Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Life Sci ; 347: 122651, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38642844

ABSTRACT

Calcium ion (Ca2+) dysregulation is one of the main causes of neuronal cell death and brain damage after cerebral ischemia. During ischemic stroke, the ability of neurons to maintain Ca2+ homeostasis is compromised. Ca2+ regulates various functions of the nervous system, including neuronal activity and adenosine triphosphate (ATP) production. Disruptions in Ca2+ homeostasis can trigger a cascade of events, including activation of the unfolded protein response (UPR) pathway, which is associated with endoplasmic reticulum (ER) stress and mitochondrial dysfunction. This response occurs when the cell is unable to manage protein folding within the ER due to various stressors, such as a high influx of Ca2+. Consequently, the UPR is initiated to restore ER function and alleviate stress, but prolonged activation can lead to mitochondrial dysfunction and, ultimately, cell death. Hence, precise regulation of Ca2+ within the cell is mandatory. The ER and mitochondria are two such organelles that maintain intracellular Ca2+ homeostasis through various calcium-operating channels, including ryanodine receptors (RyRs), inositol trisphosphate receptors (IP3Rs), sarco/endoplasmic reticulum calcium ATPases (SERCAs), the mitochondrial Na+/Ca2+ exchanger (NCLX), the mitochondrial calcium uniporter (MCU) and voltage-dependent anion channels (VDACs). These channels utilize Ca2+ sequestering and release mechanisms to maintain intracellular Ca2+ homeostasis and ensure proper cellular function and survival. The present review critically evaluates the significance of Ca2+ and its physiological role in cerebral ischemia. We have compiled recent findings on calcium's role and emerging treatment strategies, particularly targeting mitochondria and the endoplasmic reticulum, to address Ca2+ overload in cerebral ischemia.


Subject(s)
Calcium , Cell Death , Ischemic Stroke , Neurons , Humans , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Animals , Calcium/metabolism , Neurons/metabolism , Neurons/pathology , Mitochondria/metabolism , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/metabolism , Brain Ischemia/metabolism , Brain Ischemia/pathology , Unfolded Protein Response , Calcium Signaling/physiology , Homeostasis
2.
Inflammopharmacology ; 32(1): 287-305, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37991660

ABSTRACT

Pulmonary fibrosis is a devastating disorder distinguished by redundant inflammation and matrix accumulation in the lung interstitium. The early inflammatory cascade coupled with recurring tissue injury orchestrates a set of events marked by perturbed matrix hemostasis, deposition of matrix proteins, and remodeling in lung tissue. Numerous investigations have corroborated a direct correlation between the NLR family pyrin domain-containing 3 (NLRP3) activation and the development of pulmonary fibrosis. Dysregulated activation of NLRP3 within the pulmonary microenvironment exacerbates inflammation and may incite fibrogenic responses. Nevertheless, the precise mechanisms through which the NLRP3 inflammasome elicits pro-fibrogenic responses remain inadequately defined. Contemporary findings suggest that the pro-fibrotic consequences stemming from NLRP3 signaling primarily hinge on the action of interleukin-1ß (IL-1ß). IL-1ß instigates IL-1 receptor signaling, potentiating the activity of transforming growth factor-beta (TGF-ß). This signaling cascade, in turn, exerts influence over various transcription factors, including SNAIL, TWIST, and zinc finger E-box-binding homeobox 1 (ZEB 1/2), which collectively foster myofibroblast activation and consequent lung fibrosis. Here, we have connected the dots to illustrate how the NLRP3 inflammasome orchestrates a multitude of signaling events, including the activation of transcription factors that facilitate myofibroblast activation and subsequent lung remodeling. In addition, we have highlighted the prominent role played by various cells in the formation of myofibroblasts, the primary culprit in lung fibrosis. We also provided a concise overview of various compounds that hold the potential to impede NLRP3 inflammasome signaling, thus offering a promising avenue for the treatment of pulmonary fibrosis.


