Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Article in English | MEDLINE | ID: mdl-34161259

ABSTRACT

The quantity and quality of food intake have been considered crucial for peoples' wellness. Only recently has it become appreciated that the timing of food intake is also critical. Nondipping blood pressure (BP) is prevalent in diabetic patients and is associated with increased cardiovascular events. However, the causes and mechanisms of nondipping BP in diabetes are not fully understood. Here, we report that food intake and BP were arrhythmic in diabetic db/db mice fed a normal chow diet ad libitum. Imposing a food intake diurnal rhythm by time-restricted feeding (TRF; food was only available for 8 h during the active phase) prevented db/db mice from developing nondipping BP and effectively restored the already disrupted BP circadian rhythm in db/db mice. Interestingly, increasing the time of food availability from 8 h to 12 h during the active dark phase in db/db mice prompted isocaloric feeding and still provided robust protection of the BP circadian rhythm in db/db mice. In contrast, neither 8-h nor 12-h TRF affected BP dipping in wild-type mice. Mechanistically, we demonstrate that TRF protects the BP circadian rhythm in db/db mice via suppressing the sympathetic activity during the light phase when they are inactive and fasting. Collectively, these data reveal a potentially pivotal role of the timing of food intake in the prevention and treatment of nondipping BP in diabetes.


Subject(s)
Blood Pressure/physiology , Circadian Rhythm/physiology , Diabetes Mellitus, Experimental/physiopathology , Fasting/physiology , Animals , Energy Intake , Mice , Sympathetic Nervous System/physiopathology , Time Factors
2.
Arterioscler Thromb Vasc Biol ; 38(5): 1063-1075, 2018 05.
Article in English | MEDLINE | ID: mdl-29437576

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm (AAA) has high mortality rate when ruptured, but currently, there is no proven pharmacological therapy for AAA because of our poor understanding of its pathogenesis. The current study explored a novel role of smooth muscle cell (SMC) BMAL1 (brain and muscle Arnt-like protein-1)-a transcription factor known to regulate circadian rhythm-in AAA development. APPROACH AND RESULTS: SMC-selective deletion of BMAL1 potently protected mice from AAA induced by (1) MR (mineralocorticoid receptor) agonist deoxycorticosterone acetate or aldosterone plus high salt intake and (2) angiotensin II infusion in hypercholesterolemia mice. Aortic BMAL1 was upregulated by deoxycorticosterone acetate-salt, and deletion of BMAL1 in SMCs selectively upregulated TIMP4 (tissue inhibitor of metalloproteinase 4) and suppressed deoxycorticosterone acetate-salt-induced MMP (matrix metalloproteinase) activation and elastin breakages. Moreover, BMAL1 bound to the Timp4 promoter and suppressed Timp4 transcription. CONCLUSIONS: These results reveal an important, but previously unexplored, role of SMC BMAL1 in AAA. Moreover, these results identify TIMP4 as a novel target of BMAL1, which may mediate the AAA protective effect of SMC BMAL1 deletion.


Subject(s)
ARNTL Transcription Factors/deficiency , Aortic Aneurysm, Abdominal/prevention & control , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , ARNTL Transcription Factors/genetics , Aldosterone , Angiotensin II , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Binding Sites , Desoxycorticosterone Acetate , Dilatation, Pathologic , Disease Models, Animal , Elastin/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Promoter Regions, Genetic , Sodium Chloride, Dietary , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , Transcription, Genetic , Tissue Inhibitor of Metalloproteinase-4
4.
Arterioscler Thromb Vasc Biol ; 33(7): 1568-79, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23661677

ABSTRACT

OBJECTIVE: Elevated plasma aldosterone concentrations in patients have been linked to a spectrum of cardiovascular diseases. Mineralocorticoid receptor antagonists provide additional benefits in patients with heart failure. However, whether aldosterone and the mineralocorticoid receptor are involved in aortic aneurysm is unknown. APPROACH AND RESULTS: We report that administration of deoxycorticosterone acetate (DOCA) and salt or aldosterone and salt, but not DOCA or salt alone, to C57BL/6 male mice induced abdominal and thoracic aortic aneurysm formation and rupture in an age-dependent manner. DOCA and salt- or aldosterone and salt-induced aortic aneurysm mimicked human aortic aneurysm with respect to elastin degradation, inflammatory cell infiltration, smooth muscle cell degeneration and apoptosis, and oxidative stress. Aortic aneurysm formation did not correlate with the increase in blood pressure induced by DOCA and salt. Systemic administration of the angiotensin-converting enzyme inhibitor, enalapril, or angiotensin type 1 receptor antagonist, losartan, did not affect DOCA and salt-induced aortic aneurysm. In contrast, the mineralocorticoid receptor antagonists, spironolactone or eplerenone, significantly attenuated DOCA and salt- or aldosterone and salt-induced aortic aneurysm. CONCLUSIONS: The current study describes a novel aortic aneurysm animal model induced by mineralocorticoid receptor agonist and high salt, and reveals a previously unrecognized but potentially significant role of aldosterone in the pathogenesis of aortic aneurysm. These findings imply that mineralocorticoid receptor antagonists may be effective in the treatment of some aortic aneurysms.


Subject(s)
Aorta/metabolism , Aortic Aneurysm, Abdominal/etiology , Aortic Aneurysm, Thoracic/etiology , Aortic Rupture/etiology , Desoxycorticosterone , Receptors, Mineralocorticoid/metabolism , Sodium Chloride, Dietary , Aldosterone/blood , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Aorta/drug effects , Aorta/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/physiopathology , Aortic Aneurysm, Thoracic/chemically induced , Aortic Aneurysm, Thoracic/drug therapy , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Aortic Aneurysm, Thoracic/physiopathology , Aortic Rupture/chemically induced , Aortic Rupture/drug therapy , Aortic Rupture/metabolism , Aortic Rupture/pathology , Aortic Rupture/physiopathology , Apoptosis , Blood Pressure , Disease Models, Animal , Elastin/metabolism , Enalapril/administration & dosage , Eplerenone , Losartan/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mineralocorticoid Receptor Antagonists/administration & dosage , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Oxidative Stress , Receptors, Mineralocorticoid/agonists , Spironolactone/administration & dosage , Spironolactone/analogs & derivatives , Time Factors
5.
bioRxiv ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39005289

ABSTRACT

Type 2 diabetics have an increased prevalence of hypertension and nondipping blood pressure (BP), which worsen cardiovascular outcomes. Exenatide, a short acting glucagon-like peptide-1 receptor agonist (GLP-1RA) used to treat type 2 diabetes, also demonstrates blood pressure (BP)-lowering effects. However, the mechanisms behind this and the impact of administration timing on BP dipping remain unclear. We investigated the effects of exenatide intraperitoneal injected at light onset (ZT0) or dark onset (ZT12) in diabetic (db/db) mice and nondiabetic controls. Using radio-telemetry and BioDAQ cages, we continuously monitored BP and food intake. Db/db mice exhibited non-dipping BP and increased food intake. ZT0 exenatide administration restored BP dipping by specifically lowering light-phase BP, while ZT12 exenatide reversed dipping by lowering dark-phase BP. These effects correlated with altered food intake patterns, and importantly, were abolished when food access was removed. Additionally, urinary norepinephrine excretion, measured by HPLC, was significantly reduced 6 hours post-exenatide at both ZT0 and ZT12, suggesting sympathetic nervous system involvement. Notably, combining exenatide with either ganglionic blocker mecamylamine or α-blocker prazosin did not enhance BP reduction beyond the individual effects of each blocker. These findings reveal that exenatide, when administered at light onset, restores BP dipping in db/db mice by suppressing light-phase food intake and sympathetic activity. Importantly, the efficacy of exenatide is dependent on food availability and its timing relative to circadian rhythms, highlighting the potential for chronotherapy in optimizing GLP-1RA- based treatments for type 2 diabetes and hypertension. Article Highlights: Maintaining a normal blood pressure (BP) circadian rhythm is vital for cardiovascular health, but diabetes often disrupts this rhythm. The effect of exenatide, a GLP-1 receptor agonist (GLP-1RA), on BP rhythm in diabetes is uncertain.This study investigates the impact of exenatide administration timing on BP patterns in diabetic db/db mice.Findings indicate that exenatide given at the onset of rest restores normal BP dipping, while at the start of the active phase worsens BP rhythm by modulating food intake and sympathetic activity.Timing GLP-1 RA administration may optimize BP control and provide cardiovascular benefits for type 2 diabetes patients.

6.
J Clin Invest ; 134(15)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900572

ABSTRACT

Androgen has long been recognized for its pivotal role in the sexual dimorphism of cardiovascular diseases, including aortic aneurysms (AAs), a devastating vascular disease with a higher prevalence and fatality rate in men than in women. However, the mechanism by which androgen mediates AAs is largely unknown. Here, we found that male, not female, mice developed AAs when exposed to aldosterone and high salt (Aldo-salt). We revealed that androgen and androgen receptors (ARs) were crucial for this sexually dimorphic response to Aldo-salt. We identified programmed cell death protein 1 (PD-1), an immune checkpoint, as a key link between androgen and AAs. Furthermore, we demonstrated that administration of anti-PD-1 Ab and adoptive PD-1-deficient T cell transfer reinstated Aldo-salt-induced AAs in orchiectomized mice and that genetic deletion of PD-1 exacerbated AAs induced by a high-fat diet and angiotensin II (Ang II) in nonorchiectomized mice. Mechanistically, we discovered that the AR bound to the PD-1 promoter to suppress the expression of PD-1 in the spleen. Thus, our study unveils a mechanism by which androgen aggravates AAs by suppressing PD-1 expression in T cells. Moreover, our study suggests that some patients with cancer might benefit from screenings for AAs during immune checkpoint therapy.


Subject(s)
Androgens , Aortic Aneurysm , Programmed Cell Death 1 Receptor , Receptors, Androgen , Animals , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Mice , Male , Female , Androgens/pharmacology , Androgens/metabolism , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Aortic Aneurysm/metabolism , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Aldosterone/metabolism , Mice, Knockout , Humans , Angiotensin II/pharmacology
7.
J Biol Chem ; 287(29): 24739-53, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22637477

ABSTRACT

Whether group VIA phospholipase A(2) (iPLA(2)ß) is involved in vascular inflammation and neointima formation is largely unknown. Here, we report that iPLA(2)ß expression increases in the vascular tunica media upon carotid artery ligation and that neointima formation is suppressed by genetic deletion of iPLA(2)ß or by inhibiting its activity or expression via perivascular delivery of bromoenol lactone or of antisense oligonucleotides, respectively. To investigate whether smooth muscle-specific iPLA(2)ß is involved in neointima formation, we generated transgenic mice in which iPLA(2)ß is expressed specifically in smooth muscle cells and demonstrate that smooth muscle-specific expression of iPLA(2)ß exacerbates ligation-induced neointima formation and enhanced both production of proinflammatory cytokines and vascular infiltration by macrophages. With cultured vascular smooth muscle cell, angiotensin II, arachidonic acid, and TNF-α markedly induce increased expression of IL-6 and TNF-α mRNAs, all of which were suppressed by inhibiting iPLA(2)ß activity or expression with bromoenol lactone, antisense oligonucleotides, and genetic deletion, respectively. Similar suppression also results from genetic deletion of 12/15-lipoxygenase or inhibiting its activity with nordihydroguaiaretic acid or luteolin. Expression of iPLA(2)ß protein in cultured vascular smooth muscle cells was found to depend on the phenotypic state and to rise upon incubation with TNF-α. Our studies thus illustrate that smooth muscle cell-specific iPLA(2)ß participates in the initiation and early progression of vascular inflammation and neointima formation and suggest that iPLA(2)ß may represent a novel therapeutic target for preventing cardiovascular diseases.


Subject(s)
Calcium/metabolism , Inflammation/metabolism , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Neointima/immunology , Neointima/metabolism , Phospholipases A2, Calcium-Independent/metabolism , Angiotensin II , Animals , Blotting, Western , Carotid Arteries/immunology , Carotid Arteries/metabolism , Cells, Cultured , Immunohistochemistry , Inflammation/immunology , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotides, Antisense , Phospholipases A2, Calcium-Independent/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Rabbits , Rats , Rats, Sprague-Dawley
8.
Am J Physiol Heart Circ Physiol ; 305(1): H104-13, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23604714

ABSTRACT

Recent data revealed that protein kinase C-potentiated myosin phosphatase inhibitor of 17 kDa (CPI-17), a myosin phosphatase inhibitory protein preferentially expressed in smooth muscle, is upregulated/activated in several diseases but whether this CPI-17 increase plays a causal role in pathologically enhanced vascular smooth muscle contractility and blood pressure remains unclear. To address this possibility, we generated a smooth muscle-specific CPI-17 transgenic mouse model (CPI-17-Tg) and demonstrated that the CPI-17 transgene was selectively expressed in smooth muscle-enriched tissues, including mesenteric arteries. The isometric contractions in the isolated second-order branch of mesenteric artery helical strips from CPI-17-Tg mice were significantly enhanced compared with controls in response to phenylephrine, U-46619, serotonin, ANG II, high potassium, and calcium. The perfusion pressure increases in isolated perfused mesenteric vascular beds in response to norepinephrine were also enhanced in CPI-17-Tg mice. The hypercontractility was associated with increased phosphorylation of CPI-17 and 20-kDa myosin light chain under basal and stimulated conditions. Surprisingly, the protein levels of rho kinase 2 and protein kinase Cα/δ were significantly increased in CPI-17-Tg mouse mesenteric arteries. Radiotelemetry measurements demonstrated that blood pressure was significantly increased in CPI-17-Tg mice. However, no vascular remodeling was detected by morphometric analysis. Taken together, our results demonstrate that increased CPI-17 expression in smooth muscle promotes vascular smooth muscle contractility and increases blood pressure, implicating a pathological significant role of CPI-17 upregulation.


Subject(s)
Blood Pressure , Isometric Contraction/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/physiology , Phosphoproteins/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Angiotensin II/pharmacology , Animals , Calcium/pharmacology , Intracellular Signaling Peptides and Proteins , Isometric Contraction/drug effects , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Phenylephrine/pharmacology , Phosphoproteins/genetics , Potassium/pharmacology , Serotonin/pharmacology , Transcription, Genetic , Up-Regulation , Vasoconstrictor Agents/pharmacology
9.
Sci Rep ; 13(1): 2748, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36797364

ABSTRACT

Previous study from our lab has revealed a new role of CD47 in regulating adipose tissue function, energy homeostasis and the development of obesity and metabolic disease in CD47 deficient mice. In this study, the therapeutic potential of an antisense oligonucleotide (ASO) targeting to CD47 in obesity and its-associated complications was determined in two obese mouse models (diet induced and genetic models). In diet induced obesity, male C57BL6 mice were fed with high fat (HF) diet to induce obesity and then treated with CD47ASO or control ASO for 8 weeks. In genetic obese mouse model, male six-week old ob/ob mice were treated with ASOs for 9 weeks. We found that CD47ASO treatment reduced HF diet-induced weight gain, decreased fat mass, prevented dyslipidemia, and improved glucose tolerance. These changes were accompanied by reduced inflammation in white adipose tissue and decreased hepatic steatosis. This protection was also seen in CD47ASO treated ob/ob mice. Mechanistically, CD47ASO treatment increased mice physical activity and energy expenditure, contributing to weight loss and improved metabolic outcomes in obese mice. Collectively, these findings suggest that CD47ASO might serve as a new treatment option for obesity and its-associated metabolic complications.


Subject(s)
Insulin Resistance , Oligonucleotides, Antisense , Animals , Male , Mice , CD47 Antigen/metabolism , Diet, High-Fat , Liver/metabolism , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Oligonucleotides, Antisense/genetics
10.
bioRxiv ; 2023 Jan 22.
Article in English | MEDLINE | ID: mdl-36711644

ABSTRACT

Androgen has long been recognized for its pivotal role in the sexual dimorphism of cardiovascular diseases, including aortic aneurysms, a devastating vascular disease with a higher prevalence and mortality rate in men than women. However, the molecular mechanism by which androgen mediates aortic aneurysms is largely unknown. Here, we report that male but not female mice develop aortic aneurysms in response to aldosterone and high salt (Aldo-salt). We demonstrate that both androgen and androgen receptors (AR) are crucial for the sexually dimorphic response to Aldo-salt. We identify T cells expressing programmed cell death protein 1 (PD-1), an immune checkpoint molecule important in immunity and cancer immunotherapy, as a key link between androgen and aortic aneurysms. We show that intraperitoneal injection of anti-PD-1 antibody reinstates Aldo-salt-induced aortic aneurysms in orchiectomized mice. Mechanistically, we demonstrate that AR binds to the PD-1 promoter to suppress its expression in the spleen. Hence, our study reveals an important but unexplored mechanism by which androgen contributes to aortic aneurysms by suppressing PD-1 expression in T cells. Our study also suggests that cancer patients predisposed to the risk factors of aortic aneurysms may be advised to screen for aortic aneurysms during immune checkpoint therapy.

11.
J Biol Chem ; 286(52): 44646-58, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22057271

ABSTRACT

Mice deficient in regulator of G-protein signaling-2 (RGS2) have severe hypertension, and RGS2 genetic variations occur in hypertensive humans. A potentially important negative feedback loop in blood pressure homeostasis is that angiotensin II (Ang II) increases vascular smooth muscle cell (VSMC) RGS2 expression. We reported that Group VIA phospholipase A(2) (iPLA(2)ß) is required for this response (Xie, Z., Gong, M. C., Su, W., Turk, J., and Guo, Z. (2007) J. Biol. Chem. 282, 25278-25289), but the specific molecular causes and consequences of iPLA(2)ß activation are not known. Here we demonstrate that both protein kinases C (PKC) and A (PKA) participate in Ang II-induced VSMC RGS2 mRNA up-regulation, and that actions of PKC and PKA precede and follow iPLA(2)ß activation, respectively. Moreover, we identified a conserved cAMP-response element (CRE) in the murine RGS2 promoter that is critical for cAMP-response element-binding protein (CREB) binding and RGS2 promoter activation. Forskolin-stimulated RGS2 mRNA up-regulation is inhibited by CREB sequestration or specific disruption of the CREB-RGS2 promoter interaction, and Ang II-induced CREB phosphorylation and nuclear localization are blocked by iPLA(2)ß pharmacologic inhibition or genetic ablation. Ang II-induced intracellular cyclic AMP accumulation precedes CREB phosphorylation and is diminished by inhibiting iPLA(2), cyclooxygenase, or lipoxygenase. Moreover, three single nucleotide polymorphisms identified in hypertensive patients are located in the human RGS2 promoter CREB binding site. Point mutations corresponding to these single nucleotide polymorphisms interfere with stimulation of human RGS2 promoter activity by forskolin. Our studies thus delineate a negative feedback loop to attenuate Ang II signaling in VSMC with potential importance in blood pressure homeostasis and the pathogenesis of human essential hypertension.


