Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Neurochem Res ; 47(11): 3385-3401, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35922743

ABSTRACT

Parkinson's disease (PD) is a progressive motor neurodegenerative disorder significantly associated with protein aggregation related neurodegenerative mechanisms. In view of no disease modifying drugs, the present study was targeted to investigate the therapeutic effects of pharmacological agent 4-phenylbutyric acid (4PBA) in PD pathology. 4PBA is an FDA approved monocarboxylic acid with inhibitory activity towards histone deacetylase and clinically treats urea cycle disorder. First, we observed the significant protective effects of 4PBA on PD specific neuromuscular coordination, level of tyrosine hydroxylase, α-synuclein level and neurotransmitter dopamine in both substantia nigra and striatal regions of the experimental rat model of PD. Further results revealed that treatment with 4PBA drug exhibited significant protection against disease related oxidative stress and augmented nitrite levels. The disease pathology-related depletion in mitochondrial membrane potential and augmented level of calcium as well as mitochondrion membrane located VDAC1 protein level and cytochrome-c translocation were also significantly attenuated with 4PBA administration. Inhibited neuronal apoptosis and restored neuronal morphology were also observed with 4PBA treatment as measured by level of pro-apoptotic proteins t-Bid, Bax and cleaved caspase-3 along with cresyl violet staining in both substantia nigra and striatal regions. Lastly, PD-linked astrocyte activation was significantly inhibited with 4PBA treatment. Altogether, our findings suggest that 4PBA exerts broad-spectrum neuroprotective effects in PD animal model.


Subject(s)
Motor Disorders , Neuroprotective Agents , Parkinson Disease , Animals , Astrocytes/metabolism , Calcium/metabolism , Caspase 3/metabolism , Cytochromes/metabolism , Cytochromes/pharmacology , Cytochromes/therapeutic use , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons , Histone Deacetylases/metabolism , Mitochondria/metabolism , Motor Disorders/drug therapy , Motor Disorders/metabolism , Motor Disorders/pathology , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitrites/metabolism , Parkinson Disease/metabolism , Phenylbutyrates , Protein Aggregates , Rats , Tyrosine 3-Monooxygenase/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Voltage-Dependent Anion Channel 1/therapeutic use , alpha-Synuclein/metabolism , bcl-2-Associated X Protein/metabolism
2.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2078-2096, 2018 06.
Article in English | MEDLINE | ID: mdl-29551729

ABSTRACT

Piracetam, a nootropic drug that has been clinically used for decades but remains enigmatic due to no distinct understanding of its mechanism of action. The present study aimed to investigate the role of caspase independent pathway in piracetam mediated neuroprotection. LPS administration caused significant alterations in oxidative stress related parameters like glutathione, glutathione reductase and increased lipid peroxidation. LPS administration also caused augmented expression of inflammatory cytokines and astrocytes activation. Piracetam treatment offered significant protection against LPS induced oxidative and inflammatory parameters and inhibited astrocytes activation. LPS administration caused augmented level of reactive oxygen species and depleted mitochondrial membrane potential which were attenuated with piracetam treatment. This study for the first time demonstrates the role of caspase independent death factors in piracetam induced neuroprotective effects in rat brain. Translocation of mitochondrial resident apoptosis inducing factor and endonuclease G to nucleus through cytosol after LPS administration was significantly blocked with piracetam treatment. Further, LPS induced DNA fragmentation along with up regulated Poly [ADP-ribose] polymerase 1 (PARP1) levels were also inhibited with piracetam treatment. Apoptotic death was confirmed by the cleavage of caspase 3 as well as histological alteration in rat brain regions. LPS administration caused significantly increased level of cleaved caspase 3, altered neuronal morphology and decreased neuronal density which were restored with piracetam treatment. Collectively our findings indicate that piracetam offered protection against LPS induced inflammatory responses and cellular death including its antioxidative antiapoptotic activity with its attenuation against mitochondria mediated caspase independent pathway.


Subject(s)
Mitochondria/drug effects , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Piracetam/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Endodeoxyribonucleases/metabolism , Humans , Lipid Peroxidation/drug effects , Lipopolysaccharides/toxicity , Male , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondria/pathology , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Piracetam/therapeutic use , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
4.
J Nat Prod ; 79(5): 1276-84, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27136692

ABSTRACT

This study identified koenidine (4) as a metabolically stable antidiabetic compound, when evaluated in a rodent type 2 model (leptin receptor-deficient db/db mice), and showed a considerable reduction in the postprandial blood glucose profile with an improvement in insulin sensitivity. Biological studies were directed from the preliminary in vitro evaluation of the effects of isolated carbazole alkaloids (1-6) on glucose uptake and GLUT4 translocation in L6-GLUT4myc myotubes, followed by an investigation of their activity (2-5) in streptozotocin-induced diabetic rats. The effect of koenidine (4) on GLUT4 translocation was mediated by the AKT-dependent signaling pathway in L6-GLUT4myc myotubes. Moreover, in vivo pharmacokinetic studies of compounds 2 and 4 clearly showed that compound 4 was 2.7 times more bioavailable than compound 2, resulting in a superior in vivo efficacy. Therefore, these studies suggested that koenidine (4) may serve as a promising lead natural scaffold for managing insulin resistance and diabetes.


Subject(s)
Carbazoles/isolation & purification , Carbazoles/pharmacology , Hypoglycemic Agents/isolation & purification , Hypoglycemic Agents/pharmacology , Murraya/chemistry , Alkaloids/pharmacology , Animals , Blood Glucose/metabolism , Carbazoles/chemistry , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/chemistry , Insulin/pharmacology , Insulin Resistance , Male , Mice , Molecular Structure , Muscle Fibers, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Streptozocin/pharmacology
5.
Food Chem Toxicol ; 155: 112433, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34302886

ABSTRACT

The present study was undertaken to evaluate the mechanism for antiParkinsonian effect of resveratrol employing 6-hydroxydopamine (6-OHDA) induced experimental model of Parkinson's disease (PD). Resveratrol treatment significantly protects the PD related pathological markers like level of tyrosine hydroxylase, dopamine and apoptotic proteins (Bax and cleaved caspase-3). Disease pathology involves significantly decreased level of dopamine transporter, synaptophysin and postsynaptic density protein 95 (PSD-95) along with augmented level of vesicular monoamine transporter and considerably affected the dendrite arborization. Such affected neuronal communication was significantly restored with resveratrol treatment. Biochemical alterations include the depleted level of glutathione (GSH), mitochondrial complex-I activity with concomitant increased level of lipid peroxidation, nitrite level and calcium levels, which were also significantly inhibited with resveratrol treatment. Altered calcium level induces the endoplasmic reticulum (ER) stress related signalling and phosphorylated Nuclear factor erythroid 2-related factor 2 (Nrf2), and with resveratrol treatment the level of phosphorylated Nrf2 was further increased. The concurrent depleted level of proteasome activity was observed which was attenuated with resveratrol treatment. Proinflammatory cytokines and activated astrocytes were observed which was inhibited with resveratrol treatment. In conclusion, findings suggested that resveratrol exhibits the interference in neuronal communication, oxidative stress, mitochondrial pathophysiology, ER stress, protein degradation mechanism and inflammatory responses and could be utilize in clinics to treat the PD patients.


Subject(s)
Antiparkinson Agents/therapeutic use , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Neuronal Plasticity/drug effects , Parkinson Disease, Secondary/drug therapy , Resveratrol/therapeutic use , Animals , Dendrites/drug effects , Disks Large Homolog 4 Protein/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Male , Neuroprotective Agents/therapeutic use , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Rats, Sprague-Dawley , Synaptophysin/metabolism , Vesicular Monoamine Transport Proteins/metabolism
6.
Neurochem Int ; 129: 104463, 2019 10.
Article in English | MEDLINE | ID: mdl-31078578

ABSTRACT

Nigral dopaminergic (DAergic) cell degeneration and depletion of dopamine neurotransmitter in the midbrain are cardinal features of Parkinson's disease (PD). Dopamine system regulates different aspects of behavioural phenotypes such as motor control, reward, anxiety and depression via acting on dopamine receptors (D1-D5). Recent studies have shown the potential effects of dopamine on modulation of neurogenesis, a process of newborn neuron formation from neural stem cells (NSCs). Reduced proliferative capacity of NSCs and net neurogenesis has been reported in subventricular zone, olfactory bulb and hippocampus of patients with PD. However, the molecular and cellular mechanism of dopamine mediated modulation of DAergic neurogenesis is not defined. In this study, we attempted to investigate the molecular mechanism of dopamine receptors mediated control of DAergic neurogenesis and whether it affects mitochondrial biogenesis in 6-hydroxydopamine (6-OHDA) induced rat model of PD-like phenotypes. Unilateral administration of 6-OHDA into medial forebrain bundle potentially reduced tyrosine hydroxylase immunoreactivity, dopamine content in substantia nigra pars compacta (SNpc) and striatum region and impaired motor functions in adult rats. We found decreased D1 receptor expression, mitochondrial biogenesis, mitochondrial functions and DAergic differentiation associated with down-regulation of Wnt/ß-catenin signalling in SNpc of 6-OHDA lesioned rats. Pharmacological stimulation of D1 receptor enhanced mitochondrial biogenesis, mitochondrial functions and DAergic neurogenesis that lead to improved motor functions in 6-OHDA lesioned rats. D1 agonist induced effects were attenuated following administration of D1 antagonist, whereas shRNA mediated knockdown of Axin-2, a negative regulator of Wnt signalling significantly abolished D1 antagonist induced impairment in mitochondrial biogenesis and DAergic neurogenesis in 6-OHDA lesioned rats. Our results suggest that dopamine receptor regulates DAergic neurogenesis and mitochondrial functions by activation of Wnt/ß-catenin signaling in rat model of PD-like phenotypes.


Subject(s)
Dopamine/pharmacology , Mitochondria/drug effects , Oxidative Stress/drug effects , Wnt Signaling Pathway/drug effects , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Male , Mitochondria/metabolism , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Oxidopamine/pharmacology , Rats, Sprague-Dawley , Receptors, Dopamine D1/metabolism
7.
Eur J Med Chem ; 182: 111632, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31499363

ABSTRACT

ß-amino acids and their analogues are gathering increased attention not only because of their antibacterial and antifungal activity, but also for their use in designing peptidomimetics with increased oral bioavailability and resistance to metabolic degradation. In this study, a series of α-phenyl substituted chalcones, α-phenyl, ß-amino substituted dihydrochalcones and ß-amino acid derivatives were synthesized and evaluated for their antileishmanial efficacy against experimental visceral leishmaniasis (VL). Among all synthesized derivatives, 10c showed promising antileishmanial efficacy against both extracellular promastigote and intracellular amastigote (IC50 8.2 µM and 20.5 µM respectively) of L. donovani with negligible cytotoxic effect towards J774 macrophages and Vero cells. 10c effectively reduced spleen and liver parasite burden (>90%) in both hamster and Balb/c model of VL without any hepatotoxicity. In vitro pharmacokinetic analysis showed that 10c was stable in gastric fluid and plasma of Balb/c mice at 10 µg/ml. Further analysis of the molecular mechanism revealed that 10c entered into the parasite by depolarizing the plasma membrane rather than forming nonspecific pores and induced molecular events like loss in mitochondrial membrane potential with a gradual decline in ATP production. This, in turn, did not induce programmed cell death of the parasite; rather 10c induced bioenergetic collapse of the parasite by decreasing ATP synthesis through specific inhibition of mitochondrial complex III activity. Altogether, our results allude to the therapeutic potential of ß-amino acid derivatives as novel antileishmanials, identifying them as lead compounds for further exploration in the design of potent candidates for the treatment of visceral leishmaniasis.


Subject(s)
Amino Acids/pharmacology , Antiprotozoal Agents/pharmacology , Electron Transport Complex III/antagonists & inhibitors , Leishmania donovani/drug effects , Leishmaniasis, Visceral/drug therapy , Mitochondria/drug effects , Amino Acids/chemistry , Animals , Antiprotozoal Agents/chemistry , Cell Survival/drug effects , Chlorocebus aethiops , Cricetinae , Dose-Response Relationship, Drug , Electron Transport Complex III/metabolism , Leishmania donovani/metabolism , Leishmaniasis, Visceral/metabolism , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship , Vero Cells
8.
Eur J Pharmacol ; 815: 462-469, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28974347

ABSTRACT

Endothelial dysfunction plays an important role in structural remodeling occurring in the pulmonary vasculature during pulmonary hypertension (PH). Endothelial injury causes apoptosis and activation of endothelial cells. However, some endothelial cells show apoptosis-resistance and later proliferate extensively leading to vascular oculopathy and formation of plexiform lesions in PH. Studies have shown that rapidly proliferating cells exhibit increased expression of Fatty acid synthase (FAS), a regulatory enzyme responsible for the production of fatty acids. Our previous study has shown that FAS inhibition prevented smooth muscle cell proliferation, reversed pulmonary vascular remodeling and improved pulmonary vasoreactivity in monocrotaline induced PH model. However, the role of FAS in pulmonary artery endothelial cell proliferation and angiogenesis has not been explored. The present study was designed to explore the role of FAS in proliferation, metabolic dysfunctions, and angiogenesis in endothelial dysfunction associated with PH. The human pulmonary artery endothelial cells (HPAECs) were exposed to hypoxia and FAS siRNA (60nM) was used for the FAS inhibition. Increased expression and activity of FAS were observed in hypoxic HPAECs. Inhibition of FAS increased apoptosis and glucose oxidation, but decreased cellular proliferation, markers of autophagy and glycolysis in hypoxic HPAECs. FAS inhibition decreased the angiogenesis as evident by decreased tubule length and VEGF expression in hypoxic HPAECs. Inhibition of FAS also increased expression of endothelial NOS in hypoxic HPAECs, a marker of endothelial function. Our results proved, and further supported previous findings, that inhibition of FAS is beneficial for endothelial function in pulmonary hypertension.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/pathology , Fatty Acid Synthases/metabolism , Neovascularization, Physiologic , Pulmonary Artery/pathology , Autophagy , Cell Hypoxia , Cell Proliferation , Gene Expression Regulation, Enzymologic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nitric Oxide Synthase Type III/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology
9.
J Med Chem ; 60(3): 1041-1059, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28059524

ABSTRACT

A series of pyrazolo(dihydro)pyridines was synthesized and evaluated for antileishmanial efficacy against experimental visceral leishmaniasis (VL). Among all compounds, 6d and 6j exhibited better activity than miltefosine against intracellular amastigotes. Compound 6j (50 mg/kg/day) was further studied against Leishmania donovani/BALB/c mice via the intraperitoneal route for 5 days and displayed >91 and >93% clearance of splenic and liver parasitic burden, respectively. Combination treatment of 6j with a subcurative dose of miltefosine (5 mg/kg) in BALB/c mice almost completely ameliorated the disease (>97% inhibition) by augmenting nitric oxide generation and shifting the immune response toward Th1. Furthermore, investigating the effect of 6j on Leishmania promastigotes revealed that it induced molecular events, such as a loss in mitochondrial membrane potential, externalization of phosphatidylserine, and DNA fragmentation, that ultimately resulted in the programmed cell death of the parasite. These results along with pharmacokinetic studies suggest that 6j could be a promising lead for treating VL as an adjunct therapy with miltefosine.


Subject(s)
Antiprotozoal Agents/pharmacology , Leishmania/drug effects , Phosphorylcholine/analogs & derivatives , Pyrazoles/pharmacology , Pyridines/pharmacology , Animals , Antiprotozoal Agents/pharmacokinetics , Antiprotozoal Agents/therapeutic use , Mice , Mice, Inbred BALB C , Phosphorylcholine/pharmacokinetics , Phosphorylcholine/therapeutic use , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyridines/pharmacokinetics , Pyridines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL