Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Respir Med ; 227: 107658, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704051

ABSTRACT

Pulmonary hypertension (PH) in chronic obstructive pulmonary disease (COPD) is associated with worse clinical outcomes and decreased survival rates. In absence of disease specific diagnostic/therapeutic targets and unclear pathophysiology, there is an urgent need for the identification of potential genetic/molecular markers and disease associated pathways. The present study aims to use a bioinformatics approach to identify and validate hypoxia-associated gene signatures in COPD-PH patients. Additionally, hypoxia-related inflammatory profile is also explored in these patients. Microarray dataset obtained from the Gene Expression Omnibus repository was used to identify differentially expressed genes (DEGs) in a hypoxic PH mice model. The top three hub genes identified were further validated in COPD-PH patients, with chemokine (C-X-C motif) ligand 9 (CXCL9) and CXCL12 showing significant changes in comparison to healthy controls. Furthermore, multiplexed analysis of 10 inflammatory cytokines, tumor necrosis factor alpha (TNF-α), transforming growth factor ß (TGF-ß), interleukin 1-beta (IL-1ß), IL-4, IL-5, IL-6, IL-13, IL-17, IL-18 and IL-21 was also performed. These markers showed significant changes in COPD-PH patients as compared to controls. They also exhibited the ability to differentially diagnose COPD-PH patients in comparison to COPD. Additionally, IL-6 and IL-17 showed significant positive correlation with systolic pulmonary artery pressure (sPAP). This study is the first report to assess the levels of CXCL9 and CXCL12 in COPD-PH patients and also explores their link with the inflammatory profile of these patients. Our findings could be extended to better understand the underlying disease mechanism and possibly used for tailoring therapies exclusive for the disease.


Subject(s)
Chemokine CXCL12 , Computational Biology , Cytokines , Hypertension, Pulmonary , Hypoxia , Pulmonary Disease, Chronic Obstructive , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Cytokines/metabolism , Cytokines/genetics , Computational Biology/methods , Humans , Hypoxia/genetics , Hypoxia/metabolism , Animals , Mice , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Hypertension, Pulmonary/genetics , Chemokine CXCL9/genetics , Gene Expression Profiling , Male , Female , Disease Models, Animal , Inflammation/genetics , Inflammation/metabolism , Middle Aged
2.
J Cardiovasc Pharmacol ; 82(3): 180-188, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37341530

ABSTRACT

ABSTRACT: Cardiovascular diseases (CVDs) are the foremost cause of morbidity and mortality worldwide. Current clinical interventions include invasive approaches for progressed conditions and pharmacological assistance for initial stages, which has systemic side effects. Preventive, curative, diagnostic, and theranostic (therapeutic + diagnostic) approaches till date are not very useful in combating the ongoing CVD epidemic, which demands a promising efficient alternative approach. To combat the growing CVD outbreak globally, the ideal strategy is to make the therapeutic intervention least invasive and direct to the heart to reduce the bystander effects on other organs and increase the bioavailability of the therapeutics to the myocardium. The application of nanoscience and nanoparticle-mediated approaches have gained a lot of momentum because of their efficient passive and active myocardium targeting capability owing to their improved specificity and controlled release. This review provides extensive insight into the various types of nanoparticles available for CVDs, their mechanisms of targeting (eg, direct or indirect), and the utmost need for further development of bench-to-bedside cardiac tissue-based nanomedicines. Furthermore, the review aims to summarize the different ideas and methods of nanoparticle-mediated therapeutic approaches to the myocardium till date with present clinical trials and future perspectives. This review also reflects the potential of such nanoparticle-mediated tissue-targeted therapies to contribute to the sustainable development goals of good health and well-being.


Subject(s)
Cardiovascular Diseases , Heart Diseases , Humans , Myocardium , Heart Diseases/diagnosis , Heart Diseases/drug therapy , Nanomedicine , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Heart
3.
Eur J Pharmacol ; 953: 175841, 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37329972

ABSTRACT

Pathological cardiac hypertrophy is associated with ventricular fibrosis leading to heart failure. The use of thiazolidinediones as Peroxisome Proliferator-Activated Receptor-gamma (PPARγ)-modulating anti-hypertrophic therapeutics has been restricted due to major side-effects. The present study aims to evaluate the anti-fibrotic potential of a novel PPARγ agonist, deoxyelephantopin (DEP) in cardiac hypertrophy. AngiotensinII treatment in vitro and renal artery ligation in vivo were performed to mimic pressure overload-induced cardiac hypertrophy. Myocardial fibrosis was evaluated by Masson's trichrome staining and hydroxyproline assay. Our results showed that DEP treatment significantly improves the echocardiographic parameters by ameliorating ventricular fibrosis without any bystander damage to other major organs. Following molecular docking, all-atomistic molecular dynamics simulation, reverse transcription-polymerase chain reaction and immunoblot analyses, we established DEP as a PPARγ agonist stably interacting with the ligand-binding domain of PPARγ. DEP specifically downregulated the Signal Transducer and Activator of Transcription (STAT)-3-mediated collagen gene expression in a PPARγ-dependent manner, as confirmed by PPARγ silencing and site-directed mutagenesis of DEP-interacting PPARγ residues. Although DEP impaired STAT-3 activation, it did not have any effect on the upstream Interleukin (IL)-6 level implying possible crosstalk of the IL-6/STAT-3 axis with other signaling mediators. Mechanistically, DEP increased the binding of PPARγ with Protein Kinase C-delta (PKCδ) which impeded the membrane translocation and activation of PKCδ, downregulating STAT-3 phosphorylation and resultant fibrosis. This study, therefore, for the first time demonstrates DEP as a novel cardioprotective PPARγ agonist. The therapeutic potential of DEP as an anti-fibrotic remedy can be exploited against hypertrophic heart failure in the future.


Subject(s)
Heart Failure , PPAR gamma , Humans , PPAR gamma/metabolism , Interleukin-6 , PPAR-gamma Agonists , Molecular Docking Simulation , Cardiomegaly/pathology , Fibrosis
4.
Cell Signal ; 98: 110403, 2022 10.
Article in English | MEDLINE | ID: mdl-35835332

ABSTRACT

IKKγ prototypically promotes NFκBp65 activity by regulating the assembly of the IKK holocomplex. In hypertrophied cardiomyocytes, the p65-p300 complex-induced regenerative efforts are neutralized by the p53-p300 complex-mediated apoptotic load resulting in compromised cardiac function. The present study reports that nitrosative stress leads to S-Nitrosylation of IKKγ in hypertrophied cardiomyocytes in a pre-clinical model. Using a cardiomyocyte-targeted nanoconjugate, IKKγ S-Nitrosylation-resistant mutant plasmids were delivered to the pathologically hypertrophied heart that resulted in improved cardiac function by amelioration of cardiomyocyte apoptosis and simultaneous induction of their cell cycle re-entry machinery. Mechanistically, in IKKγ S-Nitrosyl mutant-transfected hypertrophied cells, increased IKKγ-p300 binding downregulated the binding of p53 and p65 with p300. This shifted the binding preference of p65 from p300 to HDAC1 resulting in upregulated expression of cyclin D1 and CDK2 via the p27/pRb pathway. This approach has therapeutic advantage over mainstream anti-hypertrophic remedies which concomitantly reduce the regenerative prowess of resident cardiomyocytes during hypertrophy upon downregulation of myocyte apoptosis. Therefore, cardiomyocyte-targeted delivery of IKKγ S-Nitrosyl mutants during hypertrophy can be exploited as a novel strategy to re-muscularize the diseased heart.


Subject(s)
I-kappa B Kinase , Myocytes, Cardiac , Cardiomegaly/pathology , Humans , I-kappa B Kinase/metabolism , Myocytes, Cardiac/metabolism , Nitrosative Stress , Tumor Suppressor Protein p53/metabolism
5.
Biochem Biophys Res Commun ; 537: 100-108, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33388412

ABSTRACT

HIF-1α acts as the cellular rheostat for oxygen sensing in cardiomyocytes. Overexpression of HIF-1α in the heart during acute myocardial infarction (MI) is known to attenuate cardiac dysfunction by upregulating pro-angiogenic HIF-1α target genes. However, the effect of HIF-1α overexpression on hypoxic cardiomyocyte apoptosis still remains obscure. In this study, we report for the first time that myocardium-targeted nanotized HIF-1α overexpression during MI downregulates cardiomyocyte apoptosis. HIF-1α overexpression attenuates bnip3-mediated apoptosis indirectly by promoting HO-1-induced anti-oxidant response. Chromatin immunoprecipitation experiment revealed that HIF-1α overexpression in hypoxic cardiomyocytes increases binding of HIF-1α to the hypoxia-responsive element in the promoter of its target anti-oxidant gene ho-1 which is known to attenuate ROS accumulation. ROS accumulation in hypoxic cardiomyocytes causes cysteine oxidation of the DNA-binding p50 subunit of NFκB, which hampers NFκB binding to κB-responsive genes like bnip3. Downregulated oxidative stress due to HIF-1α overexpression leads to decline in cysteine oxidation of NFκBp50, causing NFκB to bind to the promoter of bnip3 as a transcriptional repressor. Therefore HIF-1α overexpression-mediated attenuation of cardiomyocyte apoptosis occurs by transcriptional repression of bnip3 by NFκB. Our study thus reveals that downregulation of bnip3-mediated cardiomyocyte apoptosis occurs via ho-1 upregulation upon HIF-1α overexpression during MI, despite both being HIF-1α target genes. The cross-regulation of HIF-1α and NFκB-mediated pathways effectively downregulates apoptosis due to HIF-1α overexpression during MI, which can be exploited for possible therapeutic intervention.


Subject(s)
Antioxidants/metabolism , Apoptosis/genetics , Gene Expression Regulation , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Animals , Down-Regulation/genetics , Heme Oxygenase-1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , NF-kappa B/metabolism , Organ Specificity , Oxidation-Reduction , Oxidative Stress , Rats, Wistar , Up-Regulation/genetics
8.
Sci Rep ; 10(1): 10662, 2020 Jun 25.
Article in English | MEDLINE | ID: mdl-32581244

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
Arch Biochem Biophys ; 690: 108430, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32473132

ABSTRACT

BACKGROUND: The severity and duration of hypoxia is known to determine apoptotic fate in heart; however, its implication during myocardial infarction (MI) remains unaddressed. Therefore the aim of the study was to determine apoptotic regulation in cardiomyocytes under varied hypoxic intensity and duration and to unravel the role of HIF-1α in such modulation. METHODS: Treatment of cardiomyocytes to varied hypoxic intensity and duration was carried out in vitro, which was mimicked in vivo by dose-dependent Isoproterenol hydrochloride treatment for varied time-points. Myocardium-targeted HIF-1α knockdown in vivo was performed to decipher its role in cardiomyocyte apoptosis under varied stress. Signaling intermediates were analyzed by RT-PCR, immunoblotting and co-immunoprecipitation. DCFDA-based ROS assay, Griess assay for NO release and biochemical assays for estimating caspase activity were performed. RESULTS: Severe stress resulted in cardiomyocyte apoptosis in both shorter and longer time-points. Moderate stress, on the other hand, induced apoptosis only in the shorter time-point which was downregulated in the longer time-point. ROS activity was upregulated under severe hypoxic stress for both time-points and only in the early time-point under moderate hypoxia. Increased ROS accumulation activated ERK-1/2 which stabilized nuclear HIF-1α, promoting bnip3-mediated apoptosis. Stable HSP90-IRE-1 association in such cells caused elevated endoplasmic reticulum stress-related caspase-12 activity. Sustained moderate hypoxia caused decline in ROS activity, but upregulated NFκB-dependent NO generation. NO-stabilized HIF-1α was predominantly cytosolic, since low ROS levels downregulated ERK-1/2 activity, thereby suppressing bnip3 expression. Cytosolic HIF-1α in such cells sequestered HSP90 from IRE-1, downregulating caspase-12 activity due to proteasomal degradation of IRE-1. Accordingly, myocardium-specific in vivo silencing of HIF-1α was beneficial at both time-points under severe stress as also for lesser duration of moderate stress. However, silencing of HIF-1α aggravated apoptotic injury during sustained moderate stress. CONCLUSION: ROS-mediated HIF-1α stabilization promotes cardiomyocyte apoptosis on one hand while NO-mediated stabilization of HIF-1α disrupts apoptosis depending upon the severity and duration of hypoxia. Therefore the outcome of modulation of cardiac HIF-1α activity is regulated by both the severity and duration of ischemic stress.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Myocardial Infarction/physiopathology , Animals , Apoptosis , Caspase 12/metabolism , Cell Hypoxia , Cell Line , Endoplasmic Reticulum Stress , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Humans , Mutation , Myocytes, Cardiac/cytology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism , Severity of Illness Index , Signal Transduction , Time Factors , Transfection , NF-kappaB-Inducing Kinase
12.
Sci Rep ; 10(1): 598, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953451

ABSTRACT

Myocardial hypertrophy, an inflammatory condition of cardiac muscles is a maladaptive response of the heart to biomechanical stress, hemodynamic or neurohormonal stimuli. Previous studies indicated that knockout of Arginyltransferase (ATE1) gene in mice and embryos leads to contractile dysfunction, defective cardiovascular development, and impaired angiogenesis. Here we found that in adult rat model, downregulation of ATE1 mitigates cardiac hypertrophic, cardiac fibrosis as well as apoptosis responses in the presence of cardiac stress i.e. renal artery ligation. On contrary, in wild type cells responding to renal artery ligation, there is an increase of cellular ATE1 protein level. Further, we have shown the cardioprotective role of ATE1 silencing is mediated by the interruption of TAK1 activity-dependent JNK1/2 signaling pathway. We propose that ATE1 knockdown in presence of cardiac stress performs a cardioprotective action and the inhibition of its activity may provide a novel approach for the treatment of cardiac hypertrophy.


Subject(s)
Aminoacyltransferases/genetics , Cardiomegaly/genetics , MAP Kinase Kinase Kinases/metabolism , Myocytes, Cardiac/cytology , Signal Transduction , Animals , Apoptosis , Cardiomegaly/pathology , Cell Line , Disease Models, Animal , Fibrosis , Gene Knockdown Techniques , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar
13.
J Mol Cell Cardiol ; 139: 148-163, 2020 02.
Article in English | MEDLINE | ID: mdl-31958467

ABSTRACT

Chronic pressure overload-induced left ventricular hypertrophy in heart is preceded by a metabolic perturbation that prefers glucose over lipid as substrate for energy requirement. Here, we establish C/EBPß (CCAAT/enhancer-binding protein ß) as an early marker of the metabolic derangement that triggers the imbalance in fatty acid (FA) oxidation and glucose uptake with increased lipid accumulation in cardiomyocytes during pathological hypertrophy, leading to contractile dysfunction and endoplasmic reticulum (ER) stress. This is the first study that shows that myocardium-targeted C/EBPß knockdown prevents the impaired cardiac function during cardiac hypertrophy led by maladaptive metabolic response with persistent hypertrophic stimuli, whereas its targeted overexpression in control increases lipid accumulation significantly compared to control hearts. A new observation from this study was the dual and opposite transcriptional regulation of the alpha and gamma isoforms of Peroxisomal proliferator activated receptors (PPARα and PPARγ) by C/EBPß in hypertrophied cardiomyocytes. Before the functional and structural remodeling sets in the diseased myocardium, C/EBPß aggravates lipid accumulation with the aid of the increased FA uptake involving induced PPARγ expression and decreased fatty acid oxidation (FAO) by suppressing PPARα expression. Glucose uptake into cardiomyocytes was greatly increased by C/EBPß via PPARα suppression. The activation of mammalian target of rapamycin complex-1 (mTORC1) during increased workload in presence of glucose as the only substrate was prevented by C/EBPß knockdown, thereby abating contractile dysfunction in cardiomyocytes. Our study thus suggests that C/EBPß may be considered as a novel cellular marker for deranged metabolic milieu before the heart pathologically remodels itself during hypertrophy.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Heart/physiopathology , Adenosine Triphosphate/metabolism , Animals , Biomarkers/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Cardiomegaly/genetics , Cardiomegaly/pathology , Fatty Acids/metabolism , Gene Expression Regulation , Glucose/metabolism , Lipid Metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Myocardium/metabolism , Oxidation-Reduction , Oxygen Consumption , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Rats, Wistar , Stress, Physiological
15.
Biochim Biophys Acta Mol Basis Dis ; 1865(10): 2736-2752, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31299218

ABSTRACT

Altered cardiac adaptation of physiologically hypertrophied heart during detraining remained obscure for long time. We had previously reported the switching of protein kinase C (PKC) isoforms (-α to -δ) associated with functional deterioration of heart at detraining in mice undergone swim exercise. Here we report that, myocardium targeted overexpression of insulin-like growth factor 1 (IGF1) and knockdown of insulin-like growth factor 1 receptor (IGF1R) during detraining and exercise respectively altered the activation of PKCs and eventual cardiac condition. Moreover, downregulation of mammalian target of rapamycin complex 2 (mTORC2) was recorded in both IGF1R knockdown or detraining groups. Additionally, knocking down of mTORC2 during exercise exhibited impaired cardiac condition. Interestingly, significantly increased interactions of mTORC2 with both PKCα and δ was recorded exclusively in exercise group. This interaction resulted into hydrophobic motif phosphorylation of both PKCs (Serine657-PKCα; Serine662-PKCδ). Serine phosphorylation on one hand activated PKCα mediated cell survival and on the other hand alleviated the apoptotic activity of PKCδ during exercise. Mutation of Serine662 of PKCδ in exercised mice showed higher Tyrosine311 phosphorylation with increased apoptotic load similar to that in detrained animals. These observations confirmed that differential and conditional activation of PKCs depend upon IGF1 induced mTORC2 activation. Furthermore, blocking of PKCα resulted in activated p53 which in turn repressed IGF1 expression during swim, mimicking the condition of detrained heart. In conclusion, this is the first report to unravel the intricate molecular mechanism of switching a physiologically hypertrophied heart to a pathologically hypertrophied heart during exercise withdrawal.


Subject(s)
Heart Diseases/metabolism , Insulin-Like Growth Factor I/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Myocytes, Cardiac/metabolism , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/metabolism , Animals , Disease Models, Animal , Down-Regulation , Gene Expression Regulation , Insulin-Like Growth Factor I/genetics , Male , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Transcriptome
16.
Chemosphere ; 230: 51-58, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31102871

ABSTRACT

Pesticide stress is one of the important factors for global bee declines. Apart from physiological and developmental anomalies, pesticides also impose cognitive damages on bees. The present study investigates the visual acuity of wild populations of honey bees, in an agricultural intensification landscape, and corroborates the findings with controlled laboratory experiments. Even though overall morphometric examinations revealed no significant differences between the populations, correct color choices by bees in pesticide exposed populations were significantly reduced. The study reports, for the first time, the significant reduction in ommatidia facet diameter in these populations, as viewed under scanning electron microscope, along with the molecular underpinnings to these findings. Western blot studies revealed a significant reduction in expression of two visual proteins - blue-sensitive opsin and rhodopsin - in the pesticide exposed populations in both field and laboratory conditions. The novel findings from this study form the basis for further investigations into the effects of field realistic doses of multiple pesticide exposures on wild populations of honey bees.


Subject(s)
Bees/embryology , Eye Abnormalities/chemically induced , Eye Abnormalities/embryology , Eye/embryology , Pesticides/toxicity , Visual Acuity/drug effects , Agriculture , Animals , Bees/drug effects , Microscopy, Electron, Scanning , Opsins/biosynthesis , Rhodopsin/biosynthesis
17.
Sci Rep ; 9(1): 5872, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30971740

ABSTRACT

Cardiorenal syndrome is defined by primary heart failure conditions influencing or leading to renal injury or dysfunction. Dilated cardiomyopathy (DCM) is a major co-existing form of heart failure (HF) with renal diseases. Myocardin (MYOCD), a cardiac-specific co-activator of serum response factor (SRF), is increased in DCM porcine and patient cardiac tissues and plays a crucial role in the pathophysiology of DCM. Inhibiting the increased MYOCD has shown to be partially rescuing the DCM phenotype in porcine model. However, expression levels of MYOCD in the cardiac tissues of the cardiorenal syndromic patients and the effect of inhibiting MYOCD in a cardiorenal syndrome model remains to be explored. Here, we analyzed the expression levels of MYOCD in the DCM patients with and without renal diseases. We also explored, whether cardiac specific silencing of MYOCD expression could ameliorate the cardiac remodeling and improve cardiac function in a renal artery ligated rat model (RAL). We observed an increase in MYOCD levels in the endomyocardial biopsies of DCM patients associated with renal failure compared to DCM alone. Silencing of MYOCD in RAL rats by a cardiac homing peptide conjugated MYOCD siRNA resulted in attenuation of cardiac hypertrophy, fibrosis and restoration of the left ventricular functions. Our data suggest hyper-activation of MYOCD in the pathogenesis of the cardiorenal failure cases. Also, MYOCD silencing showed beneficial effects by rescuing cardiac hypertrophy, fibrosis, size and function in a cardiorenal rat model.


Subject(s)
Cardio-Renal Syndrome/pathology , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Angiotensin II/pharmacology , Animals , Cardio-Renal Syndrome/metabolism , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Heart Ventricles/pathology , Male , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Rats , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Ventricular Function
18.
Antioxid Redox Signal ; 30(5): 713-732, 2019 02 10.
Article in English | MEDLINE | ID: mdl-29631413

ABSTRACT

AIMS: Metabolic remodeling of cardiac muscles during pathological hypertrophy is characterized by downregulation of fatty acid oxidation (FAO) regulator, peroxisome proliferator-activated receptor alpha (PPARα). Thereby, we hypothesized that a cardiac-specific induction of PPARα might restore the FAO-related protein expression and resultant energy deficit. In the present study, consequences of PPARα augmentation were evaluated for amelioration of chronic oxidative stress, myocyte apoptosis, and cardiac function during pathological cardiac hypertrophy. RESULTS: Nanotized PPARα overexpression targeted to myocardium was done by a stearic acid-modified carboxymethyl-chitosan (CMC) conjugated to a 20-mer myocyte-targeted peptide (CMCP). Overexpression of PPARα ameliorated pathological hypertrophy and improved cardiac function. Augmented PPARα in hypertrophied myocytes revealed downregulated p53 acetylation (lys 382), leading to reduced apoptosis. Such cells showed increased binding of PPARα with p53 that in turn reduced interaction of p53 with glycogen synthase kinase-3ß (GSK3ß), which upregulated inactive phospho-GSK3ß (serine [Ser]9) expression within mitochondrial protein fraction. Altogether, the altered molecular milieu in PPARα-overexpressed hypertrophy groups restored mitochondrial structure and function both in vitro and in vivo. INNOVATION: Cardiomyocyte-targeted overexpression of a protein of interest (PPARα) by nanotized plasmid has been described for the first time in this study. Our data provide a novel insight towards regression of pathological hypertrophy by ameliorating mitochondrial oxidative stress in targeted PPARα-overexpressed myocardium. CONCLUSION: PPARα-overexpression during pathological hypertrophy showed substantial betterment of mitochondrial structure and function, along with downregulated apoptosis. Myocardium-targeted overexpression of PPARα during pathological cardiac hypertrophy led to an overall improvement of cardiac energy deficit and subsequent cardiac function, thereby, opening up a potential avenue for cardiac tissue engineering during hypertrophic cardiac pathophysiology.


Subject(s)
Cardiomegaly/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Mitochondria/pathology , Myocardium/metabolism , Nanoparticles/metabolism , PPAR alpha/biosynthesis , Tumor Suppressor Protein p53/metabolism , Animals , Humans , Mitochondria/metabolism , Nanoparticles/chemistry , Oxidative Stress , PPAR alpha/chemistry , PPAR alpha/genetics
19.
J Insect Sci ; 18(3)2018 May 01.
Article in English | MEDLINE | ID: mdl-29762714

ABSTRACT

Pesticides have been reported to be one of the major drivers in the global pollinator losses. The large-scale decline in honey bees, an important pollinator group, has resulted in comprehensive studies on honey bee colonies. Lack of information on native wild pollinators has paved the way for this study, which highlights the underlying evolutionary changes occurring in the wild honey bee populations exposed to pesticides along an agricultural intensification landscape. The study reports an increased genetic diversity in native Apis cerana Fabricius (Hymenoptera: Apidae) populations continually exposed to pesticide stress. An increased heterozygosity, evidenced by a higher electrophoretic banding pattern, was observed in the pesticide-exposed populations for two isozymes involved with xenobiotic metabolism-esterase and glucose-6-phosphate dehydrogenase. Differential banding patterns also revealed a higher percentage of polymorphic loci, number of polymorphic bands, Nei's genetic distance, etc. observed in these populations in the Randomly Amplified Polymorphic DNA-Polymerase Chain Reaction (RAPD-PCR) experiments using three random decamer primers. Higher heterozygosity, being indicative of a more resistant population, implies population survival within the threshold pesticide stress. This study reports such changes for the first time in native wild Indian honey bee populations exposed to pesticides and has far-reaching implications on the population adaptability under pesticide stress.


Subject(s)
Adaptation, Physiological , Bees/genetics , Genetic Variation , Pesticides , Stress, Physiological , Animals , Esterases/genetics , Glucose-6-Phosphatase/genetics , India , Random Amplified Polymorphic DNA Technique
20.
J Cell Biochem ; 119(2): 1575-1590, 2018 02.
Article in English | MEDLINE | ID: mdl-28771799

ABSTRACT

Pathological hypertrophy and myocardial infarction (MI) are two etiologically different cardiac disorders having differential molecular mechanisms of disease manifestation. However, no study has been conducted so far to analyze and compare the differential status of energy metabolism in these two disease forms. It was shown recently by our group that production of ATP is significantly impaired during MI along with inhibition of pyruvate dehydrogenase E1-ß (PDHE1 B) by pyruvate dehydrogenase kinase 4 (PDK4). However, the ATP levels showed no significant change during pathological hypertrophy compared to control group. To seek a plausible explanation of this phenomenon, the peroxisome proliferator-activated receptor alpha (PPAR) pathway was studied in all the experimental groups which revealed that PGC1α- ERRα axis remains active in MI while the same remained inactive during pathological hypertrophy possibly by NF-κB that plays a significant role in deactivating this pathway during hypertrophy. At the same time, it was observed that reactive oxygen species (ROS) negatively regulates NF-κB activity during MI by oxidation of cysteine residues of p50- the DNA binding subunit of NF-κB. Thus, this study reports for the first time, a possible mechanism for the differential status of energy metabolism during two etiologically different cardiac pathophysiological conditions involving PGC1α-ERRα axis along with p50 subunit of NF-κB.


Subject(s)
Adenosine Triphosphate/metabolism , Cardiomegaly/metabolism , Myocardial Infarction/metabolism , NF-kappa B p50 Subunit/metabolism , Reactive Oxygen Species/metabolism , Animals , Disease Models, Animal , Energy Metabolism , Humans , Male , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats , Rats, Wistar , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...