Subject(s)
Angiotensin II/metabolism , Cell Nucleus/metabolism , GTP-Binding Protein Regulators/biosynthesis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RGS Proteins/biosynthesis , Response Elements/physiology , Transcription, Genetic/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Angiotensin II/pharmacology , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Nucleus/genetics , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Protein Regulators/genetics , Group VI Phospholipases A2/genetics , Group VI Phospholipases A2/metabolism , Humans , Hypertension/genetics , Hypertension/metabolism , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Phosphorylation/drug effects , Phosphorylation/physiology , Polymorphism, Single Nucleotide , Protein Kinase C/genetics , Protein Kinase C/metabolism , RGS Proteins/genetics , Rabbits , Rats , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology , Vasoconstrictor Agents/metabolism , Vasoconstrictor Agents/pharmacology
12.
Am J Physiol Heart Circ Physiol ; 302(3): H621-33, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22140039

ABSTRACT

This study was designed to determine whether the 24-h rhythms of clock gene expression and vascular smooth muscle (VSM) contractile responses are altered in type 2 diabetic db/db mice. Control and db/db mice were euthanized at 6-h intervals throughout the day. The aorta, mesenteric arteries, heart, kidney, and brain were isolated. Clock and target gene mRNA levels were determined by either real-time PCR or in situ hybridization. Isometric contractions were measured in isolated aortic helical strips, and pressor responses to an intravenous injection of vasoconstrictors were determined in vivo using radiotelemetry. We found that the 24-h mRNA rhythms of the following genes were suppressed in db/db mice compared with control mice: the clock genes period homolog 1/2 (Per1/2) and cryptochrome 1/2 (Cry1/2) and their target genes D site albumin promoter-binding protein (Dbp) and peroxisome proliferator-activated receptor-γ (Pparg) in the aorta and mesenteric arteries; Dbp in the heart; Per1, nuclear receptor subfamily 1, group D, member 1 (Rev-erba), and Dbp in the kidney; and Per1 in the suprachiasmatic nucleus. The 24-h contractile variations in response to phenylephrine (α(1)-agonist), ANG II, and high K(+) were significantly altered in the aortas from db/db mice compared with control mice. The diurnal variations of the in vivo pressor responses to phenylephrine and ANG II were lost in db/db mice. Moreover, the 24-h mRNA rhythms of the contraction-related proteins Rho kinase 1/2, PKC-potentiated phosphatase inhibitory protein of 17 kDa, calponin-3, tropomyosin-1/2, and smooth muscle protein 22-α were suppressed in db/db mice compared with control mice. Together, our data demonstrated that the 24-h rhythms of clock gene mRNA, mRNA levels of several contraction-related proteins, and VSM contraction were disrupted in db/db mice, which may contribute to the disruption of their blood pressure circadian rhythm.


Subject(s)
Cryptochromes/genetics , Diabetes Mellitus, Type 2/genetics , Muscle, Smooth, Vascular/physiology , Period Circadian Proteins/genetics , Animals , Aorta/physiology , Blood Pressure/genetics , Circadian Rhythm/genetics , DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/physiopathology , Gene Expression/physiology , Heart/physiology , Kidney/physiology , Male , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , PPAR gamma/genetics , Suprachiasmatic Nucleus/physiology , Transcription Factors/genetics , Vasoconstriction/genetics
13.
Circ Res ; 106(3): 611-9, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20019328

ABSTRACT

RATIONALE: Angiotensin II (Ang II) has diverse effects on smooth muscle cells (SMCs). The diversity of effects may relate to the regional location of this cell type. OBJECTIVE: The aim of this study was to define whether Ang II exerted divergent effects on smooth muscle cells in the aorta and determine the role of blood pressure and specific oxidant mechanisms. METHODS AND RESULTS: Ang II (1000 ng/kg per minute) infusion for 28 days into mice increased systolic blood pressure and promoted medial expansion of equivalent magnitude throughout the entire aorta. Both effects were ablated by angiotensin II type 1a (AT(1a)) receptor deficiency. Similar increases in systolic blood pressure by administration of norepinephrine promoted no changes in aortic medial thickness. Increased medial thickness was attributable to SMC expansion owing to hypertrophy in most aortic regions, with the exception of hyperplasia of the ascending aorta. Deficiency of the p47(phox) component of NADPH oxidase ablated Ang II-induced medial expansion in all aortic regions. Analysis of mRNA and protein throughout the aorta revealed a much higher abundance of the inhibitor of differentiation 3 (Id3) in the ascending aorta compared to all other regions. A functional role was demonstrated by Id3 deficiency inhibiting Ang II-induced SMC hyperplasia of the ascending aorta. CONCLUSIONS: In conclusion, Ang II promotes both aortic medial hypertrophy and hyperplasia in a region-specific manner via an oxidant mechanism. The ascending aortic hyperplasia is dependent on Id3.


Subject(s)
Angiotensin II/toxicity , Aorta/drug effects , Inhibitor of Differentiation Proteins/physiology , Receptor, Angiotensin, Type 1/physiology , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Animals , Aorta/pathology , Blood Pressure/drug effects , Blood Pressure/physiology , Hyperplasia , Hypertension/chemically induced , Hypertension/drug therapy , Hypertension/prevention & control , Losartan/pharmacology , Losartan/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Norepinephrine/pharmacology , Organ Specificity , Renin/blood , Tunica Media/pathology
14.
Front Nutr ; 9: 969345, 2022.
Article in English | MEDLINE | ID: mdl-36159491

ABSTRACT

Disruption of blood pressure (BP) circadian rhythm, independent of hypertension, is emerging as an index for future target organ damage and is associated with a higher risk of cardiovascular events. Previous studies showed that changing food availability time alters BP rhythm in several mammalian species. However, the underlying mechanisms remain largely unknown. To address this, the current study specifically investigates (1) the relationship between rhythms of food intake and BP in wild-type mice; (2) effects of light-phase time-restricted feeding (TRF, food only available during light-phase) on BP circadian rhythm in wild-type and diabetic db/db mice; (3) the roles of the autonomic system and clock gene in light-phase TRF induced changes in BP circadian rhythm. Food intake and BP of C57BL/6J and db/db mice were simultaneously and continuously recorded using BioDAQ and telemetry systems under ad libitum or light-phase TRF. Per2 protein daily oscillation was recorded in vivo by IVIS spectrum in mPer2 Luc mice. Autonomic nerve activity was evaluated by heart rate variability, baroreflex, urinary norepinephrine (NE) and epinephrine (Epi) excretion, and mRNA expressions of catecholamines biosynthetic and catabolic enzymes, and alpha-adrenergic receptors in mesenteric resistance arteries. We found that in wild-type mice, the BP level was correlated with the food intake temporally across the 24 h. Reversing the feeding time by imposing light-phase TRF resulted in reverse or inverted BP dipping. Interestingly, the net changes in food intake were correlated with the net alteration in BP temporally under light-phase TRF. In db/db mice, light-phase TRF worsened the existing non-dipping BP. The food intake and BP circadian rhythm changes were associated with alterations in Per2 protein daily oscillation and the time-of-day variations in heart rate variability, baroreflex, and urinary excretion of NE and Epi, and increased mRNA expression of Slc6a2 (encoding NE transporter) and Adra1d (encoding alpha-adrenergic receptor 1d) in the mesenteric resistance arteries, indicating the sympathetic nervous system (SNS) was modulated after light-phase TRF. Collectively, our results demonstrated that light-phase TRF results in reverse dipping of BP in wild-type and diabetic db/db mice and revealed the potential role of the sympathetic pathway in light-phase TRF-induced BP circadian rhythm alteration.

15.
J Biol Chem ; 285(12): 8628-38, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20086008

ABSTRACT

Previous studies suggest that high glucose-induced RhoA/Rho kinase/CPI-17 activation is involved in diabetes-associated vascular smooth muscle hypercontractility. However, the upstream signaling that links high glucose and RhoA/Rho kinase/CPI-17 activation is unknown. Here we report that calcium-independent phospholipase A(2)beta (iPLA(2)beta) is required for high glucose-induced RhoA/Rho kinase/CPI-17 activation and thereby contributes to diabetes-associated vascular smooth muscle hypercontractility. We demonstrate that high glucose increases iPLA(2)beta mRNA, protein, and iPLA(2) activity in a time-dependent manner. Protein kinase C is involved in high glucose-induced iPLA(2)beta protein up-regulation. Inhibiting iPLA(2)beta activity with bromoenol lactone or preventing its expression by genetic deletion abolishes high glucose-induced RhoA/Rho kinase/CPI-17 activation, and restoring expression of iPLA(2)beta in iPLA(2)beta-deficient cells also restores high glucose-induced CPI-17 phosphorylation. Pharmacological and genetic inhibition of 12/15-lipoxygenases has effects on high glucose-induced CPI-17 phosphorylation similar to iPLA(2)beta inhibition. Moreover, increases in iPLA(2) activity and iPLA(2)beta protein expression are also observed in both type 1 and type 2 diabetic vasculature. Pharmacological and genetic inhibition of iPLA(2)beta, but not iPLA(2)gamma, diminishes diabetes-associated vascular smooth muscle hypercontractility. In summary, our results reveal a novel mechanism by which high glucose-induced, protein kinase C-mediated iPLA(2)beta up-regulation activates the RhoA/Rho kinase/CPI-17 via 12/15-lipoxygenases and thereby contributes to diabetes-associated vascular smooth muscle hypercontractility.


Subject(s)
Calcium/metabolism , Glucose/metabolism , Group IV Phospholipases A2/metabolism , Muscle, Smooth/metabolism , Phosphoprotein Phosphatases/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Enzyme Activation , Female , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Muscle Contraction , Muscle Proteins , Phosphorylation , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley
16.
Curr Opin Pharmacol ; 57: 125-131, 2021 04.
Article in English | MEDLINE | ID: mdl-33721615

ABSTRACT

The intrinsic vascular smooth muscle contraction and vasoconstriction show time-of-day variations, contributing to the blood pressure circadian rhythm, which is essential for cardiovascular health. This brief review provides an overview of our current understanding of the mechanisms underlying the time-of-day variations of vascular smooth muscle contraction. We discuss the potential contribution of the time-of-day variations of vasoconstriction to the physiological blood pressure circadian rhythm. Finally, we survey the data obtained in the type 2 diabetic db/db mouse model that demonstrate the alterations of the time-of-day variations of vasoconstriction and the nondipping blood pressure in diabetes.


Subject(s)
Diabetes Mellitus , Vasoconstriction , Animals , Blood Pressure , Circadian Rhythm , Mice
17.
Am J Physiol Endocrinol Metab ; 299(3): E351-63, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20530739

ABSTRACT

Atherosclerosis is the most common cause of cardiovascular diseases in the world. Although the development of atherosclerosis appears to be the result of multiple maladaptive pathways, a particularly important factor in the pathogenesis of atherosclerosis is oxidized low-density lipoprotein (ox-LDL), which contributes to endothelial damage. Data from our laboratory and others show that follistatin-related protein (FRP), which is expressed in the vasculature, has cardioprotective effects, suggesting that loss of FRP protection might play a role in the development of atherosclerosis. In the present study, we determined whether FRP overexpression protects against endothelial cell (EC) damage, an intermediate end point for atherosclerosis. We bred apoE-knockout (apoE(-/-)) mice that were FRP(+) transgenic (they overexpressed FRP). We compared them with control mice (their littermates). Human umbilical vein endothelial cells (HUVECs) were isolated and treated with ox-LDL and recombinant FRP. FRP-induced signal transduction and Bcl-2 mRNA and protein stability were analyzed. After 16 wk, apoE(-/-) FRP(+) mice had significantly fewer apoptotic ECs than controls. In vitro experiments showed that the effect of FRP on EC apoptosis was mediated by upregulation of expression of the antiapoptotic protein Bcl-2. In HUVECs, FRP upregulated Bcl-2 transcription via a PI3K-Akt-NF-kappaB pathway. We conclude that FRP overexpression maintains EC viability by preventing apoptosis via Bcl-2 upregulation. FRP may be a novel therapeutic target for the prevention and treatment of vascular EC injury and of atherosclerosis.


Subject(s)
Apolipoproteins E/metabolism , Apoptosis/drug effects , Atherosclerosis/metabolism , Endothelium, Vascular/metabolism , Follistatin-Related Proteins/pharmacology , Lipoproteins, LDL/metabolism , Animals , Apoptosis/physiology , Atherosclerosis/pathology , Cell Survival/drug effects , Cell Survival/physiology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/pathology , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Proteins/metabolism
18.
Cell Metab ; 32(1): 44-55.e6, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32402267

ABSTRACT

Age is a non-modifiable risk factor for the inflammation that underlies age-associated diseases; thus, anti-inflammaging drugs hold promise for increasing health span. Cytokine profiling and bioinformatic analyses showed that Th17 cytokine production differentiates CD4+ T cells from lean, normoglycemic older and younger subjects, and mimics a diabetes-associated Th17 profile. T cells from older compared to younger subjects also had defects in autophagy and mitochondrial bioenergetics that associate with redox imbalance. Metformin ameliorated the Th17 inflammaging profile by increasing autophagy and improving mitochondrial bioenergetics. By contrast, autophagy-targeting siRNA disrupted redox balance in T cells from young subjects and activated the Th17 profile by activating the Th17 master regulator, STAT3, which in turn bound IL-17A and F promoters. Mitophagy-targeting siRNA failed to activate the Th17 profile. We conclude that metformin improves autophagy and mitochondrial function largely in parallel to ameliorate a newly defined inflammaging profile that echoes inflammation in diabetes.


Subject(s)
Aging/drug effects , Autophagy/drug effects , Hypoglycemic Agents/pharmacology , Inflammation/metabolism , Metformin/pharmacology , Mitochondria/drug effects , Adult , Aging/metabolism , Humans , Middle Aged , Mitochondria/metabolism
19.
J Biol Rhythms ; 34(1): 51-68, 2019 02.
Article in English | MEDLINE | ID: mdl-30278816

ABSTRACT

Diabetic patients have an increased prevalence of blood pressure (BP) circadian rhythm disruption, which is associated with an increased risk of target organ damage and detrimental cardiovascular events. Limited information is available regarding the role of clock genes in the disruption of BP circadian rhythm in diabetes due to the lack of a diabetic animal model that allows real-time monitoring of clock gene oscillation. Here, we generated a novel diabetic db/db-mPer2Luc mouse model by crossing type 2 diabetic db/db mice with mPer2Luc knock-in mice. The daily rhythms of BP, heart rate, locomotor activity, and food and water intake were acquired by radiotelemetry or using metabolic chambers. The daily oscillation of mPer2 bioluminescence was recorded by LumiCycle in real-time in tissue explants and using the IVIS system in vivo. Our results show that db/db-mPer2Luc mice are obese, diabetic, and glucose intolerant. The db/db-mPer2Luc mice displayed a compromised BP daily rhythm, which was associated with disrupted daily rhythms in baroreflex sensitivity, locomotor activity, and metabolism, but not heart rate or food and water intake. The phase of the mPer2 daily oscillation was advanced to different extents in the explanted peripheral tissues from db/db-mPer2Luc mice relative to control mice. In contrast, no phase shift was detected in mPer2 daily oscillations in the explanted SCN. Moreover, advanced phase shift of the mPer2 daily oscillation was detected in the liver, kidney and submandibular gland in vivo of db/db-mPer2Luc mice. In conclusion, the diabetic db/db-mPer2Luc mouse is a novel animal model that allows real-time monitoring of mPer2 circadian rhythms ex vivo and in vivo. The results from db/db-mPer2Luc mice suggest that the desynchrony of mPer2 daily oscillation in peripheral tissues contributes to the loss of BP daily oscillation in diabetes.


Subject(s)
Blood Pressure , Circadian Clocks/genetics , Circadian Rhythm , Diabetes Mellitus, Experimental/physiopathology , Disease Models, Animal , Animals , Diabetes Mellitus, Experimental/complications , Female , Male , Mice , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Suprachiasmatic Nucleus/physiology
20.
Mol Cell Biol ; 39(11)2019 06 01.
Article in English | MEDLINE | ID: mdl-30936247

ABSTRACT

Vasodilatory shock in sepsis is caused by the failure of the vasculature to respond to vasopressors, which results in hypotension, multiorgan failure, and ultimately patient death. Recently, it was reported that CPI-17, a key player in the regulation of smooth muscle contraction, was downregulated by lipopolysaccharide (LPS) in mesenteric arteries concordant with vascular hypocontractilty. While Sp1 has been shown to activate CPI-17 transcription, it is unknown whether Sp1 is involved in LPS-induced smooth muscle CPI-17 downregulation. Here we report that tumor necrosis factor (TNF) was critical for LPS-induced smooth muscle CPI-17 downregulation. Mechanistically, we identified two GC boxes as a key TNF response element in the CPI-17 promoter and demonstrated that KLF4 was upregulated by TNF, competed with Sp1 for the binding to the GC boxes in the CPI-17 promoter, and repressed CPI-17 transcription through histone deacetylases (HDACs). Moreover, genetic deletion of TNF or pharmacological inhibition of HDACs protected mice from LPS-induced smooth muscle CPI-17 downregulation, vascular hypocontractility, hypotension, and mortality. In summary, these data provide a novel mechanism of the transcriptional control of CPI-17 in vascular smooth muscle cells under inflammatory conditions and suggest a new potential therapeutic strategy for the treatment of vasodilatory shock in sepsis.


Subject(s)
Hypotension/genetics , Intracellular Signaling Peptides and Proteins/genetics , Kruppel-Like Transcription Factors/metabolism , Lipopolysaccharides/metabolism , Muscle Proteins/genetics , Muscle, Smooth, Vascular/cytology , Sp1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Down-Regulation , Gene Knockout Techniques , Humans , Hypotension/metabolism , Kruppel-Like Factor 4 , Mice , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL