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










Publication year range
1.
FASEB J ; 38(14): e23810, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39042586

ABSTRACT

Osteofibrous dysplasia (OFD) is a rare, benign, fibro-osseous lesion that occurs most commonly in the tibia of children. Tibial involvement leads to bowing and predisposes to the development of a fracture which exhibit significantly delayed healing processes, leading to prolonged morbidity. We previously identified gain-of-function mutations in the MET gene as a cause for OFD. In our present study, we test the hypothesis that gain-of-function MET mutations impair bone repair due to reduced osteoblast differentiation. A heterozygous Met exon 15 skipping (MetΔ15-HET) mouse was created to imitate the human OFD mutation. The mutation results in aberrant and dysregulation of MET-related signaling determined by RNA-seq in the murine osteoblasts extracted from the wide-type and genetic mice. Although no gross skeletal defects were identified in the mice, fracture repair was delayed in MetΔ15-HET mice, with decreased bone formation observed 2-week postfracture. Our data are consistent with a novel role for MET-mediated signaling regulating osteogenesis.


Subject(s)
Bone Diseases, Developmental , Disease Models, Animal , Fibrous Dysplasia of Bone , Fracture Healing , Osteogenesis , Proto-Oncogene Proteins c-met , Animals , Mice , Osteogenesis/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Fracture Healing/genetics , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/pathology , Humans , Fibrous Dysplasia of Bone/genetics , Fibrous Dysplasia of Bone/pathology , Fibrous Dysplasia of Bone/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Mutation , Cell Differentiation , Mice, Inbred C57BL , Male
2.
FASEB J ; 38(13): e23753, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38924591

ABSTRACT

Lunatic Fringe (LFNG) is required for spinal development. Biallelic pathogenic variants cause spondylocostal dysostosis type-III (SCD3), a rare disease generally characterized by malformed, asymmetrical, and attenuated development of the vertebral column and ribs. However, a variety of SCD3 cases reported have presented with additional features such as auditory alterations and digit abnormalities. There has yet to be a single, comprehensive, functional evaluation of causative LFNG variants and such analyses could unveil molecular mechanisms for phenotypic variability in SCD3. Therefore, nine LFNG missense variants associated with SCD3, c.564C>A, c.583T>C, c.842C>A, c.467T>G, c.856C>T, c.601G>A, c.446C>T, c.521G>A, and c.766G>A, were assessed in vitro for subcellular localization and protein processing. Glycosyltransferase activity was quantified for the first time in the c.583T>C, c.842C>A, and c.446C>T variants. Primarily, our results are the first to satisfy American College of Medical Genetics and Genomics PS3 criteria (functional evidence via well-established assay) for the pathogenicity of c.583T>C, c.842C>A, and c.446C>T, and replicate this evidence for the remaining six variants. Secondly, this work indicates that all variants that prevent Golgi localization also lead to impaired protein processing. It appears that the FRINGE domain is responsible for this phenomenon. Thirdly, our data suggests that variant proximity to the catalytic residue may influence whether LFNG is improperly trafficked and/or enzymatically dysfunctional. Finally, the phenotype of the axial skeleton, but not elsewhere, may be modulated in a variant-specific fashion. More reports are needed to continue testing this hypothesis. We anticipate our data will be used as a basis for discussion of genotype-phenotype correlations in SCD3.


Subject(s)
Dysostoses , Genetic Variation , Glycosyltransferases , Animals , Mice , Cell Line , Chlorocebus aethiops , Dysostoses/congenital , Dysostoses/genetics , Genetic Variation/genetics , Genomics , Glycosyltransferases/genetics , NIH 3T3 Cells , Protein Processing, Post-Translational/genetics , Protein Transport/genetics , Proteomics
3.
Stem Cells Transl Med ; 13(4): 362-370, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38159082

ABSTRACT

Direct reprogramming (DR) is an emerging technique that can be applied to convert fibroblasts into osteoblast-like cells, promoting bone formation and regeneration. We review the current methodology of DR in relation to the creation of induced osteoblasts, including a comparison of transcription factor-mediated reprogramming and nontranscription factor-mediated reprogramming. We review the selection of reprogramming factors and delivery systems required. Transcription factor cocktails, such as the RXOL cocktail (Runx2, Osx, OCT3/4, and L-MYC), have shown promise in inducing osteogenic differentiation in fibroblasts. Alterations to the original cocktail, such as the addition of Oct9 and N-myc, have resulted in improved reprogramming efficiency. Transcription factor delivery includes integrative and nonintegrative systems which encompass viral vectors and nonviral methods such as synthetic RNA. Recently, an integrative approach using self-replicating RNA has been developed to achieve a longer and more sustained transcription factor expression. Nontranscription factor-mediated reprogramming using small molecules, proteins, inhibitors, and agonists has also been explored. For example, IGFBP7 protein supplementation and ALK5i-II inhibitor treatment have shown potential in enhancing osteoblast reprogramming. Direct reprogramming methods hold great promise for advancing bone regeneration and tissue repair, providing a potential therapeutic approach for fracture healing and the repair of bone defects. Multiple obstacles and constraints need to be addressed before a clinically significant level of cell therapy will be reached. Further research is needed to optimize the efficiency of the reprogramming cocktails, delivery methods, and safety profile of the reprogramming process.


