Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Prog Transplant ; 28(2): 157-162, 2018 06.
Article in English | MEDLINE | ID: mdl-29558875

ABSTRACT

BACKGROUND: Despite ventricular assist devices (VADs) becoming more common in heart failure (HF) treatment, it is still uncertain which patients are more prone to complications. One potential risk factor is increased body mass index (BMI), which is known to increase both all-cause mortality and mortality from ischemic heart disease; however, the role of the BMI in predicting morbidity and mortality following device implantation is unclear. METHODS: The study population for this single-institution retrospective chart review consisted of 136 patients with HF, who underwent VAD implantation between 2004 and 2015. Patients were divided into 2 groups based on their BMI: a nonobese group (18.5 < BMI < 30.0; n = 82) or an obese group (BMI >30.0; n = 54). These groups were compared at baseline and after implantation for survival, hospital readmission, and adverse events. RESULTS: No significant difference was found in initial hospital length of stay, number or length of readmissions, or readmission diagnosis. At 1 year, rates of ongoing device support, orthotopic heart transplant (OHT), and death were not significantly different between groups ( P = .89, P = .90, and P = .70, respectively). Multivariate analysis did not identify obesity as an independent predictor of mortality ( P = .90); only biventricular assist device implantation was associated with decreased survival (hazard ratio [HR] = 5.90, P = .002). CONCLUSION: Obesity in itself should not preclude the use of VAD support in patients with HF, as carefully selected obese patients were shown to have similar rates of hospital readmission, 1-year outcomes, and survival following device implantation compared to nonobese patients.


Subject(s)
Body Mass Index , Heart Failure/surgery , Heart-Assist Devices/adverse effects , Postoperative Complications/etiology , Postoperative Complications/mortality , Adult , Aged , Cause of Death , Female , Humans , Male , Middle Aged , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Factors , Treatment Outcome
2.
Pharmacol Res ; 110: 52-64, 2016 08.
Article in English | MEDLINE | ID: mdl-27180008

ABSTRACT

Heart failure (HF) is a global epidemic with the highest degree of mortality and morbidity of any disease presently studied. G protein-coupled receptors (GPCRs) are prominent regulators of cardiovascular function. Activated GPCRs are "turned off" by GPCR kinases (GRKs) in a process known as "desensitization". GRKs 2 and 5 are highly expressed in the heart, and known to be upregulated in HF. Over the last 20 years, both GRK2 and GRK5 have been demonstrated to be critical mediators of the molecular alterations that occur in the failing heart. In the present review, we will highlight recent findings that further characterize "non-canonical" GRK signaling observed in HF. Further, we will also present potential therapeutic strategies (i.e. small molecule inhibition, microRNAs, gene therapy) that may have potential in combating the deleterious effects of GRKs in HF.


Subject(s)
Cardiovascular System/enzymology , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Heart Failure/enzymology , Animals , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Enzyme Inhibitors/therapeutic use , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 5/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 5/genetics , Genetic Therapy , Heart Failure/genetics , Heart Failure/physiopathology , Heart Failure/therapy , Humans , MicroRNAs/therapeutic use , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects
3.
J Mol Cell Cardiol ; 92: 196-202, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829117

ABSTRACT

As the average world-wide lifespan continues to increase, heart failure (HF) has dramatically increased in incidence leading to the highest degree of mortality and morbidity of any disease presently studied. G protein-coupled receptors (GPCRs) play a prominent role in regulation of cardiovascular function. GPCRs are effectively "turned off" by GPCR kinases (GRKs) in a process known as "desensitization". GRKs 2 and 5 are highly expressed in the heart, and known to be upregulated in HF. Over the last 20years, the role of GRK2 in HF has been widely studied. However, until recently, the role of GRK5 in cardiac pathophysiology had yet to be elucidated. In the present review, we will focus on GRK5's role in the myocardium in normal physiology, and its apparent critical role in the progression of HF. Further, we will also present potential therapeutic strategies (i.e. small molecule inhibition, gene therapy) that may have potential in combating the deleterious effects of GRK5 in HF.


Subject(s)
G-Protein-Coupled Receptor Kinase 5/genetics , Heart Failure/genetics , Myocardium/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , Heart Failure/metabolism , Heart Failure/pathology , Humans , Molecular Targeted Therapy , Myocardium/pathology
4.
Circ Res ; 117(12): 1001-12, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26515328

ABSTRACT

RATIONALE: G protein-coupled receptor kinases (GRKs) are dynamic regulators of cellular signaling. GRK5 is highly expressed within myocardium and is upregulated in heart failure. Although GRK5 is a critical regulator of cardiac G protein-coupled receptor signaling, recent data has uncovered noncanonical activity of GRK5 within nuclei that plays a key role in pathological hypertrophy. Targeted cardiac elevation of GRK5 in mice leads to exaggerated hypertrophy and early heart failure after transverse aortic constriction (TAC) because of GRK5 nuclear accumulation. OBJECTIVE: In this study, we investigated the role of GRK5 in physiological, swimming-induced hypertrophy (SIH). METHODS AND RESULTS: Cardiac-specific GRK5 transgenic mice and nontransgenic littermate control mice were subjected to a 21-day high-intensity swim protocol (or no swim sham controls). SIH and specific molecular and genetic indices of physiological hypertrophy were assessed, including nuclear localization of GRK5, and compared with TAC. Unlike after TAC, swim-trained transgenic GRK5 and nontransgenic littermate control mice exhibited similar increases in cardiac growth. Mechanistically, SIH did not lead to GRK5 nuclear accumulation, which was confirmed in vitro as insulin-like growth factor-1, a known mediator of physiological hypertrophy, was unable to induce GRK5 nuclear translocation in myocytes. We found specific patterns of altered gene expression between TAC and SIH with GRK5 overexpression. Further, SIH in post-TAC transgenic GRK5 mice was able to preserve cardiac function. CONCLUSIONS: These data suggest that although nuclear-localized GRK5 is a pathological mediator after stress, this noncanonical nuclear activity of GRK5 is not induced during physiological hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/pathology , G-Protein-Coupled Receptor Kinase 5/physiology , Myocytes, Cardiac/metabolism , Animals , Animals, Newborn , Cardiomegaly/genetics , Cells, Cultured , Mice , Mice, Transgenic , Myocytes, Cardiac/pathology , Rats
5.
Circ Res ; 115(12): 976-85, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25332207

ABSTRACT

RATIONALE: G protein-coupled receptor kinases (GRKs) acting in the cardiomyocyte regulate important signaling events that control cardiac function. Both GRK2 and GRK5, the predominant GRKs expressed in the heart, have been shown to be upregulated in failing human myocardium. Although the canonical role of GRKs is to desensitize G protein-coupled receptors via phosphorylation, it has been demonstrated that GRK5, unlike GRK2, can reside in the nucleus of myocytes and exert G protein-coupled receptor-independent effects that promote maladaptive cardiac hypertrophy and heart failure. OBJECTIVE: To explore novel mechanisms by which GRK5 acting in the nucleus of cardiomyocytes participates in pathological cardiac hypertrophy. METHODS AND RESULTS: In this study, we have found that GRK5-mediated pathological cardiac hypertrophy involves the activation of the nuclear factor of activated T cells (NFAT) because GRK5 causes enhancement of NFAT-mediated hypertrophic gene transcription. Transgenic mice with cardiomyocyte-specific GRK5 overexpression activate an NFAT-reporter in mice basally and after hypertrophic stimulation, including transverse aortic constriction and phenylephrine treatment. Complimentary to this, GRK5 null mice exhibit less NFAT transcriptional activity after transverse aortic constriction. Furthermore, the loss of NFATc3 expression in the heart protected GRK5 overexpressing transgenic mice from the exaggerated hypertrophy and early progression to heart failure seen after transverse aortic constriction. Molecular studies suggest that GRK5 acts in concert with NFAT to increase hypertrophic gene transcription in the nucleus via GRK5's ability to bind DNA directly without a phosphorylation event. CONCLUSIONS: GRK5, acting in a kinase independent manner, is a facilitator of NFAT activity and part of a DNA-binding complex responsible for pathological hypertrophic gene transcription.


Subject(s)
Cardiomegaly/enzymology , G-Protein-Coupled Receptor Kinase 5/metabolism , Myocytes, Cardiac/enzymology , NFATC Transcription Factors/metabolism , Animals , Binding Sites , Cardiomegaly/etiology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Line , Cell Nucleus/enzymology , Disease Models, Animal , Disease Progression , Female , G-Protein-Coupled Receptor Kinase 5/genetics , Gene Expression Regulation , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Myocytes, Cardiac/pathology , NFATC Transcription Factors/genetics , Promoter Regions, Genetic , Rats , Time Factors , Transcription, Genetic , Transfection
6.
PLoS One ; 8(3): e57324, 2013.
Article in English | MEDLINE | ID: mdl-23472081

ABSTRACT

G protein-Coupled Receptors (GPCRs) kinases (GRKs) play a crucial role in regulating cardiac hypertrophy. Recent data from our lab has shown that, following ventricular pressure overload, GRK5, a primary cardiac GRK, facilitates maladaptive myocyte growth via novel nuclear localization. In the nucleus, GRK5's newly discovered kinase activity on histone deacetylase 5 induces hypertrophic gene transcription. The mechanisms governing the nuclear targeting of GRK5 are unknown. We report here that GRK5 nuclear accumulation is dependent on Ca(2+)/calmodulin (CaM) binding to a specific site within the amino terminus of GRK5 and this interaction occurs after selective activation of hypertrophic Gq-coupled receptors. Stimulation of myocytes with phenylephrine or angiotensinII causes GRK5 to leave the sarcolemmal membrane and accumulate in the nucleus, while the endothelin-1 does not cause nuclear GRK5 localization. A mutation within the amino-terminus of GRK5 negating CaM binding attenuates GRK5 movement from the sarcolemma to the nucleus and, importantly, overexpression of this mutant does not facilitate cardiac hypertrophy and related gene transcription in vitro and in vivo. Our data reveal that CaM binding to GRK5 is a physiologically relevant event that is absolutely required for nuclear GRK5 localization downstream of hypertrophic stimuli, thus facilitating GRK5-dependent regulation of maladaptive hypertrophy.


Subject(s)
Calmodulin/metabolism , G-Protein-Coupled Receptor Kinase 5/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation, Enzymologic , Myocardium/metabolism , Active Transport, Cell Nucleus , Adenoviridae/metabolism , Angiotensin II/metabolism , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Hypertrophy , Ligands , Male , Mice , Mice, Transgenic , Phenylephrine/pharmacology , Protein Binding , Protein Structure, Tertiary , Rabbits , Rats
7.
Dalton Trans ; 39(26): 6049-55, 2010 Jul 14.
Article in English | MEDLINE | ID: mdl-20571648

ABSTRACT

Reported are the synthesis and the structural characterization of the new ternary antimonides Eu(7)Ga(8)Sb(8), Sr(7)Ga(8)Sb(8), Ba(7)Ga(8)Sb(8), and Ba(7)In(8)Sb(8). They have been synthesized from reactions of the corresponding elements, using gallium or indium as metal fluxes. The four compounds are isostructural and crystallize with the space group P6(3)/mmc (no. 194) with lattice parameters as follows: a = 4.4942(8), c = 17.274(6) A for Eu(7)Ga(8)Sb(8); a = 4.5409(8), c = 17.486(4) A for Sr(7)Ga(8)Sb(8); a = 4.7045(12), c = 18.123(9) A for Ba(7)Ga(8)Sb(8); and a = 4.8274(3), c = 18.421(2) A for Ba(7)In(8)Sb(8). The temperature dependence of the magnetic susceptibility of Eu(7)Ga(8)Sb(8) confirms that the Eu ions are in the typical divalent state (Eu(2+)) with a room temperature effective magnetic moment mu(eff) = 8.02 mu(B). A phase transition from a paramagnetic to an antiferromagnetically ordered structure occurs in this material below 6 K. These results are corroborated by (151)Eu Mössbauer spectroscopy at 4.2 and 78 K, respectively. Eu(7)Ga(8)Sb(8), Ba(7)In(8)Sb(8) and the previously reported Sr(7)Ga(2)Sb(6) derivative of the cubic Th(3)P(4) type, were also investigated by (121)Sb Mössbauer spectroscopy.

SELECTION OF CITATIONS
SEARCH DETAIL
...