Subject(s)
Inflammasomes , Pulmonary Fibrosis , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pulmonary Fibrosis/drug therapy , Inflammation , Transcription Factors
3.
ACS Chem Neurosci ; 15(1): 31-55, 2024 01 03.
Article in English | MEDLINE | ID: mdl-38118278

ABSTRACT

Neuroinflammation is a key pathophysiological feature of stroke-associated brain injury. A local innate immune response triggers neuroinflammation following a stroke via activating inflammasomes. The nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome has been heavily implicated in stroke pathobiology. Following a stroke, several stimuli have been suggested to trigger the assembly of the NLRP3 inflammasome. Recent studies have advanced the understanding and revealed several new players regulating NLRP3 inflammasome-mediated neuroinflammation. This article discussed recent advancements in NLRP3 assembly and highlighted stroke-induced mitochondrial dysfunction as a major checkpoint to regulating NLRP3 activation. The NLRP3 inflammasome activation leads to caspase-1-dependent maturation and release of IL-1ß, IL-18, and gasdermin D. In addition, genetic or pharmacological inhibition of the NLRP3 inflammasome activation and downstream signaling has been shown to attenuate brain infarction and improve the neurological outcome in experimental models of stroke. Several drug-like small molecules targeting the NLRP3 inflammasome are in different phases of development as novel therapeutics for various inflammatory conditions, including stroke. Understanding how these molecules interfere with NLRP3 inflammasome assembly is paramount for their better optimization and/or development of newer NLRP3 inhibitors. In this review, we summarized the assembly of the NLRP3 inflammasome and discussed the recent advances in understanding the upstream regulators of NLRP3 inflammasome-mediated neuroinflammation following stroke. Additionally, we critically examined the role of the NLRP3 inflammasome-mediated signaling in stroke pathophysiology and the development of therapeutic modalities to target the NLRP3 inflammasome-related signaling for stroke treatment.


Subject(s)
Brain Injuries , Stroke , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroinflammatory Diseases , Stroke/drug therapy , Interleukin-1beta/metabolism
4.
Drug Discov Today ; 28(6): 103583, 2023 06.
Article in English | MEDLINE | ID: mdl-37028501

ABSTRACT

Sirtuin 3 (SIRT3), a mitochondrial deacetylase expressed preferentially in high-metabolic-demand tissues including the brain, requires NAD+ as a cofactor for catalytic activity. It regulates various processes such as energy homeostasis, redox balance, mitochondrial quality control, mitochondrial unfolded protein response, biogenesis, dynamics and mitophagy by altering protein acetylation status. Reduced SIRT3 expression or activity causes hyperacetylation of hundreds of mitochondrial proteins, which has been linked with neurological abnormalities, neuro-excitotoxicity and neuronal cell death. A body of evidence has suggested, SIRT3 activation as a potential therapeutic modality for age-related brain abnormalities and neurodegenerative disorders.


Subject(s)
Neurodegenerative Diseases , Sirtuin 3 , Humans , Sirtuin 3/metabolism , Mitochondrial Proteins/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Biology
5.
Drug Discov Today ; 28(5): 103555, 2023 05.
Article in English | MEDLINE | ID: mdl-36931386

ABSTRACT

Tailoring drug products to personalized medicines poses challenges for conventional dosage forms. The prominent reason is the restricted availability of flexible dosage strengths in the market. Inappropriate dosage strengths lead to adverse drug reactions or compromised therapeutic effects. The situation worsens when the drug has a narrow therapeutic window. To overcome these challenges, data-enriched edible pharmaceuticals (DEEP) are novel concepts for designing solid oral products. DEEP have individualized doses and information embedded in quick response (QR) code form. When data are presented in a QR code, the information is printed with edible ink that contains the drug in tailored doses required for the patients.


Subject(s)
Drug Delivery Systems , Precision Medicine , Humans , Pharmaceutical Preparations , Technology, Pharmaceutical , Dosage Forms
6.
Pharmacol Res ; 175: 106018, 2022 01.
Article in English | MEDLINE | ID: mdl-34863823

ABSTRACT

The nuclear factor erythroid-derived 2-related factor 2 (NFE2L2/Nrf2) is a pivotal facilitator of cytoprotective responses against the oxidative/electrophilic insults. Upon activation, Nrf2 induces transcription of a wide range of cytoprotective genes having antioxidant response element (ARE) in their promoter region. Dysfunction in Nrf2 signaling has been linked to the pathogenesis of AD and several studies have suggested that boosting Nrf2 expression/activity by genetic or pharmacological approaches is beneficial in AD. Among the diverse mechanisms that regulate the Nrf2 signaling, miRNAs-mediated regulation of Nrf2 has gained much attention in recent years. Several miRNAs have been reported to directly repress the post-transcriptional expression of Nrf2 and thereby negatively regulate the Nrf2-dependent cellular cytoprotective response in AD. Moreover, several Nrf2 targeting miRNAs are misregulated in AD brains. This review is focused on the role of misregulated miRNAs that directly target Nrf2, in AD pathophysiology. Here, alongside a general description of functional interactions between miRNAs and Nrf2, we have reviewed the evidence indicating the possible role of these miRNAs in AD pathogenesis.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , MicroRNAs , NF-E2-Related Factor 2/metabolism , Animals , Humans , NF-E2-Related Factor 2/chemistry , Transcription, Genetic
7.
Mol Neurobiol ; 58(9): 4575-4587, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34110602

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 or COVID-19 has been declared as a pandemic disease by the World Health Organization (WHO). Globally, this disease affected 159 million of the population and reported ~ 3.3 million deaths to the current date (May 2021). There is no definitive treatment strategy that has been identified, although this disease has prevailed in its current form for the past 18 months. The main challenges in the (SARS-CoV)-2 infections are in identifying the heterogeneity in viral strains and the plausible mechanisms of viral infection to human tissues. In parallel to the investigations into the patho-mechanism of SARS-CoV-2 infection, understanding the fundamental processes underlying the clinical manifestations of COVID-19 is very crucial for designing effective therapies. Since neurological symptoms are very apparent in COVID-19 infected patients, here, we tried to emphasize the involvement of redox imbalance and subsequent mitochondrial dysfunction in the progression of the COVID-19 infection. It has been articulated that mitochondrial dysfunction is very apparent and also interlinked to neurological symptoms in COVID-19 infection. Overall, this article provides an in-depth overview of redox imbalance and mitochondrial dysfunction involvement in aggravating COVID-19 infection and its probable contribution to the neurological manifestation of the disease.


Subject(s)
COVID-19/complications , Mitochondria/physiology , SARS-CoV-2/pathogenicity , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19/metabolism , Central Nervous System/virology , Drug Repositioning , Endothelium, Vascular/physiopathology , Endothelium, Vascular/virology , Humans , Mice , Mitochondria/drug effects , Mitochondria/pathology , Models, Biological , Olfactory Nerve/virology , Organ Specificity , Oxidation-Reduction , Oxidative Stress/drug effects , Pandemics , SARS-CoV-2/physiology , Viral Proteins/physiology , Viral Tropism , Viremia/complications , Virulence , Virus Internalization
8.
Pharmacol Res ; 167: 105414, 2021 05.
Article in English | MEDLINE | ID: mdl-33524540

ABSTRACT

Cardiac fibrosis is characterized by excessive deposition of extracellular matrix proteins and myofibroblast differentiation. Our previous findings have implicated resistin in cardiac fibrosis; however, the molecular mechanisms underlying this process are still unclear. Here we investigated the role of resistin in fibroblast-to-myofibroblast differentiation and elucidated the pathways involved in this process. Fibroblast-to-myofibroblast transdifferentiation was induced with resistin or TGFß1 in NIH-3T3 and adult cardiac fibroblasts. mRNA and protein expression of fibrotic markers were analyzed by qPCR and immunoblotting. Resistin-knockout mice, challenged with a high-fat diet (HFD) for 20 weeks to stimulate cardiac impairment, were analyzed for cardiac function and fibrosis using histologic and molecular methods. Cardiac fibroblasts stimulated with resistin displayed increased fibroblast-to-myofibroblast conversion, with increased levels of αSma, col1a1, Fn, Ccn2 and Mmp9, with remarkable differences in the actin network appearance. Mechanistically, resistin promotes fibroblast-to-myofibroblast transdifferentiation and fibrogenesis via JAK2/STAT3 and JNK/c-Jun signaling pathways, independent of TGFß1. Resistin-null mice challenged with HFD showed an improvement in cardiac function and a decrease in tissue fibrosis and reduced mRNA levels of fibrogenic markers. These findings are the first to delineate the role of resistin in the process of cardiac fibroblast-to-myofibroblast differentiation via JAK/STAT3 and JNK/c-Jun pathways, potentially leading to stimulation of cardiac fibrosis.


Subject(s)
Cell Transdifferentiation/physiology , Fibroblasts/metabolism , MAP Kinase Signaling System/physiology , Myocytes, Cardiac/metabolism , Resistin/pharmacology , STAT3 Transcription Factor/metabolism , Animals , Cell Transdifferentiation/drug effects , Female , Fibroblasts/drug effects , HEK293 Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Janus Kinases/metabolism , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , NIH 3T3 Cells , Resistin/deficiency
10.
Development ; 140(4): 843-52, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23325759

ABSTRACT

Environmental factors can stably perturb the epigenome of exposed individuals and even that of their offspring, but the pleiotropic effects of these factors have posed a challenge for understanding the determinants of mitotic or transgenerational inheritance of the epigenetic perturbation. To tackle this problem, we manipulated the epigenetic states of various target genes using a tetracycline-dependent transcription factor. Remarkably, transient manipulation at appropriate times during embryogenesis led to aberrant epigenetic modifications in the ensuing adults regardless of the modification patterns, target gene sequences or locations, and despite lineage-specific epigenetic programming that could reverse the epigenetic perturbation, thus revealing extraordinary malleability of the fetal epigenome, which has implications for 'metastable epialleles'. However, strong transgenerational inheritance of these perturbations was observed only at transgenes integrated at the Col1a1 locus, where both activating and repressive chromatin modifications were heritable for multiple generations; such a locus is unprecedented. Thus, in our inducible animal models, mitotic inheritance of epigenetic perturbation seems critically dependent on the timing of the perturbation, whereas transgenerational inheritance additionally depends on the location of the perturbation. In contrast, other parameters examined, particularly the chromatin modification pattern and DNA sequence, appear irrelevant.


Subject(s)
Chromatin/metabolism , Collagen Type I/genetics , Epigenesis, Genetic/physiology , Inheritance Patterns/physiology , Models, Biological , Phenotype , Animals , CD4 Antigens/genetics , Chromatin/genetics , Chromatin Immunoprecipitation , Collagen Type I, alpha 1 Chain , Epigenesis, Genetic/genetics , Flow Cytometry , Green Fluorescent Proteins/metabolism , Inheritance Patterns/genetics , Mice , Mice, Transgenic , Transgenes/genetics
11.
BMC Biol ; 10: 96, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23198860

ABSTRACT

BACKGROUND: Conditional gene knockout (cKO) mediated by the Cre/LoxP system is indispensable for exploring gene functions in mice. However, a major limitation of this method is that gene KO is not reversible. A number of methods have been developed to overcome this, but each method has its own limitations. RESULTS: We describe a simple method we have named LOFT [LoxP-flippase (FLP) recognition target (FRT) Trap], which is capable of reversible cKO and free of the limitations associated with existing techniques. This method involves two alleles of a target gene: a standard floxed allele, and a multi-functional allele bearing an FRT-flanked gene-trap cassette, which inactivates the target gene while reporting its expression with green fluorescent protein (GFP); the trapped allele is thus a null and GFP reporter by default, but is convertible into a wild-type allele. The floxed and trapped alleles can typically be generated using a single construct bearing a gene-trap cassette doubly flanked by LoxP and FRT sites, and can be used independently to achieve conditional and constitutive gene KO, respectively. More importantly, in mice bearing both alleles and also expressing the Cre and FLP recombinases, sequential function of the two enzymes should lead to deletion of the target gene, followed by restoration of its expression, thus achieving reversible cKO. LOFT should be generally applicable to mouse genes, including the growing numbers of genes already floxed; in the latter case, only the trapped alleles need to be generated to confer reversibility to the pre-existing cKO models. LOFT has other applications, including the creation and reversal of hypomorphic mutations. In this study we proved the principle of LOFT in the context of T-cell development, at a hypomorphic allele of Baf57/Smarce1 encoding a subunit of the chromatin-remodeling Brg/Brahma-associated factor (BAF) complex. Interestingly, the FLP used in the current work caused efficient reversal in peripheral T cells but not thymocytes, which is advantageous for studying developmental epigenetic programming of T-cell functions, a fundamental issue in immunology. CONCLUSIONS: LOFT combines well-established basic genetic methods into a simple and reliable method for reversible gene targeting, with the flexibility of achieving traditional constitutive and conditional KO.


Subject(s)
Gene Knockout Techniques , Genetic Engineering/methods , Alleles , Animals , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation/physiology , Gene Targeting , Genetic Vectors , Integrases , Mice , Mice, Knockout , Protein Subunits
12.
Drug Discov Today ; 17(13-14): 793-804, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22406696

ABSTRACT

Increased expression of endothelin (ET) peptide and its receptors following ischemic stroke is found to regulate many critical aspects of stroke pathophysiology. Many attempts have been made to target ET receptors in various animal models of stroke, but it is very difficult to draw a definite line of conclusion, because these studies differ in many aspects, such as animal model, treatment schedule, parameters and techniques used for assessing these parameters. A meta-analysis of all studies showed a significant reduction in the lesion volume and improvement in functional outcome in focal cerebral ischemia. ET(A) receptor antagonists appear to offer an essential advantage of multiple neuroprotective mechanisms, including prevention of blood-brain barrier disruption and leukocyte infiltration.


Subject(s)
Brain Ischemia/drug therapy , Drug Discovery/methods , Neuroprotective Agents/therapeutic use , Receptors, Endothelin/metabolism , Stroke/drug therapy , Animals , Brain Ischemia/complications , Disease Models, Animal , Drug Discovery/trends , Endothelin Receptor Antagonists , Humans , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Receptors, Endothelin/agonists , Receptors, Endothelin/genetics , Signal Transduction , Stroke/etiology
13.
Curr Neurovasc Res ; 8(3): 236-45, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21722092

ABSTRACT

PPARγ agonist; 2-(Benzoylphenyl)-O-[2-(methyl-2-pyridinylamino) ethyl]-L-tyrosine (GW1929) in focal cerebral ischemic-reperfusion (IR) injury in rats. Focal cerebral IR injury resulted significant brain infarction and neurological deficits in rats. A significant increase in various inflammatory mediators like COX-2, iNOS, MMP-9, TNFα and IL-6 and massive apoptotic DNA fragmentation was also observed in the IR challenged brains. GW1929 treatment significantly attenuated the neurological damage in focal cerebral IR injury. Neuroprotective effects of GW1929 were found to be associated with significant reduction in the COX-2, iNOS, MMP-9, TNFα and IL-6 levels. Together, we have also evaluated the effects of Pioglitazone, a clinically available thiazolidinedione PPARγ agonist, against focal cerebral IR injury. Like GW1929, Pioglitazone also showed beneficial effects in cerebral IR injury associated neurological damage but at the higher dose as compared to GW1929. Neuroprotective effects of PPARγ agonists were found to be associated with significant reduction in TUNEL positive cells in IR challenged brain. In summary, these results suggested the neuroprotective potential of PPARγ agonists in cerebral IR injury and these effects may be attributed to their anti-inflammatory and anti-apoptotic potential.


Subject(s)
Apoptosis/drug effects , Benzophenones/pharmacology , Brain Ischemia/drug therapy , Inflammation/drug therapy , Neuroprotective Agents/pharmacology , Reperfusion Injury/drug therapy , Tyrosine/analogs & derivatives , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , In Situ Nick-End Labeling , Inflammation/metabolism , Inflammation/pathology , Male , PPAR gamma/agonists , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Tyrosine/pharmacology
14.
Behav Brain Res ; 216(2): 606-12, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20833208

ABSTRACT

Transient global cerebral ischemia results in acute neurodegeneration in selective brain areas. Global cerebral ischemic-reperfusion (IR) injury induced selective hippocampal damage results into various neurobehavioral deficits including spatial memory and learning deficiencies. In this study, we have investigated the protective effects of a nonthiazolidinedione PPARγ agonist, N-(2-benzoylphenyl)-O-[2-(methyl-2-pyridinylamino)ethyl]-l-tyrosine (GW1929), against global cerebral IR injury induced neurobehavioral deficits and brain damage in gerbils. Bilateral carotid artery occlusion induced global cerebral ischemia in gerbils resulted in neurological deficits, hyperlocomotion, reduced response latency in passive avoidance test and hippocampal damage. Hippocampal neurodegeneration after cerebral IR injury was also associated with significant increase in iNOS and MMP-9 immunoreactivity along with TNFα and IL-6 levels. Massive apoptotic DNA fragmentation as evident from increased TUNEL (terminal deoxynucleotidyl transferase mediated dUTP nick end labelling)-positive cells was also observed in the CA1 hippocampal region of IR challenged gerbils. GW1929 treatment significantly ameliorated cerebral IR induced neurological symptoms, hyperlocomotion, cognitive deficits and hippocampal neuronal damage in CA1 hippocampus region in gerbils. Significant reduction in IR injury induced iNOS and MMP-9 immunoreactivity, TNFα and IL-6 levels and apoptotic DNA fragmentation was also observed with GW1929 treatment. Pioglitazone, thiazolidinedione PPARγ agonist also exhibited similar effects on inflammatory parameters after global cerebral IR injury. In summary, this study demonstrates neuroprotective effects of GW1929 in global cerebral IR injury induced neurobehavioral deficits and brain pathology which may be attributed to reduced inflammation and apoptotic DNA fragmentation, suggesting therapeutic potential of PPARγ agonists in cerebral IR injury.


Subject(s)
Benzophenones/pharmacology , Brain Ischemia/drug therapy , Cognition Disorders/prevention & control , Neuroprotective Agents/pharmacology , PPAR gamma/agonists , Reperfusion Injury/drug therapy , Tyrosine/analogs & derivatives , Analysis of Variance , Animals , Brain Damage, Chronic/etiology , Brain Damage, Chronic/pathology , Brain Damage, Chronic/prevention & control , Brain Ischemia/complications , Brain Ischemia/pathology , Cognition Disorders/etiology , Cognition Disorders/pathology , DNA Fragmentation/drug effects , Gerbillinae , Hippocampus/drug effects , Hippocampus/pathology , Male , Motor Activity/drug effects , Reperfusion Injury/complications , Reperfusion Injury/pathology , Tyrosine/pharmacology
15.
Drug News Perspect ; 23(4): 241-56, 2010 May.
Article in English | MEDLINE | ID: mdl-20520853

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARgamma) has already been considered as an attractive therapeutic target for the treatment of metabolic disorders. Recently, PPARgamma agonists were shown to effectively attenuate oxidative stress, inflammation and apoptosis in the central nervous system. There are several preclinical and clinical studies indicating neuroprotective potential of PPARgamma agonists in the treatment of cerebral ischemia, Parkinson's disease, Alzheimer's disease, multiple sclerosis and amyotrophic lateral sclerosis. In these disorders, apart from inhibiting oxidative stress, inflammation and apoptosis, PPARgamma agonists have the potential to modulate various signaling molecules/pathways, including matrix metalloproteinase-9, mitogen-activated protein kinases, signal transducer and activator of transcription, mitochondrial uncoupling protein 2, mitoNEET expression, amyloid precursor protein degradation, beta-site amyloid precursor protein cleaving enzyme 1 and Wnt signaling. This article discusses evidence and mechanisms supporting the neuroprotective effects of PPARgamma agonists in central nervous system disorders.


Subject(s)
Neuroprotective Agents/pharmacology , PPAR gamma/agonists , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/genetics , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Aspartic Acid Endopeptidases/genetics , Brain Ischemia/drug therapy , Humans , Matrix Metalloproteinase Inhibitors , Multiple Sclerosis/drug therapy , Neuroprotective Agents/therapeutic use , PPAR gamma/physiology , Parkinson Disease/drug therapy
16.
Neuropharmacology ; 58(3): 585-92, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20005237

ABSTRACT

Oxidative stress resulting in excessive generation of ROS is a compelling initiator of DNA damage along with damage to various cellular proteins and other macromolecules. Poly(ADP-ribose) polymerase (PARP) activation in response to DNA damage, stirs an energy-consuming cellular metabolic cycle; culminating into cell death. The present study was designed to determine the effect of combining an antioxidant, Melatonin and a PARP inhibitor, Nicotinamide on the hallmark deficits developing in diabetic neuropathy (DN). Streptozotocin (STZ, 55 mg/kg, i.p.) was administered to induce diabetes. Six weeks post diabetes induction, two week treatment with Melatonin (3 and 10 mg/kg) and Nicotinamide (100 and 300 mg/kg) either alone or in combination was given. Effect of these interventions on the functional, behavioral and biochemical changes caused by hyperglycemia were studied in treated animals. Melatonin and Nicotinamide alone as well as in combination ameliorated the functional deficits along with improvement in pain parameters. The combination also demonstrated an essential reversal of biochemical alterations. Nitrotyrosine and Poly ADP Ribose (PAR) immunopositivity was significantly decreased in sciatic nerve micro-sections of treatment group. The results of this study advocate that simultaneous inhibition of oxidative stress-PARP activation cascade may prove useful for the pharmacotherapy of DN.


Subject(s)
Antioxidants/therapeutic use , Diabetic Neuropathies/drug therapy , Melatonin/therapeutic use , Niacinamide/therapeutic use , Vitamin B Complex/therapeutic use , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetic Neuropathies/chemically induced , Diabetic Neuropathies/pathology , Diabetic Neuropathies/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Lipid Peroxidation/drug effects , Male , Neural Conduction/drug effects , Pain Measurement , Pain Threshold/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Sciatic Nerve/blood supply , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Streptozocin , Tyrosine/analogs & derivatives , Tyrosine/metabolism
17.
J Pharmacol Sci ; 109(3): 361-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19305120

ABSTRACT

Despite of the huge socio-economic burden, stroke still represents an unmet therapeutic need. Researchers failed to reproduce preclinical efficacy in subsequent clinical development. To bridge this translation failure, the Stroke Therapy Academic Industry Round Table (STAIR) has suggested a rigorous, robust, and detailed preclinical evaluation in at least 2 species and multiple cerebral ischemia models to avoid the clinical failure. Considering these recommendations, in the present study, we have investigated the effects of pioglitazone in global model cerebral ischemic-reperfusion (IR) injury in gerbils. Global cerebral IR injury, produced by bilateral carotid artery occlusion for 5 min, was characterized by neurological deficits, hyperlocomotion, and neurodegeneration in the hippocampal CA1 region. Global ischemia was also associated with oxidative stress and DNA fragmentation as evident from increased malondialdehyde (MDA) levels and TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling)-positive cells. Global cerebral IR injury associated neurological damage was significantly attenuated by pioglitazone pretreatment as evident from reduction in neurological symptoms, hyperlocomotion, and CA1 hippocampal neuronal damage in IR-challenged gerbils. Pioglitazone pretreatment also attenuated the oxidative stress and DNA fragmentation after cerebral IR injury. Pioglitazone post-treatment has also significantly reduced the CA1 hippocampal neuronal damage and DNA fragmentation after cerebral IR injury in IR-challenged gerbils. This study demonstrates the neuroprotective activity of pioglitazone in global cerebral IR injury and its neuroprotective effects may be attributed to reduction in oxidative stress and DNA fragmentation.


Subject(s)
Brain Ischemia/prevention & control , Hypoglycemic Agents/pharmacology , Reperfusion Injury/prevention & control , Thiazolidinediones/pharmacology , Animals , Brain Ischemia/physiopathology , DNA Fragmentation/drug effects , Disease Models, Animal , Gerbillinae , Hippocampus/drug effects , Hippocampus/physiopathology , Hyperkinesis/etiology , Hyperkinesis/prevention & control , In Situ Nick-End Labeling , Male , Malondialdehyde/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Pioglitazone , Reperfusion Injury/physiopathology
18.
Free Radic Res ; 43(4): 400-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19291593

ABSTRACT

The present study investigated whether combination of resveratrol and 4-amino 1,8 naphthalimide (4-ANI) is effective in the development of diabetic neuropathy (DN). After 6 weeks of diabetes induction, rats were treated for 2 weeks with resveratrol and 4-amino 1,8 naphthalimide (4-ANI) either alone or in combination. Experimental end points included functional, behavioural and biochemical parameters along with PAR immunohistochemistry and were performed at the end of treatment. Combination of resveratrol (10 mg/kg) and 4-ANI (3 mg/kg) attenuated conduction and nerve blood flow deficits and resulted in amelioration of diabetic neuropathic pain. Significant reversal of biochemical alterations (peroxynitrite, MDA and NAD levels) were also observed, as well as PAR accumulation in the sciatic nerve. This study suggests the beneficial effect of combining resveratrol and 4-ANI in experimental diabetic neuropathy.


Subject(s)
1-Naphthylamine/analogs & derivatives , Diabetic Neuropathies/drug therapy , Naphthalimides/administration & dosage , Neuroprotective Agents/administration & dosage , Quinolones/administration & dosage , Stilbenes/administration & dosage , 1-Naphthylamine/administration & dosage , Animals , Antioxidants/administration & dosage , Diabetic Neuropathies/physiopathology , Male , Malondialdehyde/metabolism , NAD/metabolism , Neural Conduction/drug effects , Neuralgia/drug therapy , Oxidative Stress/drug effects , Pain Measurement , Peroxynitrous Acid/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Rats , Rats, Sprague-Dawley , Resveratrol , Sciatic Nerve/drug effects , Sciatic Nerve/physiopathology
19.
Behav Brain Res ; 197(2): 398-403, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-18983875

ABSTRACT

The present study was carried out to elucidate the beneficial effect of pioglitazone in cognitive impairment induced by bilateral infusion of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in substantia nigra pars compacta (SNc) in rats, a model of Parkinson's disease. MPTP-lesioned rats showed poor cognitive performance in both passive avoidance task and cued version of the Morris water maze test. This deficit in learning and memory was found to be associated with oxidative stress. Chronic administration of pioglitazone (10 and 30 mg/kg, p.o., starting 5 days prior to MPTP administration and then for next 30 days) in MPTP-lesioned rats improved cognitive performance in passive avoidance task and cued version of the Morris water maze test. Furthermore, pioglitazone treatment also reduced oxidative stress (as evident by reduced malondialdehyde and increased glutathione levels). These results demonstrate the beneficial effects of pioglitazone on cognitive impairment in MPTP induced Parkinson's disease in rats.


Subject(s)
Cognition/drug effects , MPTP Poisoning/drug therapy , Thiazolidinediones/therapeutic use , Administration, Oral , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Cognition/physiology , Discrimination Learning/drug effects , Discrimination Learning/physiology , Disease Models, Animal , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Glutathione/metabolism , Lipid Peroxidation/drug effects , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Male , Malondialdehyde/analysis , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Pioglitazone , Rats , Rats, Wistar , Spatial Behavior/drug effects , Spatial Behavior/physiology , Spectrophotometry , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Thiazolidinediones/administration & dosage
20.
Pulm Pharmacol Ther ; 22(3): 183-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19073273

ABSTRACT

Airway dysfunction and pulmonary neutrophilic inflammation are the major characteristics of inflammatory conditions of lungs like chronic obstructive pulmonary disease (COPD). Lipopolysaccharide (LPS), a constituent of cigarette smoke, has been identified as the most important risk factor for COPD development. Inhalation exposure to LPS or cigarette smoke elicits an inflammatory response accompanied by airway hyperresponsiveness, elevated proinflammatory mediators and inflammatory cells similar to COPD. In the present study, we have evaluated the effects of pioglitazone, a peroxisome proliferator-activated receptor gamma (PPAR gamma) agonist, in LPS-induced pulmonary dysfunction, inflammatory changes and oxidative stress in guinea pigs. Inhalation exposure to nebulised LPS (30 microg ml(-1)) resulted in significant increase in the breathing frequency and bronchoconstriction accompanied with a significant decrease in tidal volume. Our results demonstrated that the LPS-induced pulmonary dysfunction was temporally associated with neutrophil infiltration as evident from heavy neutrophilia, increased TNFalpha in bronchoalveolar lavage fluid (BAL), elevated myeloperoxidase (MPO) level and histology of the lung tissue. Exposure to LPS also produced significant increase in tissue malondialdehyde (MDA) level indicating underlying oxidative stress. The results also reveal that pioglitazone (3, 10 and 30 mg kg(-1), p.o.) is effective in abrogating the pulmonary dysfunction by attenuating neutrophilia, TNFalpha release and oxidative stress in LPS-induced model of acute lung inflammation. Results from the present study have added to the emergent body of evidence that PPAR gamma agonists are effective in the therapy of inflammatory disease of the lungs.


Subject(s)
Lipopolysaccharides , Lung Diseases/chemically induced , Lung Diseases/drug therapy , Neutrophils/pathology , PPAR gamma/agonists , Pneumonia/chemically induced , Pneumonia/drug therapy , Thiazolidinediones/therapeutic use , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/cytology , Escherichia coli Proteins/toxicity , Guinea Pigs , Leukocyte Count , Lung/enzymology , Lung/metabolism , Lung/pathology , Male , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Peroxidase/metabolism , Phosphodiesterase Inhibitors/pharmacology , Pioglitazone , Plethysmography, Whole Body , Pneumonia/pathology , Rolipram/pharmacology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...