Subject(s)
Cellular Reprogramming , Osteogenesis , Osteoblasts/metabolism , Cell Differentiation , Transcription Factors/metabolism , Fibroblasts , RNA/metabolism
4.
Hum Genet ; 142(11): 1571-1586, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37755482

ABSTRACT

CYP26B1 metabolizes retinoic acid in the developing embryo to regulate its levels. A limited number of individuals with pathogenic variants in CYP26B1 have been documented with a varied phenotypic spectrum, spanning from a severe manifestation involving skull anomalies, craniosynostosis, encephalocele, radio-humeral fusion, oligodactyly, and a narrow thorax, to a milder presentation characterized by craniosynostosis, restricted radio-humeral joint mobility, hearing loss, and intellectual disability. Here, we report two families with CYP26B1-related phenotypes and describe the data obtained from functional studies of the variants. Exome and Sanger sequencing were used for variant identification in family 1 and family 2, respectively. Family 1 reflects a mild phenotype, which includes craniofacial dysmorphism with brachycephaly (without craniosynostosis), arachnodactyly, reduced radioulnar joint movement, conductive hearing loss, learning disability-and compound heterozygous CYP26B1 variants: (p.[(Pro118Leu)];[(Arg234Gln)]) were found. In family 2, a stillborn fetus presented a lethal phenotype with spina bifida occulta, hydrocephalus, poor skeletal mineralization, synostosis, limb defects, and a synonymous homozygous variant in CYP26B1: c.1083C > A. A minigene assay revealed that the synonymous variant created a new splice site, removing part of exon 5 (p.Val361_Asp382del). Enzymatic activity was assessed using a luciferase assay, demonstrating a notable reduction in exogenous retinoic acid metabolism for the variant p.Val361_Asp382del. (~ 3.5 × decrease compared to wild-type); comparatively, the variants p.(Pro118Leu) and p.(Arg234Gln) demonstrated a partial loss of metabolism (1.7× and 2.3× reduction, respectively). A proximity-dependent biotin identification assay reaffirmed previously reported ER-resident protein interactions. Additional work into these interactions is critical to determine if CYP26B1 is involved with other biological events on the ER. Immunofluorescence assay suggests that mutant CYP26B1 is still localized in the endoplasmic reticulum. These results indicate that novel pathogenic variants in CYP26B1 result in varying levels of enzymatic activity that impact retinoic acid metabolism and relate to the distinct phenotypes observed.


Subject(s)
Craniosynostoses , Tretinoin , Humans , Retinoic Acid 4-Hydroxylase/genetics , Tretinoin/metabolism , Homozygote , Exons , Craniosynostoses/genetics
5.
J Mol Cell Cardiol ; 125: 162-173, 2018 12.
Article in English | MEDLINE | ID: mdl-30381233

ABSTRACT

Numerous epidemiological studies have demonstrated that approximately 40% of myocardial infarctions (MI) are associated with heart failure (HF). Resveratrol, a naturally occurring polyphenol, has been shown to be beneficial in the treatment of MI-induced HF in rodent models. However, the mechanism responsible for the effects of resveratrol are poorly understood. Interestingly, resveratrol is known to inhibit cytochrome P450 1B1 (CYP1B1) which is involved in the formation of cardiotoxic hydroxyeicosatetraenoic acid (HETE) metabolites. Therefore, we investigated whether resveratrol could improve MI-induced cardiac remodeling and HF in rats through the inhibition of CYP1B1 and its metabolites. To do this, rats were subjected to either sham surgery or a surgery to ligate the left anterior descending artery to induce a MI and subsequent HF. Three weeks post-surgery, rats with established HF were treated with control diet or administered a diet containing low dose of resveratrol. Our results showed that low dose resveratrol treatment significantly improves % ejection fraction in MI rats and reduces MI-induced left ventricular and atrial remodeling. Furthermore, non-cardiac symptoms of HF such as reduced physical activity improved with low dose resveratrol treatment. Mechanistically, low dose resveratrol treatment of rats with established HF restored levels of fatty acid oxidation and significantly improved cardiac energy metabolism as well as significantly inhibited CYP1B1 and cardiotoxic HETE metabolites induced in MI rats. Overall, the present work provides evidence that low dose resveratrol reduces the severity of MI-induced HF, at least in part, through the inhibition of CYP1B1 and cardiotoxic HETE metabolites.


Subject(s)
Heart Failure/drug therapy , Heart Failure/etiology , Heart Failure/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Myocardial Infarction/complications , Resveratrol/therapeutic use , Animals , Chromatography, Liquid , Male , Myocardium/metabolism , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization
6.
Am J Physiol Endocrinol Metab ; 315(4): E511-E519, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29870676

ABSTRACT

Oral administration of resveratrol attenuates several symptoms associated with the metabolic syndrome, such as impaired glucose homeostasis and hypertension. Recent work has shown that resveratrol can improve glucose homeostasis in obesity via changes in the gut microbiota. Studies involving fecal microbiome transplants (FMTs) suggest that either live gut microbiota or bacterial-derived metabolites from resveratrol ingestion are responsible for producing the observed benefits in recipients. Herein, we show that obese mice receiving FMTs from healthy resveratrol-fed mice have improved glucose homeostasis within 11 days of the first transplant, and that resveratrol-FMTs is more efficacious than oral supplementation of resveratrol for the same duration. The effects of FMTs from resveratrol-fed mice are also associated with decreased inflammation in the colon of obese recipient mice. Furthermore, we show that sterile fecal filtrates from resveratrol-fed mice are sufficient to improve glucose homeostasis in obese mice, demonstrating that nonliving bacterial, metabolites, or other components within the feces of resveratrol-fed mice are sufficient to reduce intestinal inflammation. These postbiotics may be an integral mechanism by which resveratrol improves hyperglycemia in obesity. Resveratrol-FMTs also reduced the systolic blood pressure of hypertensive mice within 2 wk of the first transplant, indicating that the beneficial effects of resveratrol-FMTs may also assist with improving cardiovascular conditions associated with the metabolic syndrome.


Subject(s)
Antioxidants/pharmacology , Blood Glucose/metabolism , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Metabolic Syndrome/metabolism , Obesity/metabolism , Resveratrol/pharmacology , Animals , Blood Glucose/drug effects , Blood Pressure , Colon/immunology , Cytokines/immunology , Diet, High-Fat , Dietary Sucrose , Hyperglycemia , Hypertension , Inflammation , Magnetic Resonance Spectroscopy , Metabolic Syndrome/immunology , Mice , Obesity/immunology
7.
Cardiovasc Res ; 114(10): 1350-1359, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29566148

ABSTRACT

Aims: Doxorubicin (DOX) is among the most effective chemotherapies used in paediatric cancer patients. However, the clinical utility of DOX is offset by its well-known cardiotoxicity, which often does not appear until later in life. Since hypertension significantly increases the risk of late-onset heart failure in childhood cancer survivors, we investigated whether juvenile DOX exposure impairs the ability to adapt to angiotensin II (Ang II)-induced hypertension later in life and tested a treatment that could prevent this. Methods and results: Five-week-old male mice were administered a low dose of DOX (4 mg/kg) or saline once a week for 3 weeks and then allowed to recover for 5 weeks. Following the 5-week recovery period, mice were infused with Ang II or saline for 2 weeks. In another cohort, mice were fed chow containing 0.4% resveratrol 1 week before, during, and 1 week after the DOX administrations. One week after the last DOX administration, p38 mitogen-activated protein kinase (MAPK) was activated in hearts of DOX-treated mice demonstrating molecular signs of cardiac stress; yet, there was no change in cardiac function between groups. However, DOX-treated mice failed to develop compensatory cardiac hypertrophy in response to Ang II-induced hypertension later in life. Of importance, mice receiving DOX with resveratrol co-administration displayed normalization in p38 MAPK activation in the heart and a restored capacity for cardiac hypertrophy in response to Ang II-induced hypertension. Conclusion: We have developed a juvenile mouse model of DOX-induced cardiotoxicity that displays no immediate overt physiological dysfunction; but, leads to an impaired ability of the heart to adapt to hypertension later in life. We also show that co-administration of resveratrol during DOX treatment was sufficient to normalize molecular markers of cardiotoxicity and restore the ability of the heart to undergo adaptive remodelling in response to hypertension later in life.


Subject(s)
Angiotensin II , Doxorubicin , Heart Diseases/prevention & control , Hypertension/prevention & control , Myocytes, Cardiac/drug effects , Resveratrol/pharmacology , Adaptation, Physiological , Animals , Blood Pressure/drug effects , Cardiotoxicity , Disease Models, Animal , Enzyme Activation , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/physiopathology , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Signal Transduction/drug effects , Time Factors , Ventricular Remodeling/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Am J Physiol Heart Circ Physiol ; 312(4): H842-H853, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28159807

ABSTRACT

We investigated whether treatment of mice with established pressure overload-induced heart failure (HF) with the naturally occurring polyphenol resveratrol could improve functional symptoms of clinical HF such as fatigue and exercise intolerance. C57Bl/6N mice were subjected to either sham or transverse aortic constriction surgery to induce HF. Three weeks postsurgery, a cohort of mice with established HF (%ejection fraction <45) was administered resveratrol (~450 mg·kg-1·day-1) or vehicle for 2 wk. Although the percent ejection fraction was similar between both groups of HF mice, those mice treated with resveratrol had increased total physical activity levels and exercise capacity. Resveratrol treatment was associated with altered gut microbiota composition, increased skeletal muscle insulin sensitivity, a switch toward greater whole body glucose utilization, and increased basal metabolic rates. Although muscle mass and strength were not different between groups, mice with HF had significant declines in basal and ADP-stimulated O2 consumption in isolated skeletal muscle fibers compared with sham mice, which was completely normalized by resveratrol treatment. Overall, resveratrol treatment of mice with established HF enhances exercise performance, which is associated with alterations in whole body and skeletal muscle energy metabolism. Thus, our preclinical data suggest that resveratrol supplementation may effectively improve fatigue and exercise intolerance in HF patients.NEW & NOTEWORTHY Resveratrol treatment of mice with heart failure leads to enhanced exercise performance that is associated with altered gut microbiota composition, increased whole body glucose utilization, and enhanced skeletal muscle metabolism and function. Together, these preclinical data suggest that resveratrol supplementation may effectively improve fatigue and exercise intolerance in heart failure via these mechanisms.


Subject(s)
Antioxidants/pharmacology , Heart Failure/drug therapy , Heart Failure/physiopathology , Muscle, Skeletal/drug effects , Physical Exertion/drug effects , Stilbenes/pharmacology , Animals , Energy Metabolism/drug effects , Exercise Tolerance/drug effects , Fatigue/prevention & control , Glucose/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Microbiota , Oxidation-Reduction , Oxygen Consumption/drug effects , Physical Conditioning, Animal , Resveratrol , Stroke Volume/drug effects
9.
Am J Physiol Heart Circ Physiol ; 312(3): H561-H570, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28062414

ABSTRACT

Since left ventricular hypertrophy (LVH) increases the susceptibility for the development of other cardiac conditions, pharmacotherapy that mitigates pathological cardiac remodeling may prove to be beneficial in patients with LVH. Previous work has shown that the activation of the energy-sensing kinase AMP-activated protein kinase (AMPK) can inhibit some of the molecular mechanisms that are involved in LVH. Of interest, metformin activates AMPK through its inhibition of mitochondrial complex I in the electron transport chain and can prevent LVH induced by pressure overload. However, metformin has additional cellular effects unrelated to AMPK activation, raising questions about whether mitochondrial complex I inhibition is sufficient to reduce LVH. Herein, we characterize the cardiac effects of a novel compound (R118), which is a more potent complex I inhibitor than metformin and is thus used at a much lower concentration. We show that R118 activates AMPK in the cardiomyocyte, inhibits multiple signaling pathways involved in LVH, and prevents Gq protein-coupled receptor agonist-induced prohypertrophic signaling. We also show that in vivo administration of R118 prevents LVH in a mouse model of hypertension, suggesting that R118 can directly modulate the response of the cardiomyocyte to stress. Of importance, we also show that while R118 treatment prevents adaptive remodelling in response to elevated afterload, it does so without compromising systolic function, improves myocardial energetics, and prevents a decline in diastolic function in hypertensive mice. Taken together, our data suggest that inhibition of mitochondrial complex I may be worthy of future investigation for the treatment of LVH.NEW & NOTEWORTHY Inhibition of mitochondrial complex I by R118 reduces left ventricular hypertrophy (LVH) and improves myocardial energetics as well as diastolic function without compromising systolic function. Together, these effects demonstrate the therapeutic potential of complex I inhibitors in the treatment of LVH, even in the presence of persistent hypertension.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Hypertension/complications , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/prevention & control , AMP-Activated Protein Kinases/metabolism , Angiotensin II , Animals , Blood Pressure , Energy Metabolism , Enzyme Activators/pharmacology , Hypertension/chemically induced , Hypertrophy, Left Ventricular/chemically induced , In Vitro Techniques , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Vasoconstrictor Agents
10.
Biochim Biophys Acta Gen Subj ; 1861(2): 313-322, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27871838

ABSTRACT

BACKGROUND: Although insulin resistance (IR) is a key factor in the pathogenesis of type 2 diabetes (T2D), the precise role of insulin in the development of IR remains unclear. Therefore, we investigated whether chronic basal insulin infusion is causative in the development of glucose intolerance. METHODS: Normoglycemic lean rats surgically instrumented with i.v. catheters were infused with insulin (3mU/kg/min) or physiological saline for 6weeks. At infusion-end, plasma insulin levels along with glucose tolerance were assessed. RESULTS: Six weeks of insulin infusion induced glucose intolerance and impaired insulin response in healthy rats. Interestingly, the effects of chronic insulin infusion were completely normalized following 24h withdrawal of exogenous insulin and plasma insulin response to glucose challenge was enhanced, suggesting improved insulin secretory capacity. As a result of this finding, we assessed whether the effects of insulin therapy followed by a washout could ameliorate established glucose intolerance in obese rats. Obese rats were similarly instrumented and infused with insulin or physiological saline for 7days followed by 24h washout. Seven day-insulin therapy in obese rats significantly improved glucose tolerance, which was attributed to improved insulin secretory capacity and improved insulin signaling in liver and skeletal muscle. CONCLUSION: Moderate infusion of insulin alone is sufficient to cause glucose intolerance and impair endogenous insulin secretory capacity, whereas short-term, intensive insulin therapy followed by insulin removal effectively improves glucose tolerance, insulin response and peripheral insulin sensitivity in obese rats. GENERAL SIGNIFICANCE: New insight into the link between insulin and glucose intolerance may optimize T2D management.


Subject(s)
Blood Glucose/drug effects , Glucose/metabolism , Insulin/administration & dosage , Obesity/blood , Obesity/metabolism , Thinness/blood , Thinness/metabolism , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/blood , Glucose Tolerance Test/methods , Insulin Resistance/physiology , Liver/drug effects , Liver/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley
11.
Diabetes ; 66(2): 418-425, 2017 02.
Article in English | MEDLINE | ID: mdl-27903747

ABSTRACT

Oral administration of resveratrol is able to improve glucose homeostasis in obese individuals. Herein we show that resveratrol ingestion produces taxonomic and predicted functional changes in the gut microbiome of obese mice. In particular, changes in the gut microbiome were characterized by a decreased relative abundance of Turicibacteraceae, Moryella, Lachnospiraceae, and Akkermansia and an increased relative abundance of Bacteroides and Parabacteroides Moreover, fecal transplantation from healthy resveratrol-fed donor mice is sufficient to improve glucose homeostasis in obese mice, suggesting that the resveratrol-mediated changes in the gut microbiome may play an important role in the mechanism of action of resveratrol.


Subject(s)
Blood Glucose/drug effects , Gastrointestinal Microbiome/drug effects , Obesity/metabolism , Stilbenes/pharmacology , Animals , Bacteroides , Blood Glucose/metabolism , Chromatography, Liquid , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/genetics , Glucose/metabolism , Glucose Tolerance Test , Homeostasis/drug effects , Male , Mice , Mice, Obese , Obesity/microbiology , Resveratrol , Tandem Mass Spectrometry
12.
Circ Heart Fail ; 8(1): 128-37, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25394648

ABSTRACT

BACKGROUND: Although resveratrol has multiple beneficial cardiovascular effects, whether resveratrol can be used for the treatment and management of heart failure (HF) remains unclear. In the current study, we determined whether resveratrol treatment of mice with established HF could lessen the detrimental phenotype associated with pressure-overload-induced HF and identified physiological and molecular mechanisms contributing to this. METHODS AND RESULTS: C57Bl/6 mice were subjected to either sham or transverse aortic constriction surgery to induce HF. Three weeks post surgery, a cohort of mice with established HF (% ejection fraction <45) was administered resveratrol (≈320 mg/kg per day). Despite a lack of improvement in ejection fraction, resveratrol treatment significantly increased median survival of mice with HF, lessened cardiac fibrosis, reduced gene expression of several disease markers for hypertrophy and extracellular matrix remodeling that were upregulated in HF, promoted beneficial remodeling, and improved diastolic function. Resveratrol treatment of mice with established HF also restored the levels of mitochondrial oxidative phosphorylation complexes, restored cardiac AMP-activated protein kinase activation, and improved myocardial insulin sensitivity to promote glucose metabolism and significantly improved myocardial energetic status. Finally, noncardiac symptoms of HF, such as peripheral insulin sensitivity, vascular function, and physical activity, were improved with resveratrol treatment. CONCLUSIONS: Resveratrol treatment of mice with established HF lessens the severity of the HF phenotype by lessening cardiac fibrosis, improving molecular and structural remodeling of the heart, and enhancing diastolic function, vascular function, and energy metabolism.


Subject(s)
Energy Metabolism/drug effects , Heart Failure, Diastolic/drug therapy , Myocardial Contraction/drug effects , Myocardium/metabolism , Stilbenes/therapeutic use , Ventricular Function, Left/drug effects , Ventricular Remodeling/physiology , Animals , Disease Models, Animal , Heart Failure, Diastolic/metabolism , Heart Failure, Diastolic/physiopathology , Male , Mice , Mice, Inbred C57BL , Resveratrol , Ribonucleotide Reductases/antagonists & inhibitors , Stroke Volume/drug effects , Vasodilator Agents/therapeutic use , Ventricular Remodeling/drug effects
13.
J Mol Med (Berl) ; 93(4): 413-25, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25394677

ABSTRACT

UNLABELLED: The mechanisms responsible for how resveratrol inhibits pathological left ventricular hypertrophy (LVH) but not physiological LVH have not been elucidated. Herein, we show that in rat cardiomyocytes, lower concentrations of resveratrol (0.1 and 1 µM) are efficient at selectively inhibiting important regulators involved in pathological LVH (such as nuclear factor of activated T cells (NFAT)) while not affecting pathways involved in physiological LVH (Akt and p70S6 kinase (p70S6K)). These differential responses are also observed in both mouse and rat models of in vivo physiological and pathological LVH. Interestingly, in all of the experiments involving a low concentration of resveratrol (1 µM), the observed effects on Akt, p70S6K, and NFAT were independent from AMP-activated protein kinase (AMPK) activation while these effects at higher concentrations of resveratrol (50 µM) were potentiated by AMPK activation. In summary, we show that resveratrol can concentration/dose selectively inhibit various pro-hypertrophic signaling pathways and that resveratrol has differential effects on the modification of these signaling cascades in response to pathological stimuli versus physiological stimuli. This has important clinical implications as our findings support the concept that resveratrol may be useful in the selective treatment of pathological LVH. KEY MESSAGE: Resveratrol differentially regulates pathological and physiological cardiac hypertrophy. Resveratrol dose selectively inhibits pathological cardiac signaling pathways. Resveratrol inhibits NFAT-dependent transcription. At low concentrations, effects of resveratrol are AMPK-independent. Resveratrol may be used to selectively treat pathological cardiac hypertrophy.


Subject(s)
Antioxidants/pharmacology , Heart Ventricles/drug effects , Hypertrophy, Left Ventricular/prevention & control , Myocytes, Cardiac/drug effects , Stilbenes/pharmacology , Ventricular Function/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Exercise , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Insulin-Like Growth Factor I/metabolism , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NFATC Transcription Factors/metabolism , Rats , Resveratrol , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects
14.
J Mol Med (Berl) ; 89(5): 459-69, 2011 May.
Article in English | MEDLINE | ID: mdl-21387178

ABSTRACT

As aging is a significant risk factor for the development of left ventricular hypertrophy and cardiovascular disease, we hypothesized that hearts from middle-aged mice may be more sensitive to the effects of a high fat (HF) diet than hearts from young mice. To investigate this, young (10-12 week old) and middle-aged (40-44 week old) male mice were fed a low fat (LF) or HF diet (10 or 60 kcal% fat, respectively) for 12 weeks. Following this 12-week period, we show that CD36 protein expression was not changed in hearts from young mice yet was increased 1.5-fold in the middle-aged HF group compared with LF-fed age-matched counterparts. Correlated with increased CD36 expression, middle-aged mice displayed a greater degree of cardiac hypertrophy compared with young mice when fed a HF diet, and this was observed in the absence of cardiac dysfunction. Furthermore, middle-aged CD36 knockout mice were protected against HF diet-induced cardiac hypertrophy, supporting a link between CD36 and cardiac hypertrophy. To further explore potential mechanisms that may explain why middle-aged mice are more susceptible to HF diet-induced cardiac hypertrophy, we investigated mediators of cardiac growth. We show that myocardial ceramide levels were significantly increased in middle-aged mice fed a HF diet compared with LF-fed controls, which was also correlated with inhibition of AMP-activated protein kinase (AMPK). Consistent with AMPK being a negative regulator of cardiac hypertrophy, decreased AMPK activity also resulted in the activation of the mTOR/p70S6K pathway, which is known to enhance protein synthesis associated with cardiac hypertrophy. Together, these data suggest that increased myocardial CD36 expression in hearts from middle-aged mice may contribute to HF diet-induced cardiac hypertrophy and that this may be mediated by elevated ceramide levels signaling through AMPK. Overall, we suggest that inhibition of CD36-mediated fatty acid uptake may prevent obesity-related cardiomyopathies in the middle-aged population.


Subject(s)
CD36 Antigens/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomyopathies/metabolism , Obesity/metabolism , Obesity/physiopathology , AMP-Activated Protein Kinases/metabolism , Aging/physiology , Animals , CD36 Antigens/genetics , Dietary Fats/adverse effects , Immunoblotting , Male , Mice
15.
J Mol Med (Berl) ; 89(3): 291-302, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21140129

ABSTRACT

Recent evidence has suggested that activation of AMP-activated protein kinase (AMPK) induced by short-term caloric restriction (CR) protects against myocardial ischemia-reperfusion (I/R) injury. Because AMPK plays a central role in regulating energy metabolism, we investigated whether alterations in cardiac energy metabolism contribute to the cardioprotective effects induced by CR. Hearts from control or short-term CR mice were subjected to ex vivo I/R and metabolism, as well as post-ischemic functional recovery was measured. Even in the presence of elevated levels of fatty acids, CR significantly improved recovery of cardiac function following ischemia. While rates of fatty acid oxidation or glycolysis from exogenous glucose were similar between groups, improved functional recovery post-ischemia in CR hearts was associated with high rates of glucose oxidation during reperfusion compared to controls. Consistent with CR improving energy supply, hearts from CR mice had increased ATP levels, as well as lower AMPK activity at the end of reperfusion compared to controls. Furthermore, in agreement with the emerging concept that CR is a non-conventional form of pre-conditioning, we observed a significant increase in phosphorylation of Akt and Erk1/2 at the end of reperfusion. These data also suggest that activation of the reperfusion salvage kinase (RISK) pathway also contributes to the beneficial effects of CR in reducing post-ischemia contractile dysfunction. These findings also suggest that short-term CR improves post-ischemic recovery by promoting glucose oxidation, and activating the RISK pathway. As such, pre-operative CR may be a clinically relevant strategy for increasing ischemic tolerance of the heart.


Subject(s)
Caloric Restriction , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Blotting, Western , Fatty Acids/blood , Fatty Acids/metabolism , Glycolysis/physiology , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Protein Kinases/metabolism , Signal Transduction/physiology
16.
J Biol Chem ; 283(35): 24194-201, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18562309

ABSTRACT

Whereas studies involving animal models of cardiovascular disease demonstrated that resveratrol is able to inhibit hypertrophic growth, the mechanisms involved have not been elucidated. Because studies in cells other than cardiomyocytes revealed that AMP-activated protein kinase (AMPK) and Akt are affected by resveratrol, we hypothesized that resveratrol prevents cardiac myocyte hypertrophy via these two kinase systems. Herein, we demonstrate that resveratrol reduces phenylephrine-induced protein synthesis and cell growth in rat cardiac myocytes via alterations of intracellular pathways involved in controlling protein synthesis (p70S6 kinase and eukaryotic elongation factor-2). Additionally, we demonstrate that resveratrol negatively regulates the calcineurin-nuclear factor of activated T cells pathway thus modifying a critical component of the transcriptional mechanism involved in pathological cardiac hypertrophy. Our data also indicate that these effects of resveratrol are mediated via AMPK activation and Akt inhibition, and in the case of AMPK, is dependent on the presence of the AMPK kinase, LKB1. Taken together, our data suggest that resveratrol exerts anti-hypertrophic effects by activating AMPK via LKB1 and inhibiting Akt, thus suppressing protein synthesis and gene transcription.


Subject(s)
Cardiomegaly/enzymology , Enzyme Inhibitors/pharmacology , Multienzyme Complexes/metabolism , Myocytes, Cardiac/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Animals , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Cardiotonic Agents/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Mice , Myocytes, Cardiac/pathology , Peptide Elongation Factor 2/metabolism , Phenylephrine/pharmacology , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Resveratrol , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Stilbenes , Transcription, Genetic/drug effects
17.
Diabetes ; 56(12): 2863-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17728375

ABSTRACT

OBJECTIVE: The etiology of type 2 diabetes often involves diet-induced obesity (DIO), which is associated with elevated plasma fatty acids and lipoprotein associated triglycerides. Since aberrant hepatic fatty acid uptake may contribute to this, we investigated whether increased expression of a fatty acid transport protein (CD36) in the liver during DIO contributes to the dyslipidemia that precedes development of type 2 diabetes. RESEARCH DESIGN AND METHODS: We determined the effect DIO has on hepatic CD36 protein expression and the functional consequence of this in terms of hepatic triglyceride storage and secretion. In addition, in vivo adenoviral gene delivery of CD36 to the livers of lean mice was performed to determine if increased hepatic CD36 protein was sufficient to alter hepatic fatty acid uptake and triglyceride storage and secretion. RESULTS: During DIO, CD36 protein levels in the liver are significantly elevated, and these elevated levels correlate with increased hepatic triglyceride storage and secretion. These alterations in liver lipid storage and secretion were also observed upon forced expression of hepatic CD36 in the absence of DIO and were accompanied with a marked rise in hepatic fatty acid uptake in vivo, demonstrating that increased CD36 expression is sufficient to recapitulate the aberrant liver lipid handling observed in DIO. CONCLUSIONS: Increased expression of hepatic CD36 protein in response to DIO is sufficient to exacerbate hepatic triglyceride storage and secretion. As these CD36-mediated effects contribute to the dyslipidemia that often precedes the development of type 2 diabetes, increased hepatic CD36 expression likely plays a causative role in the pathogenesis of type 2 diabetes.


Subject(s)
CD36 Antigens/genetics , Dyslipidemias/physiopathology , Liver/physiopathology , Obesity/physiopathology , Animals , Cells, Cultured , Energy Intake , Fatty Acids/metabolism , Glucose Tolerance Test , Hepatocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Palmitic Acid/metabolism
18.
Am J Physiol Heart Circ Physiol ; 292(3): H1460-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17098823

ABSTRACT

AMP-activated protein kinase (AMPK) is a major metabolic regulator in the cardiac myocyte. Recently, LKB1 was identified as a kinase that regulates AMPK. Using immunoblot analysis, we confirmed high expression of LKB1 in isolated rat cardiac myocytes but show that, under basal conditions, LKB1 is primarily localized to the nucleus, where it is inactive. We examined the role of LKB1 in cardiac myocytes, using adenoviruses that express LKB1, and its binding partners Ste20-related adaptor protein (STRADalpha) and MO25alpha. Infection of neonatal rat cardiac myocytes with all three adenoviruses substantially increased LKB1/STRADalpha/MO25alpha expression, LKB1 activity, and AMPKalpha phosphorylation at its activating phosphorylation site (threonine-172). Since activation of AMPK can inhibit hypertrophic growth and since LKB1 is upstream of AMPK, we hypothesized that expression of an active LKB1 complex would also inhibit protein synthesis associated with hypertrophic growth. Expression of the LKB1/STRADalpha/MO25alpha complex in neonatal rat cardiac myocytes inhibited the increase in protein synthesis observed in cells treated with phenylephrine (measured via [(3)H]phenylalanine incorporation). This was associated with a decreased phosphorylation of p70S6 kinase and its substrate S6 ribosomal protein, key regulators of protein synthesis. In addition, we show that the pathological cardiac hypertrophy in transgenic mice with cardiac-specific expression of activated calcineurin is associated with a significant decrease in LKB1 expression. Together, our data show that increased LKB1 activity in the cardiac myocyte can decrease hypertrophy-induced protein synthesis and suggest that LKB1 activation may be a method for the prevention of pathological cardiac hypertrophy.


Subject(s)
Cardiomegaly/physiopathology , Muscle Cells/physiology , Phenylephrine , Protein Serine-Threonine Kinases/physiology , AMP-Activated Protein Kinase Kinases , Adenoviridae , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cardiomegaly/prevention & control , Cells, Cultured , Heart/drug effects , Heart/physiopathology , Humans , Mice , Mice, Transgenic , Muscle Cells/cytology , Muscle Cells/drug effects , Plasmids , Protein Serine-Threonine Kinases/genetics , Rats , Recombinant Proteins/metabolism , Transfection
19.
Am J Physiol Heart Circ Physiol ; 290(6): H2472-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16428351

ABSTRACT

AMP-activated protein kinase (AMPK) plays a major role in the regulation of cardiac energy substrate utilization and can be negatively regulated by Akt activation in the heart. It has recently been shown that Akt directly phosphorylates AMPKalpha(1)/alpha(2) on Ser(485/491) in vitro and prevents the AMPK kinase (AMPKK) LKB1 from phosphorylating AMPKalpha at its primary activation site, Thr(172) (S Horman, D Vertommen, R Heath, D Neumann, V Mouton, A Woods, U Schlattner, T Wallimann, D Carling, L Hue, and MH Rider. J Biol Chem 281: 5335-5340, 2006). To determine whether this is also the case in the cardiac myocyte, neonatal rat cardiac myocytes (NRCM) were infected with a recombinant adenovirus expressing a constitutively active mutant of Akt1 (myrAkt1) and then with or without adenoviruses expressing the active LKB1 complex. Expression of myrAkt1 blunted LKB1-induced phosphorylation of AMPKalpha at Thr(172), which resulted in a dramatic decrease in phosphorylation of AMPK's target, acetyl CoA-carboxylase. This decrease in AMPK activity was associated with prior Akt1-dependent phosphorylation of AMPKalpha(1)/alpha(2) at Ser(485/491). To investigate whether Akt1 activation was also able to prevent other AMPKKs from phosphorylating AMPKalpha, we subjected NRCM to chemical hypoxia and noted a marked increase in phosphorylation of AMPKalpha at Thr(172), despite no change in LKB1 activity. NRCM expressing myrAkt1 demonstrated increased phosphorylation of AMPKalpha(1)/alpha(2) at Ser(485/491) and a complete inhibition of chemical hypoxia-induced phosphorylation of AMPKalpha at Thr(172). Taken together, our data show that activation of Akt1 is able to prevent activation of cardiac AMPK by LKB1 and at least one other AMPKK, likely by prior phosphorylation of AMPKalpha(1)/alpha(2) at Ser(485/491).


Subject(s)
Hypoxia/chemically induced , Myocardium/enzymology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins c-akt/physiology , AMP-Activated Protein Kinase Kinases , Adenoviridae/enzymology , Animals , Animals, Newborn , Antimetabolites , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/physiology , Hypoxia/metabolism , Myocytes, Cardiac/enzymology , Phosphorylation , Proto-Oncogene Proteins c-akt/biosynthesis , Rats
20.
J Biol Chem ; 279(31): 32771-9, 2004 Jul 30.
Article in English | MEDLINE | ID: mdl-15159410

ABSTRACT

A necessary mediator of cardiac myocyte enlargement is protein synthesis, which is controlled at the levels of both translation initiation and elongation. Eukaryotic elongation factor-2 (eEF2) mediates the translocation step of peptide-chain elongation and is inhibited through phosphorylation by eEF2 kinase. In addition, p70S6 kinase can regulate protein synthesis by phosphorylating eEF2 kinase or via phosphorylation of ribosomal protein S6. We have recently shown that eEF2 kinase is also controlled by phosphorylation by AMP-activated protein kinase (AMPK), a key regulator of cellular energy homeostasis. Moreover, the mammalian target of rapamycin has also been shown to be inhibited, indirectly, by AMPK, thus leading to the inhibition of p70S6 kinase. Although AMPK activation has been shown to modulate protein synthesis, it is unknown whether AMPK could also be a regulator of cardiac hypertrophic growth. Therefore, we investigated the role of AMPK activation in regulating protein synthesis during both phenylephrine- and Akt-induced cardiac hypertrophy. Metformin and 5-aminoimidazole-4-carboxamide 1-beta-D-ribofuranoside were used to activate AMPK in neonatal rat cardiac myocytes. Activation of AMPK significantly decreased protein synthesis induced by phenylephrine treatment or by expression of constitutively active Akt. Activation of AMPK also resulted in decreased p70S6 kinase phosphorylation and increased phosphorylation of eEF2, suggesting that inhibition of protein synthesis involves the eEF2 kinase/eEF2 axis and/or the p70S6 kinase pathway. Together, our data suggest that the inhibition of protein synthesis by pharmacological activation of AMPK may be a key regulatory mechanism by which hypertrophic growth can be controlled.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Multienzyme Complexes/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Animals, Newborn , Antibiotics, Antineoplastic/pharmacology , Cells, Cultured , Elongation Factor 2 Kinase , Enzyme Activation , Green Fluorescent Proteins , Hypertrophy , Hypoglycemic Agents/pharmacology , Immunoblotting , Luminescent Proteins/metabolism , Metformin/metabolism , Metformin/pharmacology , Microscopy, Fluorescence , Phenylephrine/pharmacology , Phosphorylation , Rats , Ribonucleotides/pharmacology , Ribose/analogs & derivatives , Ribosomal Protein S6/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL