Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
G3 (Bethesda) ; 13(12)2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37857450

ABSTRACT

Endothelial-to-mesenchymal transition (EndoMT), a specific form of epithelial-to-mesenchymal transition, drives a growing number of human (Homo sapiens) pathological conditions. This emerging knowledge opens a path to discovering novel therapeutic targets for many EndoMT-associated disorders. Here, we constructed an atlas of the endothelial-cell transcriptome and demonstrated EndoMT-induced global changes in transcriptional gene expression. Our gene ontology analyses showed that EndoMT could be a specific checkpoint for leukocyte chemotaxis, adhesion, and transendothelial migration. We also identified distinct gene expression signatures underlying EndoMT across arterial, venous, and microvascular endothelial cells. We performed protein-protein interaction network analyses, identifying a class of highly connected hub genes in endothelial cells from different vascular beds. Moreover, we found that the short-chain fatty acid acetate strongly inhibits the transcriptional program of EndoMT in endothelial cells from different vascular beds across tissues. Our results reveal the molecular signature and cell-type difference of EndoMT across distinct tissue- and vascular-bed-specific endothelial cells, providing a powerful discovery tool and resource value. These results suggest that therapeutically manipulating the endothelial transcriptome could treat an increasing number of EndoMT-associated pathological conditions.


Subject(s)
Endothelial Cells , Epithelial-Mesenchymal Transition , Humans , Endothelial Cells/metabolism , Cells, Cultured , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling
2.
Mol Biol Cell ; 34(7): br10, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37133968

ABSTRACT

The short-chain fatty acid metabolite acetyl-coenzyme A (acetyl-CoA) has emerged as a major signal transducer that can broadly affect cell fate and function, at least partly by influencing acetylation of key proteins. The mechanism by which acetyl-CoA regulates CD4+ T-cell fate determination remains poorly understood. Herein, we report that acetate modulates glyceraldehyde-3-phosphate dehydrogenase (GAPDH) acetylation and CD4+ T helper 1 (Th1) cell differentiation by altering acetyl-CoA levels. Our transcriptome profiling shows that acetate is a robust positive regulator of CD4+ T-cell gene expression typical of glycolysis. We further show that acetate potentiates GAPDH activity, aerobic glycolysis, and Th1 polarization through regulation of GAPDH acetylation levels. This acetate-dependent GAPDH acetylation occurs in a dose- and time-dependent manner, while decreasing acetyl-CoA levels by fatty acid oxidation inhibition results in a decline in acetyl-GAPDH levels. Thus, acetate functions as a potent metabolic regulator in CD4+ T-cells by promoting GAPDH acetylation and Th1 cell fate decision.


Subject(s)
Acetates , Acetyl Coenzyme A/metabolism , Acetylation , Cell Differentiation , Acetates/pharmacology , Acetates/metabolism
3.
Mol Biol Cell ; 34(8): br11, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37163337

ABSTRACT

As an important substrate for cell metabolism, the short-chain fatty acid acetate emerges as a regulator of cell fate and function. However, its role in T-cell survival and its underlying mechanisms remain largely unknown. Here, we demonstrate that acetate modulates T-cell apoptosis via potentiation of α-tubulin acetylation. We further show that acetate treatment effectively increases the expression of the tumor necrosis factor receptor (TNFR) family member CD30 by enhancing its gene transcription. Moreover, CD30 physically associates with and stabilizes the deacetylase HDAC6, which deacetylates α-tubulin to decrease microtubule stability. Proteomic profiling of CD30 knockout (Cd30-/-) T-cells reveals elevated expression of anti-apoptotic BCL2 family proteins and thus promotes T-cell survival via a microtubule-Bcl-2 axis. Taken together, our results demonstrate that acetate is a regulator of T-cell survival by controlling levels of acetylated α-tubulin. This suggests that therapeutic manipulation of acetate metabolism may facilitate optimal T-cell responses in pathological conditions.


Subject(s)
Proteomics , Tubulin , Tubulin/metabolism , Histone Deacetylase 6/metabolism , Cell Survival , T-Lymphocytes/metabolism , Apoptosis Regulatory Proteins/metabolism , Acetates/pharmacology , Fatty Acids, Volatile , Acetylation
4.
Mol Biol Cell ; 33(8)2022 07 01.
Article in English | MEDLINE | ID: mdl-35544303

ABSTRACT

Normal tissue and organ morphogenesis requires epithelial cell plasticity and conversion to a mesenchymal phenotype through a tightly regulated process-epithelial-to-mesenchymal transition (EMT). Alterations of EMT go far beyond cell-lineage segregation and contribute to pathologic conditions such as cancer. EMT is subject to intersecting control pathways; however, EMT's metabolic mechanism remains poorly understood. Here, we demonstrate that transforming growth factor ß (TGF-ß)-induced EMT is accompanied by decreased fatty acid oxidation (FAO) and reduced acetyl-coenzyme A (acetyl-CoA) levels. Acetyl-CoA is a central metabolite and the sole donor of acetyl groups to acetylate key proteins. Further, the short-chain fatty acid acetate increases acetyl-CoA levels--robustly inhibiting EMT and cancer cell migration. Acetate can restore EMT-associated α-tubulin acetylation levels, increasing microtubule stability. Transcriptome profiling and flow cytometric analysis show that acetate inhibits the global gene expression program associated with EMT and the EMT-associated G1 cell cycle arrest. Taken together, these results demonstrate that acetate is a potent metabolic regulator of EMT and that therapeutic manipulation of acetate metabolism could provide the basis for treating a wide range of EMT-linked pathological conditions, including cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Transforming Growth Factor beta , Acetates/pharmacology , Acetyl Coenzyme A , Epithelial-Mesenchymal Transition/genetics , Fatty Acids, Volatile , Transforming Growth Factor beta/pharmacology
5.
Nat Commun ; 11(1): 5105, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037191

ABSTRACT

RB1 mutational inactivation is a cancer driver in various types of cancer including lung cancer, making it an important target for therapeutic exploitation. We performed chemical and genetic vulnerability screens in RB1-isogenic lung cancer pair and herein report that aurora kinase A (AURKA) inhibition is synthetic lethal in RB1-deficient lung cancer. Mechanistically, RB1-/- cells show unbalanced microtubule dynamics through E2F-mediated upregulation of the microtubule destabilizer stathmin and are hypersensitive to agents targeting microtubule stability. Inhibition of AURKA activity activates stathmin function via reduced phosphorylation and facilitates microtubule destabilization in RB1-/- cells, heavily impacting the bipolar spindle formation and inducing mitotic cell death selectively in RB1-/- cells. This study shows that stathmin-mediated disruption of microtubule dynamics is critical to induce synthetic lethality in RB1-deficient cancer and suggests that upstream factors regulating microtubule dynamics, such as AURKA, can be potential therapeutic targets in RB1-deficient cancer.


Subject(s)
Aurora Kinase A/genetics , Lung Neoplasms/genetics , Microtubules/metabolism , Retinoblastoma Binding Proteins/genetics , Stathmin/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Microtubules/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Retinoblastoma Binding Proteins/metabolism , Stathmin/genetics , Synthetic Lethal Mutations , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
6.
Int J Biol Sci ; 16(11): 1774-1784, 2020.
Article in English | MEDLINE | ID: mdl-32398948

ABSTRACT

PTEN, a tumor suppressor, is found loss of function in many cancers, including colorectal cancer. To identify the synthetic lethal compounds working with PTEN deficiency, we performed a synthetic lethality drug screening with PTEN-isogenic colorectal cancer cells. From the screening, we found that PTEN-/- colorectal cancer cells were sensitive to anacardic acid, a p300/CBP histone acetyltransferase (HAT) inhibitor. Anacardic acid significantly reduced the viability of PTEN-/- cells not in PTEN+/+ cells via inducing apoptosis. Inhibition of HAT activity of p300/CBP by anacardic acid reduced the acetylation of histones at the promoter region and inhibited the transcription of Hsp70 family of proteins. The down-regulation of Hsp70 family proteins led to the reduction of AKT-Hsp70 complex formation, AKT destabilization and decreased the level of phosphorylated AKT at Ser473, all of which are vital for the survival of PTEN-/- colorectal cells. The synthetic lethality effect of anacardic acid was further validated in tumor xenograft mice models, where PTEN-/- colorectal tumors showed greater sensitivity to anacardic acid treatment than PTEN+/+ tumors. These data suggest that anacardic acid induced synthetic lethality by inhibiting HAT activity of p300/CBP, thereby reducing Hsp70 transcription and destabilizing AKT in PTEN deficient colorectal cancer cells.


Subject(s)
Anacardic Acids/therapeutic use , Colorectal Neoplasms/drug therapy , PTEN Phosphohydrolase/deficiency , Proto-Oncogene Proteins c-akt , p300-CBP Transcription Factors/antagonists & inhibitors , Anacardic Acids/chemistry , Anacardic Acids/pharmacology , Animals , Colorectal Neoplasms/pathology , Combinatorial Chemistry Techniques , Down-Regulation , Drug Design , Drug Discovery , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neoplasms, Experimental , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/drug therapy , Synthetic Lethal Mutations , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/metabolism
7.
Acta Pharm Sin B ; 10(4): 615-627, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32322466

ABSTRACT

Breast cancer susceptibility gene 1 (BRCA1) is a tumor suppressor gene, which is frequently mutated in breast and ovarian cancers. BRCA1 plays a key role in the homologous recombination directed DNA repair, allowing its deficiency to act as a therapeutic target of DNA damaging agents. In this study, we found that inhibition of the class I histone deacetylases (HDAC) exhibited synthetic lethality with BRCA1 deficiency in breast cancer cells. Transcriptome profiling and validation study showed that HDAC inhibition enhanced the expression of thioredoxin interaction protein (TXNIP), causing reactive oxygen species (ROS)-mediated DNA damage. This effect induced preferential apoptosis in BRCA1 -/- breast cancer cells where DNA repair system is compromised. Two animal experiments and gene expression-associated patients' survival analysis further confirmed in vivo synthetic lethality between BRCA1 and HDAC. Finally, the combination of inhibitors of HDAC and bromodomain and extra-terminal motif (BET), another BRCA1 synthetic lethality target that also works through oxidative stress-mediated DNA damage, showed a strong anticancer effect in BRCA1 -/- breast cancer cells. Together, this study provides a new therapeutic strategy for BRCA1-deficient breast cancer by targeting two epigenetic machineries, HDAC and BET.

8.
Cells ; 8(5)2019 04 28.
Article in English | MEDLINE | ID: mdl-31035320

ABSTRACT

Cholesterol is an essential structural component of cellular membranes. In addition to the structural role, it also serves as a precursor to a variety of steroid hormones and has diverse functions in intracellular signal transduction. As one of its functions in cell signaling, recent evidence suggests that cholesterol plays a key role in regulating angiogenesis. This review discusses the role of cholesterol in angiogenesis, with a particular emphasis on cholesterol trafficking in endothelial cell signaling. Small molecule inhibitors of cholesterol trafficking and their preclinical and clinical development targeting angiogenesis and cancer are also discussed.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cholesterol/physiology , Endothelial Cells , Neoplasms , Neovascularization, Pathologic , Animals , Biological Transport/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Signal Transduction/drug effects
9.
Int J Biol Sci ; 14(10): 1175-1185, 2018.
Article in English | MEDLINE | ID: mdl-30123067

ABSTRACT

Cholesterol plays a key role in membrane protein function and signaling in endothelial cells. Thus, disturbing cholesterol trafficking is an effective approach for inhibiting angiogenesis. We recently identified astemizole (AST), an antihistamine drug, as a cholesterol trafficking inhibitor from a phenotypic screen. In this study, we found that AST induced cholesterol accumulation in the lysosome by binding to the sterol-sensing domain of Niemann-Pick disease, type C1 (NPC1), a lysosomal surface protein responsible for cholesterol transport. Inhibition of cholesterol trafficking by AST led to the depletion of membrane cholesterol, causing SREBP1 nuclear localization. The depletion of membrane cholesterol resulted in dissociation of mammalian target of rapamycin (mTOR) from the lysosomal surface and inactivation of mTOR signaling. These effects were effectively rescued by addition of exogenous cholesterol. AST inhibited endothelial cell proliferation, migration and tube formation in a cholesterol-dependent manner. Furthermore, AST inhibited zebrafish angiogenesis in a cholesterol-dependent manner. Together, our data suggest that AST is a new class of NPC1 antagonist that inhibits cholesterol trafficking in endothelial cells and angiogenesis.


Subject(s)
Astemizole/therapeutic use , Cholesterol/metabolism , Neovascularization, Pathologic/drug therapy , TOR Serine-Threonine Kinases/metabolism , A549 Cells , Biological Transport/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells , Humans , Niemann-Pick C1 Protein/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
10.
Cancer Lett ; 436: 28-37, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30118842

ABSTRACT

PTEN is a tumor suppressor found mutated in many cancers. From a synthetic lethality drug screen with PTEN-isogenic colorectal cancer cells, we found that mutant-PTEN cells were resistant to dual inhibitors of FLT3 and aurora kinase-A, including KW2449 and ENMD-2076. KW2449 significantly reduced the viability of wildtype-PTEN cells causing apoptosis, while little effect was observed in mutant-PTEN counterparts. Transcriptome profiling showed that members of PI3K-AKT signaling pathway were strongly changed in cells after KW2449 treatment, indicating a potential role of the pathway in drug resistance. We found that KW2449 caused a dose-dependent, biphasic induction of AKT phosphorylation at Ser473 in mutant-PTEN cells. Co-treatment with the inhibitors of its upstream signaling completely abolished the reactivation of AKT phosphorylation by KW2449 and reversed the drug resistant phenotype. These data suggest that reactivation of AKT phosphorylation at Ser473 is a key factor to confer drug resistant phenotype of mutant-PTEN cells to the dual inhibitors and that proper drug combinations that shut down AKT reactivation is necessary for the effective treatment of mutant-PTEN cancer with the dual inhibitors in clinical settings.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Indazoles/pharmacology , PTEN Phosphohydrolase/deficiency , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Female , HCT116 Cells , Humans , Indazoles/administration & dosage , Mice, Nude , Mutation , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Piperazines/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction/drug effects , Signal Transduction/genetics , Xenograft Model Antitumor Assays/methods , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
11.
Nat Commun ; 9(1): 3212, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30097580

ABSTRACT

ARID1A, a component of the SWI/SNF chromatin remodeling complex, is a tumor suppressor with a high frequency of inactivating mutations in many cancers. Therefore, ARID1A deficiency has been exploited therapeutically for treating cancer. Here we show that ARID1A has a synthetic lethal interaction with aurora kinase A (AURKA) in colorectal cancer (CRC) cells. Pharmacological and genetic perturbations of AURKA selectively inhibit the growth of ARID1A-deficient CRC cells. Mechanistically, ARID1A occupies the AURKA gene promoter and negatively regulates its transcription. Cells lacking ARID1A show enhanced AURKA transcription, which leads to the persistent activation of CDC25C, a key protein for G2/M transition and mitotic entry. Inhibiting AURKA activity in ARID1A-deficient cells significantly increases G2/M arrest and induces cellular multinucleation and apoptosis. This study shows a novel synthetic lethality interaction between ARID1A and AURKA and indicates that pharmacologically inhibiting the AURKA-CDC25C axis represents a novel strategy for treating CRC with ARID1A loss-of-function mutations.


Subject(s)
Aurora Kinase A/metabolism , Colorectal Neoplasms/genetics , Nuclear Proteins/deficiency , Signal Transduction , Synthetic Lethal Mutations/genetics , Transcription Factors/deficiency , cdc25 Phosphatases/metabolism , Animals , Apoptosis , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins , Drug Evaluation, Preclinical , Female , G2 Phase , Gene Knockout Techniques , Humans , Mice, Inbred BALB C , Mice, Nude , Mitosis , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic
12.
Oncogene ; 37(49): 6341-6356, 2018 12.
Article in English | MEDLINE | ID: mdl-30042414

ABSTRACT

BRCA1 is a tumor suppressor frequently mutated in breast and ovarian cancer, serving it as a target for therapeutic exploitation. Here, we show that BRCA1 has a synthetic lethality interaction with an epigenetics regulator, bromodomain and extra-terminal domain (BET). BET inhibition led to gene expression changes reversing MYC-dependent transcription repression of a redox regulator, thioredoxin-interacting protein (TXNIP), via switching the promoter occupant from MYC to MondoA:MLX complex. Reversing the MYC-TXNIP axis inhibited thioredoxin activity and elevated cellular oxidative stress, causing DNA damages that are detrimental to BRCA1-deficient breast cancer cells. Tumor xenograft models and breast cancer clinical data analyses further demonstrated an in vivo synthetic lethality interaction and clinical association between BET/TXNIP and BRCA1 deficiency in the survival of breast cancer patients.


Subject(s)
BRCA1 Protein/deficiency , Breast Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Genes, BRCA1 , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Quinazolinones/pharmacology , Xenograft Model Antitumor Assays
13.
Cancer Lett ; 409: 91-103, 2017 11 28.
Article in English | MEDLINE | ID: mdl-28923401

ABSTRACT

Cholesterol is an important modulator of membrane protein function and signaling in endothelial cells, thus making it an emerging target for anti-angiogenic agents. In this study, we employed a phenotypic screen that detects intracellular cholesterol distribution in endothelial cells (HUVEC) and identified 13 existing drugs as cholesterol trafficking inhibitors. Cepharanthine, an approved drug for anti-inflammatory and cancer management use, was amongst the candidates, which was selected for in-depth mechanistic studies to link cholesterol trafficking and angiogenesis. Cepharanthine inhibited the endolysosomal trafficking of free-cholesterol and low-density lipoprotein in HUVEC by binding to Niemann-Pick disease, type C1 (NPC1) protein and increasing the lysosomal pH. The blockade of cholesterol trafficking led to a cholesterol-dependent dissociation of mTOR from the lysosomes and inhibition of its downstream signaling. Cepharanthine inhibited angiogenesis in HUVEC and in zebrafish in a cholesterol-dependent manner. Furthermore, cepharanthine suppressed tumor growth in vivo by inhibiting angiogenesis and it enhanced the antitumor activity of the standard chemotherapy cisplatin in lung and breast cancer xenografts in mice. Altogether, these results strongly support the idea that cholesterol trafficking is a viable drug target for anti-angiogenesis and that the inhibitors identified among existing drugs, such as cepharanthine, could be potential anti-angiogenic and antitumor agents.


Subject(s)
Benzylisoquinolines/pharmacology , Cholesterol/metabolism , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzylisoquinolines/administration & dosage , Cell Growth Processes/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Drug Synergism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL