Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(6): 112557, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37224019

ABSTRACT

Despite its pivotal roles in biology, how the transcriptional activity of c-MYC is tuned quantitatively remains poorly defined. Here, we show that heat shock factor 1 (HSF1), the master transcriptional regulator of the heat shock response, acts as a prime modifier of the c-MYC-mediated transcription. HSF1 deficiency diminishes c-MYC DNA binding and dampens its transcriptional activity genome wide. Mechanistically, c-MYC, MAX, and HSF1 assemble into a transcription factor complex on genomic DNAs, and surprisingly, the DNA binding of HSF1 is dispensable. Instead, HSF1 physically recruits the histone acetyltransferase general control nonderepressible 5 (GCN5), promoting histone acetylation and augmenting c-MYC transcriptional activity. Thus, we find that HSF1 specifically potentiates the c-MYC-mediated transcription, discrete from its canonical role in countering proteotoxic stress. Importantly, this mechanism of action engenders two distinct c-MYC activation states, primary and advanced, which may be important to accommodate diverse physiological and pathological conditions.


Subject(s)
DNA-Binding Proteins , Heat-Shock Response , Transcription Factors , DNA , DNA-Binding Proteins/metabolism , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Heat-Shock Response/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Humans , Cell Line, Tumor
2.
Front Cardiovasc Med ; 10: 1155444, 2023.
Article in English | MEDLINE | ID: mdl-37077734

ABSTRACT

Atherosclerosis is a major risk factor for cardiovascular diseases. Hypercholesterolemia has been both clinically and experimentally linked to cardiovascular disease and is involved in the initiation of atherosclerosis. Heat shock factor 1 (HSF1) is involved in the control of atherosclerosis. HSF1 is a critical transcriptional factor of the proteotoxic stress response that regulates the production of heat shock proteins (HSPs) and other important activities such as lipid metabolism. Recently, HSF1 is reported to directly interact with and inhibit AMP-activated protein kinase (AMPK) to promote lipogenesis and cholesterol synthesis. This review highlights roles of HSF1 and HSPs in critical metabolic pathways of atherosclerosis, including lipogenesis and proteome homeostasis.

3.
Sci Adv ; 6(46)2020 11.
Article in English | MEDLINE | ID: mdl-33177089

ABSTRACT

The role of proteomic instability in cancer, particularly amyloidogenesis, remains obscure. Heat shock factor 1 (HSF1) transcriptionally governs the proteotoxic stress response to suppress proteomic instability and enhance survival. Paradoxically, HSF1 promotes oncogenesis. Here, we report that AKT activates HSF1 via Ser230 phosphorylation. In vivo, HSF1 enables megalencephaly and hepatomegaly, which are driven by hyperactive phosphatidylinositol 3-kinase/AKT signaling. Hsf1 deficiency exacerbates amyloidogenesis and elicits apoptosis, thereby countering tissue overgrowth. Unexpectedly, HSF1 physically neutralizes soluble amyloid oligomers (AOs). Beyond impeding amyloidogenesis, HSF1 shields HSP60 from direct assault by AOs, averting HSP60 destabilization, collapse of the mitochondrial proteome, and, ultimately, mitophagy and apoptosis. The very same mechanism occurs in Alzheimer's disease. These findings suggest that amyloidogenesis may be a checkpoint mechanism that constrains uncontrolled growth and safeguards tissue homeostasis, congruent with its emerging tumor-suppressive function. HSF1, by acting as an anti-amyloid factor, promotes overgrowth syndromes and cancer but may suppress neurodegenerative disorders.

4.
Mol Cell ; 76(4): 546-561.e8, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31561952

ABSTRACT

Through transcriptional control of the evolutionarily conserved heat shock, or proteotoxic stress, response, heat shock factor 1 (HSF1) preserves proteomic stability. Here, we show that HSF1, a physiological substrate for AMP-activated protein kinase (AMPK), constitutively suppresses this central metabolic sensor. By physically evoking conformational switching of AMPK, HSF1 impairs AMP binding to the γ subunits and enhances the PP2A-mediated de-phosphorylation, but it impedes the LKB1-mediated phosphorylation of Thr172, and retards ATP binding to the catalytic α subunits. These immediate and manifold regulations empower HSF1 to both repress AMPK under basal conditions and restrain its activation by diverse stimuli, thereby promoting lipogenesis, cholesterol synthesis, and protein cholesteroylation. In vivo, HSF1 antagonizes AMPK to control body fat mass and drive the lipogenic phenotype and growth of melanomas independently of its intrinsic transcriptional action. Thus, the physical AMPK-HSF1 interaction epitomizes a reciprocal kinase-substrate regulation whereby lipid metabolism and proteomic stability intertwine.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Energy Metabolism , Heat Shock Transcription Factors/metabolism , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Adiposity , Animals , Binding Sites , Cell Proliferation , Cholesterol/biosynthesis , HEK293 Cells , HeLa Cells , Heat Shock Transcription Factors/deficiency , Heat Shock Transcription Factors/genetics , Humans , Lipogenesis , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Phosphorylation , Protein Conformation , Protein Stability , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Structure-Activity Relationship
5.
Bioessays ; 39(5)2017 05.
Article in English | MEDLINE | ID: mdl-28295473

ABSTRACT

Beyond protein synthesis and autophagy, emerging evidence has implicated mTORC1 in regulating protein folding and proteasomal degradation as well, highlighting its prominent role in cellular proteome homeostasis or proteostasis. In addition to growth signals, mTORC1 senses and responds to a wide array of stresses, including energetic/metabolic stress, genotoxic stress, oxidative stress, osmotic stress, ER stress, proteotoxic stress, and psychological stress. Whereas growth signals unanimously stimulate mTORC1, stresses exert complex impacts on mTORC1, most of which are repressive. mTORC1 suppression, as a generic adaptive strategy, empowers cell survival under various stressful conditions. In this essay, we provide an overview of the emerging role of mTORC1 in proteostasis, the distinct molecular mechanisms through which mTORC1 reacts to diverse stresses, and the schemes exploited by cancer cells to circumvent stress-induced mTORC1 suppression. Hence, acting as a stress sensor, mTORC1 intimately couples stresses to cellular proteostasis.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Proteostasis , Stress, Physiological , Animals , Carcinogenesis , Endoplasmic Reticulum Stress , Humans , Models, Biological , Neoplasms/metabolism , Osmotic Pressure , Oxidative Stress , Stress, Psychological
6.
Cell Cycle ; 15(23): 3155-3156, 2016 12.
Article in English | MEDLINE | ID: mdl-27589382
7.
Cell Mol Life Sci ; 73(22): 4231-4248, 2016 11.
Article in English | MEDLINE | ID: mdl-27289378

ABSTRACT

Proteome homeostasis, or proteostasis, is essential to maintain cellular fitness and its disturbance is associated with a broad range of human health conditions and diseases. Cells are constantly challenged by various extrinsic and intrinsic insults, which perturb cellular proteostasis and provoke proteotoxic stress. To counter proteomic perturbations and preserve proteostasis, cells mobilize the proteotoxic stress response (PSR), an evolutionarily conserved transcriptional program mediated by heat shock factor 1 (HSF1). The HSF1-mediated PSR guards the proteome against misfolding and aggregation. In addition to proteotoxic stress, emerging studies reveal that this proteostatic mechanism also responds to cellular energy state. This regulation is mediated by the key cellular metabolic sensor AMP-activated protein kinase (AMPK). In this review, we present an overview of the maintenance of proteostasis by HSF1, the metabolic regulation of the PSR, particularly focusing on AMPK, and their implications in the two major age-related diseases-diabetes mellitus and neurodegenerative disorders.


Subject(s)
Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Proteins/toxicity , Stress, Physiological/drug effects , Animals , Heat-Shock Proteins/metabolism , Humans , Models, Biological
8.
Nat Cell Biol ; 18(5): 527-39, 2016 05.
Article in English | MEDLINE | ID: mdl-27043084

ABSTRACT

To cope with proteotoxic stress, cells attenuate protein synthesis. However, the precise mechanisms underlying this fundamental adaptation remain poorly defined. Here we report that mTORC1 acts as an immediate cellular sensor of proteotoxic stress. Surprisingly, the multifaceted stress-responsive kinase JNK constitutively associates with mTORC1 under normal growth conditions. On activation by proteotoxic stress, JNK phosphorylates both RAPTOR at S863 and mTOR at S567, causing partial disintegration of mTORC1 and subsequent translation inhibition. Importantly, HSF1, the central player in the proteotoxic stress response (PSR), preserves mTORC1 integrity and function by inactivating JNK, independently of its canonical transcriptional action. Thereby, HSF1 translationally augments the PSR. Beyond promoting stress resistance, this intricate HSF1-JNK-mTORC1 interplay, strikingly, regulates cell, organ and body sizes. Thus, these results illuminate a unifying mechanism that controls stress adaptation and growth.


Subject(s)
DNA-Binding Proteins/metabolism , Heat-Shock Response , Multiprotein Complexes/metabolism , Proteins/toxicity , Stress, Physiological/drug effects , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Body Size/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Enzyme Activation/drug effects , HEK293 Cells , HeLa Cells , Heat Shock Transcription Factors , Heat-Shock Response/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/cytology , Liver/drug effects , Liver/growth & development , MAP Kinase Signaling System/drug effects , Mechanistic Target of Rapamycin Complex 1 , Mice , Organ Size/drug effects , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Transcription, Genetic/drug effects
9.
Mol Nutr Food Res ; 59(4): 646-57, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25581901

ABSTRACT

SCOPE: Epigallocatechin-3-gallate (EGCG), the most abundant catechin of green tea, has beneficial effects on physiological functions of endothelial cells (ECs), yet the detailed mechanisms are not fully understood. In this study, we investigated the role of transient receptor potential vanilloid type 1 (TRPV1), a ligand-gated nonselective calcium channel, in EGCG-mediated endothelial nitric oxide (NO) synthase (eNOS) activation and angiogenesis. METHODS AND RESULTS: In ECs, treatment with EGCG time-dependently increased the intracellular level of Ca(2+) . Removal of extracellular calcium (Ca(2+) ) by EGTA or EDTA or inhibition of TRPV1 by capsazepine or SB366791 abrogated EGCG-increased intracellular Ca(2+) level in ECs or TRPV1-transfected HEK293 cells. Additionally, EGCG increased the phsophorylation of eNOS at Ser635 and Ser1179, Akt at Ser473, calmodulin-dependent protein kinase II (CaMKII) at Thr286 and AMP-activated protein kinase (AMPK) at Thr172, all abolished by the TRPV1 antagonist capsazepine. EGCG-induced NO production was diminished by pretreatment with LY294002 (an Akt inhibitor), KN62 (a CaMKII inhibitor), and compound C (an AMPK inhibitor). Moreover, blocking TRPV1 activation prevented EGCG-induced EC proliferation, migration, and tube formation, as well as angiogenesis in Matrigel plugs in mice. CONCLUSION: EGCG may trigger activation of TRPV1-Ca(2+) signaling, which leads to phosphorylation of Akt, AMPK, and CaMKII; eNOS activation; NO production; and, ultimately, angiogenesis in ECs.


Subject(s)
Catechin/analogs & derivatives , Nitric Oxide Synthase Type III/metabolism , TRPV Cation Channels/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Anilides/pharmacology , Animals , Calcium/metabolism , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Catechin/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cinnamates/pharmacology , Endothelial Cells/drug effects , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Phosphorylation , Signal Transduction , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Tea/chemistry
10.
Int J Biol Sci ; 10(9): 990-6, 2014.
Article in English | MEDLINE | ID: mdl-25210497

ABSTRACT

14,15-epoxyeicosatrienoic acid (14,15-EET) is implicated in regulating physiological functions of endothelial cells (ECs), yet the potential molecular mechanisms underlying the beneficial effects in ECs are not fully understood. In this study, we investigated whether transient receptor potential vanilloid receptor type 1 (TRPV1) is involved in 14,15-EET-mediated Ca(2+) influx, nitric oxide (NO) production and angiogenesis. In human microvascular endothelial cells (HMECs), 14,15-EET time-dependently increased the intracellular level of Ca(2+). Removal of extracellular Ca(2+), pharmacological inhibition or genetic disruption of TRPV1 abrogated 14,15-EET-mediated increase of intracellular Ca(2+) level in HMECs or TRPV1-transfected HEK293 cells. Furthermore, removal of extracellular Ca(2+) or pharmacological inhibition of TRPV1 decreased 14,15-EET-induced NO production. 14,15-EET-mediated tube formation was abolished by TRPV1 pharmacological inhibition. In an animal experiment, 14,15-EET-induced angiogenesis was diminished by inhibition of TRPV1 and in TRPV1-deficient mice. TRPV1 may play a crucial role in 14,15-EET-induced Ca(2+) influx, NO production and angiogenesis.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Endothelial Cells/physiology , Neovascularization, Physiologic/drug effects , TRPV Cation Channels/metabolism , Vasodilator Agents/pharmacology , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Calcium/metabolism , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide , Signal Transduction , TRPV Cation Channels/genetics
11.
J Gastroenterol Hepatol ; 29(3): 494-501, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24219143

ABSTRACT

BACKGROUND AND AIM: Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders with unclear etiology and mechanism(s). Glycine N-methyltransferase (GNMT) plays a central role in inflammatory diseases such as hepatitis and atherosclerosis. However, little is known about the impact of GNMT and the involved mechanism in the pathogenesis of IBD. In the current study, we investigated the role of GNMT in the mouse model of dextran sulfate sodium (DSS)-induced colitis. METHODS: Protein expression was determined by Western blotting or immunohistochemistry. Histopathology was examined by hematoxylin and eosin staining. Levels of pro-inflammatory cytokines were evaluated by ELISA kits. RESULTS: GNMT was expressed in the epithelium of the colon under normal conditions, and with DSS treatment, its expression was predominant in infiltrated leukocytes of lesions. Mice with genetic deletion of GNMT (GNMT(-/-) ) showed increased susceptibility to DSS induction of colitis, as revealed by the progression of colitis. Additionally, severe colonic inflammation, including increased crypt loss, leukocyte infiltration, and hemorrhage, was greater with DSS treatment in GNMT(-/-) than wild-type mice. Furthermore, the expression of adhesion molecule and inflammatory mediators in the colon was significantly higher with DSS treatment in GNMT(-/-) than wild-type mice. Moreover, loss of GNMT decreased cell apoptosis in colitis lesions with DSS treatment. CONCLUSIONS: Collectively, our findings suggest that GNMT may be a crucial molecule in the pathogenesis of DSS-induced colitis. This finding may provide new information for a potential therapeutic target in treating IBD.


Subject(s)
Colitis, Ulcerative/genetics , Glycine N-Methyltransferase/genetics , Glycine N-Methyltransferase/physiology , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Dextran Sulfate , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Expression , Glycine N-Methyltransferase/metabolism , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy
12.
Am J Chin Med ; 41(5): 1079-96, 2013.
Article in English | MEDLINE | ID: mdl-24117070

ABSTRACT

Paeonol, a phenolic component purified from Paeonia suffruticosa (Cortex Moutan), is used in traditional Chinese medicine to treat inflammatory diseases. However, little is known about the effect of paeonol on cholesterol metabolism. We investigated the efficacy of paeonol on cholesterol metabolism and the underlying mechanism in macrophages and apolipoprotein E deficient (apoE(-/-)) mice. Treatment with paeonol markedly attenuated cholesterol accumulation induced by oxidized LDL in macrophages, which was due to increased cholesterol efflux. Additionally, paeonol enhanced the mRNA and protein expression of ATP-binding membrane cassette transport protein A1 (ABCA1) but did not alter the protein level of ABCG1 or other scavenger receptors. Inhibition of ABCA1 activity with a pharmacological inhibitor, neutralizing antibody or small interfering RNA (siRNA), negated the effects of paeonol on cholesterol efflux and cholesterol accumulation. Furthermore, paeonol induced the nuclear translocation of liver X receptor α (LXRα) by increasing its activity. siRNA knockdown of LXRα abolished the paeonol-induced upregulation of ABCA1, promotion of cholesterol efflux and suppression of cholesterol accumulation. Moreover, atherosclerotic lesions, hyperlipidemia and systemic inflammation were reduced and the protein expression of ABCA1 was increased in aortas of paeonol-treated apoE(-/-) mice. Paeonol may alleviate the formation of foam cells by enhancing LXRα-ABCA1-dependent cholesterol efflux.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Acetophenones/pharmacology , Cholesterol/metabolism , Foam Cells/metabolism , Gene Expression/drug effects , Orphan Nuclear Receptors/metabolism , Up-Regulation/drug effects , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/physiology , Acetophenones/therapeutic use , Animals , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Cell Culture Techniques , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Lipoproteins, LDL/metabolism , Liver X Receptors , Male , Mice , Molecular Targeted Therapy , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/physiology , Phytotherapy , RNA, Small Interfering
13.
Mol Med ; 18: 805-15, 2012.
Article in English | MEDLINE | ID: mdl-22451268

ABSTRACT

We investigated whether AMP-activated protein kinase (AMPK), a multi-functional regulator of energy homeostasis, is involved in transient receptor potential vanilloid type 1 (TRPV1)-mediated activation of endothelial nitric oxide synthase (eNOS) in endothelial cells (ECs) and mice. In ECs, treatment with evodiamine, the activator of TRPV1, increased the phosphorylation of AMPK, acetyl-CoA carboxylase (ACC) and eNOS, as revealed by western blot analysis. Inhibition of AMPK activation by compound C or dominant-negative AMPK mutant abrogated the evodiamine-induced increase in phosphorylation of AMPK and eNOS and NO bioavailability, as well as tube formation in ECs. Immunoprecipitation and two-hybrid analysis demonstrated that AMPK mediated the evodiamine-induced increase in the formation of a TRPV1-eNOS complex. Additionally, TRPV1 activation by evodiamine increased the phosphorylation of AMPK and eNOS in aortas of wild-type mice but did not activate eNOS in aortas of TRPV1-deficient mice. In mice, inhibition of AMPK activation by compound C markedly decreased evodiamine-evoked angiogenesis in Matrigel plugs and in a hind-limb ischemia model. Moreover, evodiamine-induced phosphorylation of AMPK and eNOS in aortas of apolipoprotein E deficient (ApoE(-/-)) mice was abrogated in TRPV1-deficient ApoE(-/-) mice. In conclusion, TRPV1 activation may trigger AMPK-dependent signaling, which leads to enhanced activation of AMPK and eNOS and retarded development of atherosclerosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aorta/metabolism , Nitric Oxide Synthase Type III/metabolism , TRPV Cation Channels/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Aorta/cytology , Blood Vessels/drug effects , Blotting, Western , Cattle , Cells, Cultured , Collagen , Drug Combinations , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Hindlimb/blood supply , Hindlimb/metabolism , Injections, Intraperitoneal , Laminin , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Phosphorylation/drug effects , Protein Binding , Proteoglycans , Quinazolines/administration & dosage , Quinazolines/pharmacology , TRPV Cation Channels/genetics
14.
J Cell Physiol ; 227(8): 3053-62, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22021095

ABSTRACT

We investigated whether AMP-activated protein kinase (AMPK), a multi-functional regulator of energy homeostasis, participates in the regulation of erythropoietin (EPO)-mediated activation of endothelial nitric oxide synthase (eNOS) in endothelial cells (ECs) and mice. In ECs, treatment with EPO increased the phosphorylation of AMPK, acetyl-CoA carboxylase (ACC), and eNOS, as revealed by Western blot analysis. Inhibition of AMPK activation by compound C or dominant-negative AMPK mutant abrogated the EPO-induced increase in the phosphorylation of AMPK, ACC, and eNOS, as well as nitric oxide (NO) production. Additionally, suppression of AMPK activation abolished EPO-induced EC proliferation, migration and tube formation. Immunoprecipitation analysis demonstrated that AMPK mediated the EPO-induced increase in the phosphorylation of ß common receptor (ßCR) and the formation of a ßCR-AMPK-eNOS complex. In mice, inhibition of AMPK activation by compound C markedly decreased EPO-elicited angiogenesis in Matrigel plugs. Furthermore, the phosphorylation of AMPK and eNOS was significantly higher in aortas from EPO transgenic mice than wild-type mice. Moreover, treatment with EPO neutralizing antibody greatly reduced the exercise training-induced increase in phosphorylation of AMPK and eNOS in aortas of wild-type mice. Taken together, EPO may trigger AMPK-dependent signaling, which leads to enhanced phosphorylation of ßCR and eNOS, increased ßCR-AMPK-eNOS complex formation, NO production, and, ultimately, angiogenesis.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Erythropoietin/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelium/cytology , Endothelium/metabolism , Erythropoietin/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction
15.
Cardiovasc Res ; 91(3): 492-501, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21493704

ABSTRACT

AIMS: We investigated the molecular mechanism underlying the role of transient receptor potential vanilloid type 1 (TRPV1), a Ca(2+)-permeable non-selective cation channel, in the activation of endothelial nitric oxide (NO) synthase (eNOS) in endothelial cells (ECs) and mice. METHODS AND RESULTS: In ECs, TRPV1 ligands (evodiamine or capsaicin) promoted NO production, eNOS phosphorylation, and the formation of a TRPV1-eNOS complex, which were all abrogated by the TRPV1 antagonist capsazepine. TRPV1 ligands promoted the phosphorylation of Akt, calmodulin-dependent protein kinase II (CaMKII) and TRPV1, and increased the formation of a TRPV1-Akt-CaMKII complex. Removal of extracellular Ca(2+) abolished the ligand-induced increase in the phosphorylation of Akt and CaMKII, formation of a TRPV1-eNOS complex, and eNOS activation. Inhibition of PI3K and CaMKII suppressed the ligand-induced increase in TRPV1 phosphorylation, formation of a TRPV1-eNOS complex, and eNOS activation. TRPV1 activation increased the phosphorylation of Akt, CaMKII, and eNOS in the aortas of wild-type mice but failed to activate eNOS in TRPV1-deficient aortas. Additionally, TRPV1 ligand-induced angiogenesis was diminished in eNOS- or TRPV1-deficient mice. When compared with apolipoprotein E (ApoE)-deficient mice, ApoE/TRPV1-double-knockout mice displayed reduced phosphorylation of eNOS, Akt, and CaMKII in aortas but worsened atherosclerotic lesions. CONCLUSION: TRPV1 activation in ECs may trigger Ca(2+)-dependent PI3K/Akt/CaMKII signalling, which leads to enhanced phosphorylation of TRPV1, increased TRPV1-eNOS complex formation, eNOS activation and, ultimately, NO production.


Subject(s)
Endothelial Cells/enzymology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , TRPV Cation Channels/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cattle , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Enzyme Activation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , RNA Interference , Signal Transduction , TRPV Cation Channels/deficiency , TRPV Cation Channels/drug effects , TRPV Cation Channels/genetics , Time Factors , Transfection
16.
J Cell Physiol ; 226(12): 3330-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21321940

ABSTRACT

Erythropoietin (EPO), the key hormone for erythropoiesis, also increases nitric oxide (NO) bioavailability in endothelial cells (ECs), yet the definitive mechanisms are not fully understood. Increasing evidence has demonstrated that ß common receptor (ßCR) plays a crucial role in EPO-mediated non-hematopoietic effects. We investigated the role of ßCR in EPO-induced endothelial NO synthase (eNOS) activation in bovine aortic ECs (BAECs) and the molecular mechanisms involved. Results of confocal microscopy and immunoprecipitation analyses revealed that ßCR was colocalized and interacted with EPO receptor (EPOR) in ECs. Inhibition of ßCR or EPOR by neutralizing antibodies or small interfering RNA abolished the EPO-induced NO production. Additionally, blockage of ßCR abrogated the EPO-induced increase in the phosphorylation of eNOS, Akt, Src, or Janus kinase 2 (JAK2). Immunoprecipitation analysis revealed that treatment with EPO increased the interaction between ßCR and eNOS, which was suppressed by inhibition of Src, JAK2, or Akt signaling with specific pharmacological inhibitors. Furthermore, EPO-induced EC proliferation, migration, and tube formation were blocked by pretreatment with ßCR antibody and Src, JAK2, or PI3K/Akt inhibitors. Moreover, in vivo experiments showed that EPO increased the level of phosphorylated eNOS, Src, JAK2, and Akt, as well as ßCR-eNOS association in aortas and promoted the angiogenesis in Matrigel plug, which was diminished by ßCR or EPOR neutralizing antibodies. Our findings suggest that ßCR may play an integrative role in the EPO signaling-mediated activation of eNOS in ECs.


Subject(s)
Cytokine Receptor Common beta Subunit/metabolism , Endothelial Cells/enzymology , Erythropoietin/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptors, Interleukin-3/metabolism , Signal Transduction , Animals , Antibodies, Neutralizing/pharmacology , Cattle , Cell Movement , Cell Proliferation , Cells, Cultured , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/immunology , Endothelial Cells/drug effects , Enzyme Activation , Erythropoietin/genetics , Humans , Immunoprecipitation , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Neovascularization, Physiologic , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/immunology , Receptors, Erythropoietin/metabolism , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Recombinant Proteins , Signal Transduction/drug effects , Time Factors , Transfection , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
17.
Free Radic Biol Med ; 50(1): 47-54, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21034810

ABSTRACT

α-Lipoic acid (α-LA), a key cofactor in cellular energy metabolism, has protective activities in atherosclerosis, yet the detailed mechanisms are not fully understood. In this study, we examined whether α-LA affects foam cell formation and its underlying molecular mechanisms in murine macrophages. Treatment with α-LA markedly attenuated oxidized low-density lipoprotein (oxLDL)-mediated cholesterol accumulation in macrophages, which was due to increased cholesterol efflux. Additionally, α-LA treatment dose-dependently increased protein levels of ATP-binding cassette transporter A1 (ABCA1) and ABCG1 but had no effect on the protein expression of SR-A, CD36, or SR-BI involved in cholesterol homeostasis. Furthermore, α-LA increased the mRNA expression of ABCA1 and ABCG1. The upregulation of ABCA1 and ABCG1 by α-LA depended on liver X receptor α (LXRα), as evidenced by an increase in the nuclear levels of LXRα and LXRE-mediated luciferase activity and its prevention of the expression of ABCA1 and ABCG1 after inhibition of LXRα activity by the pharmacological inhibitor geranylgeranyl pyrophosphate (GGPP) or knockdown of LXRα expression with small interfering RNA (siRNA). Consistently, α-LA-mediated suppression of oxLDL-induced lipid accumulation was abolished by GGPP or LXRα siRNA treatment. In conclusion, LXRα-dependent upregulation of ABCA1 and ABCG1 may mediate the beneficial effect of α-LA on foam cell formation.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Foam Cells/drug effects , Lipoproteins/genetics , Orphan Nuclear Receptors/physiology , Thioctic Acid/pharmacology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Cells, Cultured , Foam Cells/metabolism , Foam Cells/pathology , Humans , Lipoproteins/metabolism , Lipoproteins, LDL/metabolism , Liver X Receptors , Mice , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/metabolism , Polyisoprenyl Phosphates/pharmacology , RNA, Small Interfering/pharmacology , Response Elements/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Transfection , Up-Regulation/drug effects
18.
J Nutr Biochem ; 22(11): 1015-21, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21190831

ABSTRACT

Wogonin, one component in Scutellaria baicalensis Georgi extracts, has several beneficial properties for cancers and inflammatory diseases. However, the efficacy of wogonin in cholesterol metabolism of macrophages remains unknown. In macrophages, cholesterol uptake is controlled by scavenger receptors (SR-A and CD36) and cholesterol efflux by SR-BI, ATP-binding cassette transporter-A1 (ABCA1) and ABCG1. In the present study, we investigated the effect and underlying molecular mechanism of wogonin on the formation of macrophage foam cells by murine J774.A1 macrophages. Wogonin attenuated oxidized low-density lipoprotein (oxLDL)-induced cholesterol accumulation in macrophages. The binding of oxLDL to macrophages and protein expression of SR-A and CD36 were not affected by wogonin. Wogonin enhanced cholesterol efflux and increased the protein level of ABCA1 without affecting the protein expression of SR-BI or ABCG1. Inhibition of ABCA1 by pharmacological inhibitor 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid disodium salt or neutralizing antibody abolished this suppressive effect of wogonin on lipid accumulation. Moreover, the up-regulation of ABCA1 protein by wogonin resulted from a decrease in degradation rate of ABCA1 protein, with no effect on ABCA1 mRNA expression. This reduction in ABCA1 degradation was due to increased protein phosphatase 2B (PP2B)-mediated ABCA1 dephosphorylation, as evidenced by increased interaction between ABCA1 and PP2B; pharmacological inhibition of PP2B would prevent wogonin-induced ABCA1 protein expression, dephosphorylation and attenuation of lipid accumulation. Collectively, wogonin increases the protein stability of ABCA1 via PP2B-mediated dephosphorylation, thus leading to reduced cholesterol accumulation in macrophage foam cells.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Calcineurin/metabolism , Cholesterol/metabolism , Flavanones/pharmacology , Macrophages/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Animals , Cells, Cultured , Foam Cells/drug effects , Lipoproteins/metabolism , Lipoproteins, LDL/metabolism , Macrophages/drug effects , Mice , Scavenger Receptors, Class A/metabolism , Scavenger Receptors, Class B/metabolism
19.
Cardiovasc Res ; 88(3): 415-23, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20615914

ABSTRACT

AIMS: Accumulation of foam cells in the intima is a hallmark of early-stage atherosclerotic lesions. Ginkgo biloba extract (EGb761) has been reported to exert anti-oxidative and anti-inflammatory properties in atherosclerosis, yet the significance and the molecular mechanisms of action of EGb761 in the formation of macrophage foam cells are not fully understood. METHODS AND RESULTS: Treatment with EGb761 resulted in a dose-dependent decrease in oxidized low-density lipoprotein (oxLDL)-mediated cholesterol accumulation in macrophages, a consequence that was due to a decrease in cholesterol uptake and an increase in cholesterol efflux. Additionally, EGb761 significantly down-regulated the mRNA and protein expression of class A scavenger receptor (SR-A) by decreasing expression of activator protein 1 (AP-1); however, EGb761 increased the protein stability of ATP-binding cassette transporter A1 (ABCA1) by reducing calpain activity without affecting ABCA1 mRNA expression. Small interfering RNA (siRNA) targeting haem oxygenase-1 (HO-1) abolished the EGb761-induced protective effects on the expression of AP-1, SR-A, ABCA1, and calpain activity. Accordingly, EGb761-mediated suppression of lipid accumulation in foam cells was also abrogated by HO-1 siRNA. Moreover, the lesion size of atherosclerosis was smaller in EGb761-treated, apolipoprotein E-deficient mice compared with the vehicle-treated mice, and the expression of HO-1, SR-A, and ABCA1 in aortas was modulated similar to that observed in macrophages. CONCLUSION: These findings suggest that EGb761 confers a protection from the formation of foam cells by a novel HO-1-dependent regulation of cholesterol homeostasis in macrophages.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Foam Cells/drug effects , Foam Cells/metabolism , Heme Oxygenase-1/metabolism , Plant Extracts/pharmacology , Scavenger Receptors, Class A/metabolism , ATP Binding Cassette Transporter 1 , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Calpain/metabolism , Cholesterol/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Foam Cells/pathology , Ginkgo biloba , Homeostasis/drug effects , Lipid Metabolism/drug effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout
20.
Circulation ; 121(16): 1828-37, 2010 Apr 27.
Article in English | MEDLINE | ID: mdl-20385932

ABSTRACT

BACKGROUND: In addition to the hematopoietic effect of erythropoietin, increasing evidence suggests that erythropoietin also exerts protective effects for cardiovascular diseases. However, the role of erythropoietin and its underlying mechanism in macrophage foam cell formation are poorly understood. METHODS AND RESULTS: Compared with wild-type specimens, erythropoietin was increased in atherosclerotic aortas of apolipoprotein E-deficient (apoE(-/-)) mice, mainly in the macrophage foam cells of the lesions. Erythropoietin levels in culture medium and macrophages were significantly elevated in response to oxidized low-density lipoprotein in a dose-dependent manner. Furthermore, erythropoietin markedly attenuated lipid accumulation in oxidized low-density lipoprotein-treated macrophages, a result that was due to an increase in cholesterol efflux. Erythropoietin treatment significantly increased ATP-binding cassette transporters (ABC) A1 and ABCG1 mRNA and protein levels without affecting protein expression of scavenger receptors, including scavenger receptor-A, CD36, and scavenger receptor-BI. The upregulation of ABCA1 and ABCG1 by erythropoietin resulted from liver X receptor alpha activation, which was confirmed by its prevention on expression of ABCA1 and ABCG1 after pharmacological or small interfering RNA inhibition of liver X receptor alpha. Moreover, the erythropoietin-mediated attenuation on lipid accumulation was abolished by such inhibition. Finally, reduced lipid accumulation and marked increase in ABCA1 and ABCG1 were demonstrated in erythropoietin-overexpressed macrophages. CONCLUSIONS: Our data suggest that erythropoietin suppresses foam cell formation via the liver X receptor alpha-dependent upregulation of ABCA1 and ABCG1.


Subject(s)
Atherosclerosis/drug therapy , Cardiotonic Agents/pharmacology , Erythropoietin/pharmacology , Foam Cells/drug effects , Orphan Nuclear Receptors/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Aortic Diseases/drug therapy , Aortic Diseases/immunology , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , CD36 Antigens/genetics , Cells, Cultured , Foam Cells/metabolism , Foam Cells/pathology , Lipids/biosynthesis , Lipoproteins/genetics , Lipoproteins/metabolism , Lipoproteins, LDL/pharmacology , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class B/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...