Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Cardiovasc Pharmacol ; 72(5): 231-241, 2018 11.
Article in English | MEDLINE | ID: mdl-30399060

ABSTRACT

We investigated whether resveratrol (RSV) can attenuate obesity and diabetes progression and improve diabetes-induced vascular dysfunction, and we attempted to delineate its underlying mechanisms. Male C57Bl/6 mice were administered a high-fat diet (HFD) for 17 weeks. Mice developed type 2 diabetes with increased body weight, hyperglycemia, hyperinsulinemia, and hyperlipidemia. Oral gavage with RSV significantly reversed the symptoms induced by the HFD. Insulin sensitivity likewise improved after the RSV intervention in these mice. Phenylephrine-induced cremaster arteriolar constriction was impaired, whereas RSV treatment significantly mitigated the vessel responsiveness to phenylephrine. The obese diabetic mice exhibited increased leukocyte rolling, adhesion, and transmigration in the postcapillary venules of the cremaster muscle. By contrast, RSV treatment significantly attenuated HFD-induced extravasation. RSV significantly recovered phosphorylated Akt and eNOS expression in the thoracic aorta. In addition, activated adenosine monophosphate-activated protein kinase in the thoracic aorta was involved in the improvement of epithelial function after RSV intervention. RSV considerably upregulated the plasma NO level in HFD mice. Moreover, RSV-enhanced human umbilical vein endothelial cells healing through Sirt1/ER pathway may be involved in the prevention of leukocyte extravasation. Collectively, RSV attenuates diabetes-induced vascular dysfunction by activating Akt/eNOS/NO and Sirt1/ER pathway. Our mechanistic study provides a potential RSV-based therapeutic strategy against cardiovascular disease.


Subject(s)
Abdominal Muscles/blood supply , Blood Vessels/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/prevention & control , Diet, High-Fat , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/metabolism , Resveratrol/pharmacology , Sirtuin 1/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Blood Vessels/enzymology , Blood Vessels/physiopathology , Cells, Cultured , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/etiology , Diabetic Angiopathies/enzymology , Diabetic Angiopathies/etiology , Diabetic Angiopathies/physiopathology , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Male , Mice, Inbred C57BL , Microvessels/drug effects , Microvessels/enzymology , Microvessels/physiopathology , Phosphorylation , Signal Transduction/drug effects
2.
Mol Pharm ; 12(11): 3885-95, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26412161

ABSTRACT

The overexpression of the serine/threonine specific polo-like kinase 1 (Plk1) is associated with poor prognosis in many types of cancer. Consequently, Plk1 has emerged as a valid therapeutic target for anticancer drug design. Volasertib is a potent inhibitor of Plk1 that inhibits the proliferation of multiple human cancer cell lines by promoting cell cycle arrest at nanomolar concentrations. However, the risk of developing drug resistance, which is often associated with the overexpression of the ATP-binding cassette (ABC) transporter ABCB1 (P-glycoprotein), can present a therapeutic challenge for volasertib and many other therapeutic drugs. Although volasertib is highly effective against the proliferation of numerous cancer cell lines, we found that the overexpression of ABCB1 in cancer cells leads to cellular resistance to volasertib and reduces the level of volasertib-stimulated G2/M cell cycle arrest and subsequent onset of apoptosis. Furthermore, we demonstrate that volasertib competitively inhibits the function of ABCB1 and stimulates the basal ATPase activity of ABCB1 in a concentration-dependent manner, which is consistent with substrate transport by ABCB1. More importantly, we discovered that the coadministration of an inhibitor or drug substrate of ABCB1 restored the anticancer activity of volasertib in ABCB1-overexpressing cancer cells. In conclusion, the results of our study reveal that ABCB1 negatively affects the efficacy of volasertib and supports its combination with a modulator of ABCB1 to improve clinical responses.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adenosine Triphosphatases/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Neoplasms/metabolism , Neoplasms/pathology , Polo-Like Kinase 1
3.
J Med Biol Eng ; 35(2): 156-164, 2015.
Article in English | MEDLINE | ID: mdl-25960704

ABSTRACT

Pulsed-mode ultrasound (pUS) in combination with intravenously (IV) administered microbubbles (MBs) can enhance local drug delivery by temporarily enhancing capillary permeability. This study evaluates the use of epidermal growth factor receptor (EGFR)-targeting MBs after pUS treatment to enhance the effects of therapeutic-EGFR antibody delivery to glioma tumor cells in mice. Three animal groups were compared: (1) IV-injected non-targeting MBs, (2) IV-injected targeting MBs, and (3) IV-injected targeting MBs combined with pUS treatment. All animals were analyzed using high-frequency small-animal US imaging. The mean halftime of circulating targeting MBs was significantly increased from 3.13 min of targeting bubble alone to 5.86 min by targeting MBs combined with pUS treatment, compared to 2.34 min for non-targeting MBs. Compared to targeting bubble administration alone, pUS exposure prior to injection of targeting MBs was also significantly better at suppressing tumor growth when monitored for up to 35 days (p < 0.05). The final relative tumor volumes were 2664, 700, and 188 mm3 for non-targeting MBs, targeting MBs, and targeting MBs combined with pUS treatment, respectively. pUS treatment prolonged the mean circulatory halftime of targeting MBs and enhanced the anti-tumor effect of EGFR antibodies in a human glioma model in mice. Targeting MBs combined with pUS treatment thus has potential for enhanced therapeutic antibody delivery for facilitating anti-glioma treatment.

4.
J Biomed Sci ; 21: 37, 2014 May 03.
Article in English | MEDLINE | ID: mdl-24884386

ABSTRACT

BACKGROUND: An endogenous dopaminergic (DA) tone acting on D3 receptors has been shown to inhibit tuberoinfundibular (TI) DA neuron activity and stimulate prolactin (PRL) surge in the afternoon of estrogen-primed ovariectomized (OVX+E2) rats. Whether D2 receptor (D2R) is also involved in the regulation of TIDA and PRL rhythms was determined in this study. RESULTS: Intracerebroventricular (icv) injection of PHNO, a D2R agonist, in the morning inhibited TIDA and midbrain DA neurons' activities, and stimulated PRL secretion. The effects of PHNO were significantly reversed by co-administration of raclopride, a D2R antagonist. A single injection of raclopride at 1200 h significantly reversed the lowered TIDA neuron activity and the increased serum PRL level at 1500 h. Dopamine D2R mRNA expression in medial basal hypothalamus (MBH) exhibited a diurnal rhythm, i.e., low in the morning and high in the afternoon, which was opposite to that of TIDA neuron activity. The D2R rhythm was abolished in OVX+E2 rats kept under constant lighting but not in OVX rats with regular lighting exposures. Pretreatment with an antisense oligodeoxynucleotides (AODN, 10 µg/3 µl/day, icv) against D2R mRNA for 2 days significantly reduced D2R mRNAs in central DA neurons, and reversed both lowered TIDA neuron activity and increased serum PRL level in the afternoon on day 3. A diurnal rhythm of D2R mRNA expression was also observed in midbrain DA neurons and the rhythm was significantly knocked down by the AODN pretreatment. CONCLUSIONS: We conclude that a diurnal change of D2R mRNA expression in MBH may underlie the diurnal rhythms of TIDA neuron activity and PRL secretion in OVX+E2 rats.


Subject(s)
Circadian Rhythm/genetics , Dopaminergic Neurons/metabolism , Prolactin/metabolism , Receptors, Dopamine D2/metabolism , Animals , Dopamine/metabolism , Estrogens/metabolism , Female , Hypothalamus/metabolism , Infusions, Intraventricular , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Prolactin/blood , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/agonists
5.
bioRxiv ; 2024 May 13.
Article in English | MEDLINE | ID: mdl-38765991

ABSTRACT

Lipids are essential for tumours because of their structural, energetic, and signaling roles. While many cancer cells upregulate lipid synthesis, growing evidence suggests that tumours simultaneously intensify the uptake of circulating lipids carried by lipoproteins. Which mechanisms promote the uptake of extracellular lipids, and how this pool of lipids contributes to cancer progression, are poorly understood. Here, using functional genetic screens, we find that lipoprotein uptake confers resistance to lipid peroxidation and ferroptotic cell death. Lipoprotein supplementation robustly inhibits ferroptosis across numerous cancer types. Mechanistically, cancer cells take up lipoproteins through a pathway dependent on sulfated glycosaminoglycans (GAGs) linked to cell-surface proteoglycans. Tumour GAGs are a major determinant of the uptake of both low and high density lipoproteins. Impairment of glycosaminoglycan synthesis or acute degradation of surface GAGs decreases the uptake of lipoproteins, sensitizes cells to ferroptosis and reduces tumour growth in mice. We also find that human clear cell renal cell carcinomas, a distinctively lipid-rich tumour type, display elevated levels of lipoprotein-derived antioxidants and the GAG chondroitin sulfate than non-malignant human kidney. Altogether, our work identifies lipoprotein uptake as an essential anti-ferroptotic mechanism for cancer cells to overcome lipid oxidative stress in vivo, and reveals GAG biosynthesis as an unexpected mediator of this process.

6.
Langmuir ; 29(39): 12075-83, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24011019

ABSTRACT

A series of amphiphilic poly(amidoamine) dendrimers (PAMAM, G2-G5) composed of a hydrophilic core and a hydrophobic shell of aniline pentamer (AP) were synthesized and characterized. The modified dendrimers self-assembled to vesicular aggregates in water with the critical aggregation concentration (CAC) decreased in the order of G2 > G3 > G4 > G5. It was found that the modified dendrimers self-organized into spherical aggregates with a bilayer vesicular structures and that the dendrimers in higher generation have more order structure, which may be attributed to the crystallization induced by the compacted effect of AP segments. In addition, larger spherical vesicles were observed under acidic and alkaline conditions, as compared with sizes of aggregates in neutral medium. At low pH, the tertiary amine groups of PAMAM-AP were transformed to ammonium salts; the polarons were formed from AP units by doping with strong acids, thereby leading to the stability of vesicular aggregates being better than that in double distilled water. Nevertheless, in high pH environment, the deprotonation of PAMAM-AP caused the enhancement of π-π interactions, resulting in generation of twins or multilayered vesicles.


Subject(s)
Aniline Compounds/chemistry , Dendrimers/chemical synthesis , Polyamines/chemical synthesis , Surface-Active Agents/chemical synthesis , Dendrimers/chemistry , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Structure , Particle Size , Polyamines/chemistry , Surface Properties , Surface-Active Agents/chemistry
7.
Cancer Cell ; 7(6): 575-89, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15950906

ABSTRACT

Epidermal growth factor receptor (EGFR) exists in the nucleus of highly proliferative cells where it functions as a transcription factor. Although EGFR has transactivational activity, it lacks a DNA binding domain and, therefore, may require a DNA binding transcription cofactor for its transcriptional function. Here, we report that EGFR physically interacts with signal transducers and activators of transcription 3 (STAT3) in the nucleus, leading to transcriptional activation of inducible nitric oxide synthase (iNOS). In breast carcinomas, nuclear EGFR positively correlates with iNOS. This study describes a mode of transcriptional control involving cooperated efforts of STAT3 and nuclear EGFR. Our work suggests that the deregulated iNOS/NO pathway may partly contribute to the malignant biology of tumor cells with high levels of nuclear EGFR and STAT3.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , ErbB Receptors/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/physiology , Trans-Activators/metabolism , Animals , Base Sequence , Binding Sites/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cell Survival/drug effects , Chromatin Immunoprecipitation , Cricetinae , Cricetulus , Drug Synergism , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , Female , Gene Expression/drug effects , Gene Expression Regulation, Neoplastic , Genes, bcl-1/genetics , Genes, fos/genetics , HeLa Cells , Humans , Janus Kinase 2 , Microscopy, Fluorescence , Microscopy, Immunoelectron , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Phosphorylation/drug effects , Prognosis , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , S-Nitroso-N-Acetylpenicillamine/pharmacology , STAT3 Transcription Factor , Signal Transduction/drug effects , Survival Analysis
8.
Proc Natl Acad Sci U S A ; 107(37): 16125-30, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20802156

ABSTRACT

EGF induces the translocation of EGF receptor (EGFR) from the cell surface to the nucleus where EGFR activates gene transcription through its binding to an AT-rich sequence (ATRS) of the target gene promoter. However, how EGFR, without a DNA-binding domain, can bind to the gene promoter is unclear. In the present study, we show that RNA helicase A (RHA) is an important mediator for EGFR-induced gene transactivation. EGF stimulates the interaction of EGFR with RHA in the nucleus of cancer cells. The EGFR/RHA complex then associates with the target gene promoter through binding of RHA to the ATRS of the target gene promoter to activate its transcription. Knockdown of RHA expression in cancer cells abrogates the binding of EGFR to the target gene promoter, thereby reducing EGF/EGFR-induced gene expression. In addition, interruption of EGFR-RHA interaction decreases the EGFR-induced promoter activity. Consistently, we observed a positive correlation of the nuclear expression of EGFR, RHA, and cyclin D1 in human breast cancer samples. These results indicate that RHA is a DNA-binding partner for EGFR-mediated transcriptional activation in the nucleus.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , ErbB Receptors/metabolism , RNA Helicases/metabolism , Transcriptional Activation , Active Transport, Cell Nucleus , Breast Neoplasms/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Promoter Regions, Genetic , Protein Binding , RNA Helicases/genetics
9.
J Phys Chem Lett ; 14(25): 5924-5931, 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37343274

ABSTRACT

To explore non-adiabatic effects caused by electromagnetic (EM) vacuum fluctuations in molecules, we develop a general theory of internal conversion (IC) in the framework of quantum electrodynamics and propose a new mechanism, "quantum electrodynamic internal conversion" (QED-IC). The theory allows us to compute the rates of the conventional IC and QED-IC processes at the first-principles level. Our simulations manifest that, under experimentally feasible weak light-matter coupling conditions, EM vacuum fluctuations can significantly affect IC rates by an order of magnitude. Moreover, our theory elucidates three key factors in the QED-IC mechanism: the effective mode volume, coupling-weighted normal mode alignment, and molecular rigidity. The theory successfully captures the nucleus-photon interaction in the factor "coupling-weighted normal mode alignment". In addition, we find that molecular rigidity plays a totally different role in conventional IC versus QED-IC rates. Our study provides applicable design principles for exploiting QED effects on IC processes.

10.
J Biol Chem ; 286(23): 20558-68, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21487020

ABSTRACT

Epidermal growth factor receptor (EGFR), an aberrantly overexpressed or activated receptor-tyrosine kinase in many cancers, plays a pivotal role in cancer progression and has been an attractive target for cancer therapy. Gefitinib and erlotinib, two EGFR-tyrosine kinase inhibitors, have been approved for non-small cell lung cancer. However, durable clinical efficacy of these EGFR inhibitors is severely limited by the emergence of acquired resistance. For example, the expression of breast cancer-resistant protein (BCRP/ABCG2) has been shown to confer acquired resistance of wild-type EGFR (wtEGFR)-expressing cancer cells to gefitinib. However, the underlying molecular mechanisms still remain unclear. Here, we show that wtEGFR expression is elevated in the nucleus of acquired gefitinib-resistant cancer cells. Moreover, nuclear translocation of EGFR requires phosphorylation at Ser-229 by Akt. In the nucleus, EGFR then targets the proximal promoter of BCRP/ABCG2 and thereby enhances its gene transcription. The nuclear EGFR-mediated BCRP/ABCG2 expression may contribute at least in part to the acquired resistance of wtEGFR-expressing cancer cells to gefitinib. Our findings shed light on the role of nuclear EGFR in the sensitivity of wtEGFR-expressing cancer cells to EGFR tyrosine kinase inhibitors and also deciphered a putative molecular mechanism contributing to gefitinib resistance through BCRP/ABCG2 expression.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Nucleus/metabolism , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Female , Gefitinib , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics
11.
J Cell Physiol ; 227(4): 1446-54, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21618535

ABSTRACT

Hepatitis B virus (HBV) X protein (HBx) has been implicated in HBV-associated carcinogenesis through activation of IκB kinase (IKK)/nuclear factor kappa B (NF-κB) signaling pathway. Besides activating NF-κB in the cytoplasm, IKKα was found in the nucleus to regulate gene expression epigenetically in response to various stimuli. However, it is unknown whether nuclear IKKα plays a role in HBx-associated tumor progression. Moreover, the molecular mechanism underlying IKKα nuclear transport also remains to be elucidated. Here, we disclosed HBx as a new inducer of IKKα nuclear transport in hepatoma cells. HBx induced IKKα nuclear transport in an Akt-dependent manner. HBx-activated Akt promoted IKKα nuclear translocation via phosphorylating its threonine-23 (Thr23). In addition, IKKα ubiquitination enhanced by HBx and Akt also contributed to the IKKα accumulation in the nucleus, indicating the involvement of ubiquitination in Akt-increased IKKα nuclear transport in response to HBx. Furthermore, inhibition of IKKα nuclear translocation by mutation of its nuclear localization signal and Thr23 diminished IKKα-dependent cell migration. Taken together, our findings shed light on the molecular mechanism of IKKα nuclear translocation and provide a potential role of nuclear IKKα in HBx-mediated hepatocellular carcinoma (HCC) progression.


Subject(s)
Carcinoma, Hepatocellular/etiology , Hepatitis B virus/pathogenicity , I-kappa B Kinase/metabolism , Liver Neoplasms/etiology , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/toxicity , Viral Proteins/toxicity , Active Transport, Cell Nucleus/drug effects , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/physiopathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/physiology , Disease Progression , HEK293 Cells , Hep G2 Cells , Humans , I-kappa B Kinase/genetics , Liver Neoplasms/pathology , Liver Neoplasms/physiopathology , Mutagenesis, Site-Directed , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Nuclear Localization Signals/genetics , Phosphorylation , Ubiquitination , Viral Regulatory and Accessory Proteins
12.
Pharmaceutics ; 14(8)2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35893779

ABSTRACT

Epigallocatechin gallate (EGCG), a major tea catechin, enhances cellular uptake of magnetic nanoparticles (MNPs), but the mechanism remains unclear. Since EGCG may interact with the 67-kDa laminin receptor (67LR) and epidermal growth factor receptor (EGFR), we investigate whether a receptor and its downstream signaling may mediate EGCG's enhancement effects on nanoparticle uptake. As measured using a colorimetric iron assay, EGCG induced a concentration-dependent enhancement effect of MNP internalization by LN-229 glioma cells, which was synergistically enhanced by the application of a magnetic field. Transmission electron microscopy demonstrated that EGCG increased the number, but not the size, of internalized vesicles, whereas EGCG and the magnet synergistically increased the size of vesicles. EGCG appears to enhance particle-particle interaction and thus aggregation following a 5-min magnet application. An antibody against 67LR, knockdown of 67LR, and a 67LR peptide (amino acid 161-170 of 67LR) attenuated EGCG-induced MNP uptake by 35%, 100%, and 45%, respectively, suggesting a crucial role of 67LR in the effects of EGCG. Heparin, the 67LR-binding glycosaminoglycan, attenuated EGCG-induced MNP uptake in the absence, but not presence, of the magnet. Such enhancement effects of EGCG were attenuated by LY294002 (a phosphoinositide 3-kinase inhibitor) and Akt inhibitor, but not by agents affecting cGMP levels, suggesting potential involvement of signaling downstream of 67LR. In contrast, the antibody against EGFR exerted no effect on EGCG-enhanced internalization. These results suggest that 67LR may be potentially amenable to tumor-targeted therapeutics.

13.
Front Cell Dev Biol ; 10: 890419, 2022.
Article in English | MEDLINE | ID: mdl-35602596

ABSTRACT

TEAD4 (TEA Domain Transcription Factor 4) is well recognized as the DNA-anchor protein of YAP transcription complex, which is modulated by Hippo, a highly conserved pathway in Metazoa that controls organ size through regulating cell proliferation and apoptosis. To acquire full transcriptional activity, TEAD4 requires co-activator, YAP (Yes-associated protein) or its homolog TAZ (transcriptional coactivator with PDZ-binding motif) the signaling hub that relays the extracellular stimuli to the transcription of target genes. Growing evidence suggests that TEAD4 also exerts its function in a YAP-independent manner through other signal pathways. Although TEAD4 plays an essential role in determining that differentiation fate of the blastocyst, it also promotes tumorigenesis by enhancing metastasis, cancer stemness, and drug resistance. Upregulation of TEAD4 has been reported in several cancers, including colon cancer, gastric cancer, breast cancer, and prostate cancer and serves as a valuable prognostic marker. Recent studies show that TEAD4, but not other members of the TEAD family, engages in regulating mitochondrial dynamics and cell metabolism by modulating the expression of mitochondrial- and nuclear-encoded electron transport chain genes. TEAD4's functions including oncogenic activities are tightly controlled by its subcellular localization. As a predominantly nuclear protein, its cytoplasmic translocation is triggered by several signals, such as osmotic stress, cell confluency, and arginine availability. Intriguingly, TEAD4 is also localized in mitochondria, although the translocation mechanism remains unclear. In this report, we describe the current understanding of TEAD4 as an oncogene, epigenetic regulator and mitochondrial modulator. The contributing mechanisms will be discussed.

14.
Biochem Biophys Res Commun ; 404(3): 865-9, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21184736

ABSTRACT

Protein arginine methylation plays crucial roles in numerous cellular processes. Heterogeneous nuclear ribonucleoprotein K (hnRNP K) is a multi-functional protein participating in a variety of cellular functions including transcription and RNA processing. HnRNP K is methylated at multiple sites in the glycine- and arginine-rich (RGG) motif. Using various RGG domain deletion mutants of hnRNP K as substrates, here we show by direct methylation assay that protein arginine methyltransferase 1 (PRMT1) methylated preferentially in a.a. 280-307 of the RGG motif. Kinetic analysis revealed that deletion of a.a. 280-307, but not a.a. 308-327, significantly inhibited rate of methylation. Importantly, nuclear localization of hnRNP K was significantly impaired in mutant hnRNP K lacking the PRMT1 methylation region or upon pharmacological inhibition of methylation. Together our results identify preferred PRMT1 methylation sequences of hnRNP K by direct methylation assay and implicate a role of arginine methylation in regulating intracellular distribution of hnRNP K.


Subject(s)
Arginine/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , HEK293 Cells , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Humans , Methylation , Transfection
15.
Biochem Biophys Res Commun ; 404(1): 68-73, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21094134

ABSTRACT

Alteration of epidermal growth factor receptor (EGFR) is involved in various human cancers and has been intensively investigated. A plethora of evidence demonstrates that posttranslational modifications of EGFR play a pivotal role in controlling its function and metabolism. Here, we show that EGFR can be acetylated by CREB binding protein (CBP) acetyltransferase. Interestingly, EGFR acetylation affects its tyrosine phosphorylation, which may contribute to cancer cell resistance to histone deacetylase inhibitors (HDACIs). Since there is an increasing interest in using HDACIs to treat various cancers in the clinic, our current study provides insights and rationale for selecting effective therapeutic regimen. Consistent with the previous reports, we also show that HDACI combined with EGFR inhibitors achieves better therapeutic outcomes and provides a molecular rationale for the enhanced effect of combination therapy. Our results unveil a critical role of EGFR acetylation that regulates EGFR function, which may have an important clinical implication.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/enzymology , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Acetylation/drug effects , Animals , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Lysine/genetics , Lysine/metabolism , Mice , Mice, Inbred Strains , RNA, Small Interfering/genetics , Vorinostat
16.
Cancers (Basel) ; 13(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34298755

ABSTRACT

Arginine is an amino acid critically involved in multiple cellular processes including the syntheses of nitric oxide and polyamines, and is a direct activator of mTOR, a nutrient-sensing kinase strongly implicated in carcinogenesis. Yet, it is also considered as a non- or semi-essential amino acid, due to normal cells' intrinsic ability to synthesize arginine from citrulline and aspartate via ASS1 (argininosuccinate synthase 1) and ASL (argininosuccinate lyase). As such, arginine can be used as a dietary supplement and its depletion as a therapeutic strategy. Strikingly, in over 70% of tumors, ASS1 transcription is suppressed, rendering the cells addicted to external arginine, forming the basis of arginine-deprivation therapy. In this review, we will discuss arginine as a signaling metabolite, arginine's role in cancer metabolism, arginine as an epigenetic regulator, arginine as an immunomodulator, and arginine as a therapeutic target. We will also provide a comprehensive summary of ADI (arginine deiminase)-based arginine-deprivation preclinical studies and an update of clinical trials for ADI and arginase. The different cell killing mechanisms associated with various cancer types will also be described.

17.
Polymers (Basel) ; 13(2)2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33451036

ABSTRACT

In this present work, an electrochemical sensor was developed for the sensing of uric acid (UA). The sensor was based on a carbon paste electrode (CPE) modified with electroactive polyimide (EPI) synthesized using aniline tetramer (ACAT) decorated with reduced nanoparticles (NPs) of Au, Pt, and Ag. The initial step involved the preparation and characterization of ACAT. Subsequently, the ACAT-based EPI synthesis was performed by chemical imidization of its precursors 4,4'-(4.4'-isopropylidene-diphenoxy) bis (phthalic anhydride) BPADA and ACAT. Then, EPI was doped with distinctive particles of Ag, Pt and Au, and the doped EPIs were abbreviated as EPIS, EPIP and EPIG, respectively. Their structures were characterized by XRD, XPS, and TEM, and the electrochemical properties were determined by cyclic voltammetry and chronoamperometry. Among these evaluated sensors, EPI with Au NPs turned out the best with a sensitivity of 1.53 uA uM-1 UA, a low limit of detection (LOD) of 0.78 uM, and a linear detection range (LDR) of 5-50 uM UA at a low potential value of 310 mV. Additionally, differential pulse voltammetric (DPV) analysis showed that the EPIG sensor showed the best selectivity for a tertiary mixture of UA, dopamine (DA), and ascorbic acid (AA) as compared to EPIP and EPIS.

18.
Nat Commun ; 12(1): 2398, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893278

ABSTRACT

Arginine plays diverse roles in cellular physiology. As a semi-essential amino acid, arginine deprivation has been used to target cancers with arginine synthesis deficiency. Arginine-deprived cancer cells exhibit mitochondrial dysfunction, transcriptional reprogramming and eventual cell death. In this study, we show in prostate cancer cells that arginine acts as an epigenetic regulator to modulate histone acetylation, leading to global upregulation of nuclear-encoded oxidative phosphorylation (OXPHOS) genes. TEAD4 is retained in the nucleus by arginine, enhancing its recruitment to the promoter/enhancer regions of OXPHOS genes and mediating coordinated upregulation in a YAP1-independent but mTOR-dependent manner. Arginine also activates the expression of lysine acetyl-transferases and increases overall levels of acetylated histones and acetyl-CoA, facilitating TEAD4 recruitment. Silencing of TEAD4 suppresses OXPHOS functions and prostate cancer cell growth in vitro and in vivo. Given the strong correlation of TEAD4 expression and prostate carcinogenesis, targeting TEAD4 may be beneficially used to enhance arginine-deprivation therapy and prostate cancer therapy.


Subject(s)
Arginine/pharmacology , DNA-Binding Proteins/genetics , Epigenesis, Genetic/drug effects , Epigenomics/methods , Gene Expression Regulation, Neoplastic/drug effects , Muscle Proteins/genetics , Oxidative Phosphorylation/drug effects , Prostatic Neoplasms/genetics , Transcription Factors/genetics , Animals , Arginine/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Humans , Male , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Muscle Proteins/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , TEA Domain Transcription Factors , Transcription Factors/metabolism
19.
Theranostics ; 11(15): 7527-7545, 2021.
Article in English | MEDLINE | ID: mdl-34158865

ABSTRACT

Rationale: One of the most common metabolic defects in cancers is the deficiency in arginine synthesis, which has been exploited therapeutically. Yet, challenges remain, and the mechanisms of arginine-starvation induced killing are largely unclear. Here, we sought to demonstrate the underlying mechanisms by which arginine starvation-induced cell death and to develop a dietary arginine-restriction xenograft model to study the in vivo effects. Methods: Multiple castration-resistant prostate cancer cell lines were treated with arginine starvation followed by comprehensive analysis of microarray, RNA-seq and ChIP-seq were to identify the molecular and epigenetic pathways affected by arginine starvation. Metabolomics and Seahorse Flux analyses were used to determine the metabolic profiles. A dietary arginine-restriction xenograft mouse model was developed to assess the effects of arginine starvation on tumor growth and inflammatory responses. Results: We showed that arginine starvation coordinately and epigenetically suppressed gene expressions, including those involved in oxidative phosphorylation and DNA repair, resulting in DNA damage, chromatin-leakage and cGAS-STING activation, accompanied by the upregulation of type I interferon response. We further demonstrated that arginine starvation-caused depletion of α-ketoglutarate and inactivation of histone demethylases are the underlying causes of epigenetic silencing. Significantly, our dietary arginine-restriction model showed that arginine starvation suppressed prostate cancer growth in vivo, with evidence of enhanced interferon responses and recruitment of immune cells. Conclusions: Arginine-starvation induces tumor cell killing by metabolite depletion and epigenetic silencing of metabolic genes, leading to DNA damage and chromatin leakage. The resulting cGAS-STING activation may further enhance these killing effects.


Subject(s)
Arginine/deficiency , Chromatin/metabolism , DNA Repair , Gene Expression Regulation, Neoplastic , Gene Silencing , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Nucleotidyltransferases/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Chromatin/genetics , Chromatin/pathology , Humans , Male , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Nucleotidyltransferases/genetics , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology
20.
Cancer Res ; 67(19): 9066-76, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17909010

ABSTRACT

Aberrant epidermal growth factor receptor (EGFR) signaling is a major cause of tumor progression and metastasis; the underlying mechanisms, however, are not well understood. In particular, it remains elusive whether deregulated EGFR pathway is involved in epithelial-mesenchymal transition (EMT), an early event that occurs during metastasis of cancers of an epithelial origin. Here, we show that EGF induces EGFR-expressing cancer cells to undergo a transition from the epithelial to the spindle-like mesenchymal morphology. EGF reduced E-cadherin expression and increased that of mesenchymal proteins. In search of a downstream mediator that may account for EGF-induced EMT, we focused on transcription repressors of E-cadherin, TWIST, SLUG, and Snail and found that cancer cells express high levels of TWIST and that EGF enhances its expression. EGF significantly increases TWIST transcripts and protein in EGFR-expressing lines. Forced expression of EGFR reactivates TWIST expression in EGFR-null cells. TWIST expression is suppressed by EGFR and Janus-activated kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) inhibitors, but not significantly by those targeting phosphoinositide-3 kinase and MEK/ERK. Furthermore, constitutively active STAT3 significantly activates the TWIST promoter, whereas the JAK/STAT3 inhibitor and dominant-negative STAT3 suppressed TWIST promoter. Deletion/mutation studies further show that a 26-bp promoter region contains putative STAT3 elements required for the EGF-responsiveness of the TWIST promoter. Chromatin immunoprecipitation assays further show that EGF induces binding of nuclear STAT3 to the TWIST promoter. Immunohistochemical analysis of 130 primary breast carcinomas indicates positive correlations between non-nuclear EGFR and TWIST and between phosphorylated STAT3 and TWIST. Together, we report here that EGF/EGFR signaling pathways induce cancer cell EMT via STAT3-mediated TWIST gene expression.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Squamous Cell/pathology , ErbB Receptors/metabolism , STAT3 Transcription Factor/metabolism , Twist-Related Protein 1/biosynthesis , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CHO Cells , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cricetinae , Cricetulus , Dogs , Epidermal Growth Factor/pharmacology , Epithelial Cells/pathology , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Mesoderm/pathology , Mice , Molecular Sequence Data , Promoter Regions, Genetic , STAT3 Transcription Factor/genetics , Swiss 3T3 Cells , Twist-Related Protein 1/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL