poor prognosis in glioblastoma patients. This work highlights the instrumental role of PGK1 autophosphorylation in its activation and PTEN protein phosphatase activity in governing glycolysis and tumorigenesis.


Brain Neoplasms/enzymology , Glioblastoma/enzymology , Glucose/metabolism , Glycolysis , PTEN Phosphohydrolase/metabolism , Phosphoglycerate Kinase/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Glioblastoma/genetics , Glioblastoma/pathology , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , PTEN Phosphohydrolase/genetics , Phosphoglycerate Kinase/genetics , Phosphorylation , Prognosis , Signal Transduction , Time Factors , Tumor Burden , Tyrosine
11.
Cancer Discov ; 8(1): 94-107, 2018 01.
Article En | MEDLINE | ID: mdl-29074724

Tumors override energy stress to grow. However, how nucleotide synthesis is regulated under energy stress is unclear. We demonstrate here that glucose deprivation or hypoxia results in the AMPK-mediated phosphorylation of phosphoribosyl pyrophosphate synthetase 1 (PRPS1) S180 and PRPS2 S183, leading to conversion of PRPS hexamers to monomers and thereby inhibiting PRPS1/2 activity, nucleotide synthesis, and nicotinamide adenine dinucleotide (NAD) production. Knock-in of nonphosphorylatable PRPS1/2 mutants, which have uninhibited activity, in brain tumor cells under energy stress exhausts cellular ATP and NADPH and increases reactive oxygen species levels, thereby promoting cell apoptosis. The expression of those mutants inhibits brain tumor formation and enhances the inhibitory effect of the glycolysis inhibitor 2-deoxy-d-glucose on tumor growth. Our findings highlight the significance of recalibrating tumor cell metabolism by fine-tuning nucleotide and NAD synthesis in tumor growth.Significance: Our findings elucidate an instrumental function of AMPK in direct regulation of nucleic acid and NAD synthesis in tumor cells in response to energy stress. AMPK phosphorylates PRPS1/2, converts PRPS1/2 hexamers to monomers, and inhibits PRPS1/2 activity and subsequent nucleotide and NAD synthesis to maintain tumor cell growth and survival. Cancer Discov; 8(1); 94-107. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1.


AMP-Activated Protein Kinases/metabolism , Mass Spectrometry/methods , Nucleotides/metabolism , Phosphorylation/drug effects , Ribose-Phosphate Pyrophosphokinase/metabolism , Animals , Humans , Mice , Rabbits , Transfection
12.
Nat Commun ; 8(1): 949, 2017 10 16.
Article En | MEDLINE | ID: mdl-29038421

Phosphofructokinase 1 (PFK1) plays a critical role in glycolysis; however, its role and regulation in tumorigenesis are not well understood. Here, we demonstrate that PFK1 platelet isoform (PFKP) is the predominant PFK1 isoform in human glioblastoma cells and its expression correlates with total PFK activity. We show that PFKP is overexpressed in human glioblastoma specimens due to an increased stability, which is induced by AKT activation resulting from phosphatase and tensin homologue (PTEN) loss and EGFR-dependent PI3K activation. AKT binds to and phosphorylates PFKP at S386, and this phosphorylation inhibits the binding of TRIM21 E3 ligase to PFKP and the subsequent TRIM21-mediated polyubiquitylation and degradation of PFKP. PFKP S386 phosphorylation increases PFKP expression and promotes aerobic glycolysis, cell proliferation, and brain tumor growth. In addition, S386 phosphorylation in human glioblastoma specimens positively correlates with PFKP expression, AKT S473 phosphorylation, and poor prognosis. These findings underscore the potential role and regulation of PFKP in human glioblastoma development.Phosphofructokinase 1 (PFK1) plays a critical role in glycolysis. Here the authors show that PFK1 platelet isoform is upregulated in Glioblastoma and is required for tumor growth mechanistically, such upregulation is due to an increased stability induced by AKT activation via phosphorylation on residue S386.


Brain Neoplasms/metabolism , Carcinogenesis , Glioblastoma/metabolism , Phosphofructokinase-1, Type C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , Cell Proliferation , Glycolysis , Humans , PTEN Phosphohydrolase/metabolism , Phosphorylation
13.
Mol Cell ; 65(5): 917-931.e6, 2017 Mar 02.
Article En | MEDLINE | ID: mdl-28238651

Autophagy is crucial for maintaining cell homeostasis. However, the precise mechanism underlying autophagy initiation remains to be defined. Here, we demonstrate that glutamine deprivation and hypoxia result in inhibition of mTOR-mediated acetyl-transferase ARD1 S228 phosphorylation, leading to ARD1-dependent phosphoglycerate kinase 1 (PGK1) K388 acetylation and subsequent PGK1-mediated Beclin1 S30 phosphorylation. This phosphorylation enhances ATG14L-associated class III phosphatidylinositol 3-kinase VPS34 activity by increasing the binding of phosphatidylinositol to VPS34. ARD1-dependent PGK1 acetylation and PGK1-mediated Beclin1 S30 phosphorylation are required for glutamine deprivation- and hypoxia-induced autophagy and brain tumorigenesis. Furthermore, PGK1 K388 acetylation levels correlate with Beclin1 S30 phosphorylation levels and poor prognosis in glioblastoma patients. Our study unearths an important mechanism underlying cellular-stress-induced autophagy initiation in which the protein kinase activity of the metabolic enzyme PGK1 plays an instrumental role and reveals the significance of the mutual regulation of autophagy and cell metabolism in maintaining cell homeostasis.


Autophagosomes/enzymology , Autophagy , Beclin-1/metabolism , Brain Neoplasms/enzymology , Glioblastoma/enzymology , Phosphoglycerate Kinase/metabolism , Acetylation , Animals , Autophagosomes/pathology , Beclin-1/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Class III Phosphatidylinositol 3-Kinases/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Female , Glioblastoma/genetics , Glioblastoma/pathology , Glutamine/deficiency , HEK293 Cells , Humans , Mice, Nude , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/genetics , N-Terminal Acetyltransferase E/metabolism , Phosphoglycerate Kinase/genetics , Phosphorylation , Protein Binding , RNA Interference , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection , Tumor Burden , Tumor Hypoxia
14.
Cell Rep ; 15(12): 2665-78, 2016 06 21.
Article En | MEDLINE | ID: mdl-27292636

The histone demethylase LSD1 facilitates epithelial-to-mesenchymal transition (EMT) and tumor progression by repressing epithelial marker expression. However, little is known about how its function may be modulated. Here, we report that LSD1 is acetylated in epithelial but not mesenchymal cells. Acetylation of LSD1 reduces its association with nucleosomes, thus increasing histone H3K4 methylation at its target genes and activating transcription. The MOF acetyltransferase interacts with LSD1 and is responsible for its acetylation. MOF is preferentially expressed in epithelial cells and is downregulated by EMT-inducing signals. Expression of exogenous MOF impedes LSD1 binding to epithelial gene promoters and histone demethylation, thereby suppressing EMT and tumor invasion. Conversely, MOF depletion enhances EMT and tumor metastasis. In human cancer, high MOF expression correlates with epithelial markers and a favorable prognosis. These findings provide insight into the regulation of LSD1 and EMT and identify MOF as a critical suppressor of EMT and tumor progression.


Epithelial-Mesenchymal Transition , Histone Acetyltransferases/metabolism , Histone Demethylases/metabolism , Acetylation , Cadherins/metabolism , Cell Line, Tumor , Disease Progression , Down-Regulation/genetics , Embryo, Mammalian/cytology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Multiprotein Complexes/metabolism , Mutation/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Nucleosomes/metabolism
15.
Oncotarget ; 7(18): 25113-24, 2016 May 03.
Article En | MEDLINE | ID: mdl-26848621

The Warburg effect, which reflects cancer cells' preference for aerobic glycolysis over glucose oxidation, contributes to tumor growth, progression and therapy resistance. The restraint on pyruvate flux into mitochondrial oxidative metabolism in cancer cells is in part attributed to the inhibition of pyruvate dehydrogenase (PDH) complex. Src is a prominent oncogenic non-receptor tyrosine kinase that promotes cancer cell proliferation, invasion, metastasis and resistance to conventional and targeted therapies. However, the potential role of Src in tumor metabolism remained unclear. Here we report that activation of Src attenuated PDH activity and generation of reactive oxygen species (ROS). Conversely, Src inhibitors activated PDH and increased cellular ROS levels. Src inactivated PDH through direct phosphorylation of tyrosine-289 of PDH E1α subunit (PDHA1). Indeed, Src was the main kinase responsible for PDHA1 tyrosine phosphorylation in cancer cells. Expression of a tyrosine-289 non-phosphorable PDHA1 mutant in Src-hyperactivated cancer cells restored PDH activity, increased mitochondrial respiration and oxidative stress, decreased experimental metastasis, and sensitized cancer cells to pro-oxidant treatment. The results suggest that Src contributes to the Warburg phenotype by inactivating PDH through tyrosine phosphorylation, and the metabolic effect of Src is essential for Src-driven malignancy and therapy resistance. Combination therapies consisting of both Src inhibitors and pro-oxidants may improve anticancer efficacy.


Drug Resistance, Neoplasm/physiology , Neoplasms/metabolism , Pyruvate Dehydrogenase Complex/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Glycolysis/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Reactive Oxygen Species/metabolism , Tyrosine/metabolism
16.
Sci Rep ; 5: 17545, 2015 Dec 02.
Article En | MEDLINE | ID: mdl-26626148

Green tea polyphenols (GTP) are highly effective in inhibiting a variety of tumorigenic effects induced by carcinogens. In this study we assessed GTP mitigation on biomarkers of fumonisin B1 (FB1), a class 2B carcinogen, in blood and urine samples collected from an intervention trial. A total of 124 exposed people were recruited and randomly assigned to low-dose (GTP 500 mg, n = 42), high-dose (GTP 1,000 mg, n = 41) or placebo (n = 41) for 3 months. After one-month of intervention, urinary FB1 was significantly decreased in high-dose group compared to that of placebo group (p = 0.045), with reduction rates of 18.95% in the low-dose group and 33.62% in the high-dose group. After three-month intervention, urinary FB1 showed significant decrease in both low-dose (p = 0.016) and the high-dose (p = 0.0005) groups compared to that of both placebo group and baseline levels, with reduction rates of 40.18% in the low-dose group and 52.6% in the high-dose group. GTP treatment also significantly reduced urinary excretion of sphinganine (Sa), sphingosine (So), and Sa/So ratio, but had no effect on serum Sa, So, and Sa/So ratio. Analysis with mixed-effect model revealed significant interactions between time and treatment effects of GTP on both urinary free FB1 levels and Sa/So ratios.


Biomarkers, Tumor , Carcinoma, Hepatocellular , Fumonisins , Liver Neoplasms , Polyphenols/administration & dosage , Tea/chemistry , Adult , Biomarkers, Tumor/blood , Biomarkers, Tumor/urine , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/urine , Female , Fumonisins/blood , Fumonisins/urine , Humans , Liver Neoplasms/blood , Liver Neoplasms/drug therapy , Liver Neoplasms/urine , Male , Middle Aged , Polyphenols/chemistry
17.
Cancer Lett ; 362(1): 70-82, 2015 Jun 28.
Article En | MEDLINE | ID: mdl-25827072

The Snail family of transcription factors are core inducers of epithelial-to-mesenchymal transition (EMT). Here we show that the F-box protein FBXO11 recognizes and promotes ubiquitin-mediated degradation of multiple Snail family members including Scratch. The association between FBXO11 and Snai1 in vitro is independent of Snai1 phosphorylation. Overexpression of FBXO11 in mesenchymal cells reduces Snail protein abundance and cellular invasiveness. Conversely, depletion of endogenous FBXO11 in epithelial cancer cells causes Snail protein accumulation, EMT, and tumor invasion, as well as loss of estrogen receptor expression in breast cancer cells. Expression of FBXO11 is downregulated by EMT-inducing signals TGFß and nickel. In human cancer, high FBXO11 levels correlate with expression of epithelial markers and favorable prognosis. The results suggest that FBXO11 sustains the epithelial state and inhibits cancer progression. Inactivation of FBXO11 in mice leads to neonatal lethality, epidermal thickening, and increased Snail protein levels in epidermis, validating that FBXO11 is a physiological ubiquitin ligase of Snail. Moreover, in C. elegans, the FBXO11 mutant phenotype is attributed to the Snail factors as it is suppressed by inactivation/depletion of Snail homologs. Collectively, these findings suggest that the FBXO11-Snail regulatory axis is evolutionarily conserved and critically governs carcinoma progression and mammalian epidermal development.


Breast Neoplasms/metabolism , Epidermis/metabolism , F-Box Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/pathology , Caenorhabditis elegans , Disease Progression , Epidermis/growth & development , Epithelial-Mesenchymal Transition , F-Box Proteins/genetics , Female , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Protein-Arginine N-Methyltransferases/genetics , Snail Family Transcription Factors , Ubiquitination
18.
Cancer Lett ; 356(2 Pt A): 156-64, 2015 Jan 28.
Article En | MEDLINE | ID: mdl-24732809

Compared to normal cells, cancer cells strongly upregulate glucose uptake and glycolysis to give rise to increased yield of intermediate glycolytic metabolites and the end product pyruvate. Moreover, glycolysis is uncoupled from the mitochondrial tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) in cancer cells. Consequently, the majority of glycolysis-derived pyruvate is diverted to lactate fermentation and kept away from mitochondrial oxidative metabolism. This metabolic phenotype is known as the Warburg effect. While it has become widely accepted that the glycolytic intermediates provide essential anabolic support for cell proliferation and tumor growth, it remains largely elusive whether and how the Warburg metabolic phenotype may play a role in tumor progression. We hereby review the cause and consequence of the restrained oxidative metabolism, in particular in the context of tumor metastasis. Cells change or lose their extracellular matrix during the metastatic process. Inadequate/inappropriate matrix attachment generates reactive oxygen species (ROS) and causes a specific type of cell death, termed anoikis, in normal cells. Although anoikis is a barrier to metastasis, cancer cells have often acquired elevated threshold for anoikis and hence heightened metastatic potential. As ROS are inherent byproducts of oxidative metabolism, forced stimulation of glucose oxidation in cancer cells raises oxidative stress and restores cells' sensitivity to anoikis. Therefore, by limiting the pyruvate flux into mitochondrial oxidative metabolism, the Warburg effect enables cancer cells to avoid excess ROS generation from mitochondrial respiration and thus gain increased anoikis resistance and survival advantage for metastasis. Consistent with this notion, pro-metastatic transcription factors HIF and Snail attenuate oxidative metabolism, whereas tumor suppressor p53 and metastasis suppressor KISS1 promote mitochondrial oxidation. Collectively, these findings reveal mitochondrial oxidative metabolism as a critical suppressor of metastasis and justify metabolic therapies for potential prevention/intervention of tumor metastasis.


Anoikis/physiology , Glycolysis/physiology , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Cell Proliferation , Citric Acid Cycle , Glucose/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kisspeptins/metabolism , Lactic Acid/metabolism , Neoplasm Metastasis , Oxidative Phosphorylation , Oxidative Stress/physiology , Pyruvic Acid/metabolism , Reactive Oxygen Species/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
19.
PLoS One ; 9(3): e92992, 2014.
Article En | MEDLINE | ID: mdl-24664296

Benzo[a]pyrene (BaP), a well-known environmental carcinogen, promotes oxidative stress and DNA damage. Curcumin and vitamin E (VE) have potent antioxidative activity that protects cells from oxidative stress and cellular damage. The objectives of the present study were to investigate the adverse effects of BaP on normal human lung epithelial cells (BEAS-2B), the potential protective effects of curcumin and VE against BaP-induced cellular damage, and the molecular mechanisms of action. MTT assay, flow cytometry, fluorescence microplate assay, HPLC, qRT-PCR, and western blot were performed to analyze cytotoxicity, cell cycle, reactive oxygen species (ROS), BaP diol-epoxidation (BPDE)-DNA adducts, gene expression, and protein expression, respectively. Curcumin or VE prevented cells from BaP-induced cell cycle arrest and growth inhibition, significantly suppressed BaP-induced ROS levels, and decreased BPDE-DNA adducts. While CYP1A1 and 1B1 were induced by BaP, these inductions were not significantly reduced by curcumin or VE. Moreover, the level of activated p53 and PARP-1 were significantly induced by BaP, whereas this induction was markedly reduced after curcumin and VE co-treatment. Survivin was significantly down-regulated by BaP, and curcumin significantly restored survivin expression in BaP-exposed cells. The ratio of Bax/Bcl-2 was also significantly increased in cells exposed to BaP and this increase was reversed by VE co-treatment. Taken together, BaP-induced cytotoxicity occurs through DNA damage, cell cycle arrest, ROS production, modulation of metabolizing enzymes, and the expression/activation of p53, PARP-1, survivin, and Bax/Bcl-2. Curcumin and VE could reverse some of these BaP-mediated alterations and therefore be effective natural compounds against the adverse effects of BaP in lung cells.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Benzo(a)pyrene/toxicity , Curcumin/pharmacology , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , Vitamin E/pharmacology , Cell Line , DNA Damage , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Reactive Oxygen Species/metabolism , Respiratory Mucosa/pathology
20.
J Biol Chem ; 288(38): 27680-27691, 2013 Sep 20.
Article En | MEDLINE | ID: mdl-23928305

Chromatin readers decipher the functional readouts of histone modifications by recruiting specific effector complexes for subsequent epigenetic reprogramming. The LSD1 (also known as KDM1A) histone demethylase complex modifies chromatin and represses transcription in part by catalyzing demethylation of dimethylated histone H3 lysine 4 (H3K4me2), a mark for active transcription. However, none of its currently known subunits recognizes methylated histones. The Snai1 family transcription factors are central drivers of epithelial-to-mesenchymal transition (EMT) by which epithelial cells acquire enhanced invasiveness. Snai1-mediated transcriptional repression of epithelial genes depends on its recruitment of the LSD1 complex and ensuing demethylation of H3K4me2 at its target genes. Through biochemical purification, we identified the MBT domain-containing protein SFMBT1 as a novel component of the LSD1 complex associated with Snai1. Unlike other mammalian MBT domain proteins characterized to date that selectively recognize mono- and dimethylated lysines, SFMBT1 binds di- and trimethyl H3K4, both of which are enriched at active promoters. We show that SFMBT1 is essential for Snai1-dependent recruitment of LSD1 to chromatin, demethylation of H3K4me2, transcriptional repression of epithelial markers, and induction of EMT by TGFß. Carcinogenic metal nickel is a widespread environmental and occupational pollutant. Nickel alters gene expression and induces EMT. We demonstrate the nickel-initiated effects are dependent on LSD1-SFMBT1-mediated chromatin modification. Furthermore, in human cancer, expression of SFMBT1 is associated with mesenchymal markers and unfavorable prognosis. These results highlight a critical role of SFMBT1 in epigenetic regulation, EMT, and cancer.


Chromatin/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Histone Demethylases/metabolism , Histones/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Repressor Proteins/metabolism , Carcinogens/pharmacology , Chromatin/genetics , Chromatin/pathology , Epithelial Cells/pathology , HEK293 Cells , Histone Demethylases/genetics , Histones/genetics , Humans , Methylation , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Nickel/adverse effects , Nickel/pharmacology , Repressor Proteins/genetics , Snail Family Transcription Factors , Trace Elements , Transcription Factors/genetics , Transcription Factors/metabolism
...