Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 18(2): 519-533, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36608690

ABSTRACT

Demyelinating disorders of the central nervous system (CNS) occur when myelin and oligodendrocytes are damaged or lost. Remyelination and regeneration of oligodendrocytes can be achieved from endogenous oligodendrocyte precursor cells (OPCs) that reside in the adult CNS tissue. Using a cuprizone mouse model of demyelination, we show that infusion of fractalkine (CX3CL1) into the demyelinated murine brain increases de novo oligodendrocyte formation and enhances remyelination in the corpus callosum and cortical gray matter. This is achieved by increased OPC proliferation in the cortical gray matter as well as OPC differentiation and attenuation of microglia/macrophage activation both in corpus callosum and cortical gray matter. Finally, we show that activated OPCs and microglia/macrophages express fractalkine receptor CX3CR1 in vivo, and that in OPC-microglia co-cultures fractalkine increases in vitro oligodendrocyte differentiation by modulating both OPC and microglia biology. Our results demonstrate a novel pro-regenerative role of fractalkine in a demyelinating mouse model.


Subject(s)
Demyelinating Diseases , Remyelination , Mice , Animals , Chemokine CX3CL1 , Oligodendroglia/physiology , Myelin Sheath , Disease Models, Animal , Cell Differentiation/physiology , Mice, Inbred C57BL
2.
Exp Neurol ; 334: 113454, 2020 12.
Article in English | MEDLINE | ID: mdl-32877653

ABSTRACT

Individuals with demyelinating diseases often experience difficulties during social interactions that are not well studied in preclinical models. Here, we describe a novel juvenile focal corpus callosum demyelination murine model exhibiting a social interaction deficit. Using this preclinical murine demyelination model, we discover that application of metformin, an FDA-approved drug, in this model promotes oligodendrocyte regeneration and remyelination and improves the social interaction. This beneficial effect of metformin acts through stimulating Ser436 phosphorylation in CBP, a histone acetyltransferase. In addition, we found that metformin acts through two distinct molecular pathways to enhance oligodendrocyte precursor (OPC) proliferation and differentiation, respectively. Metformin enhances OPC proliferation through early-stage autophagy inhibition, while metformin promotes OPC differentiation into mature oligodendrocytes through activating CBP Ser436 phosphorylation. In summary, we identify that metformin is a promising remyelinating agent to improve juvenile demyelination-associated social interaction deficits by promoting oligodendrocyte regeneration and remyelination.


Subject(s)
Demyelinating Diseases/drug therapy , Demyelinating Diseases/metabolism , Histone Acetyltransferases/metabolism , Metformin/therapeutic use , Remyelination/drug effects , Social Interaction/drug effects , Animals , Demyelinating Diseases/psychology , Female , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Remyelination/physiology , Serine/metabolism
3.
Theranostics ; 10(14): 6337-6360, 2020.
Article in English | MEDLINE | ID: mdl-32483456

ABSTRACT

Rationale: Monoacylglycerol lipase (Mgll), a hydrolase that breaks down the endocannabinoid 2-arachidonoyl glycerol (2-AG) to produce arachidonic acid (ARA), is a potential target for neurodegenerative diseases, such as Alzheimer's disease (AD). Increasing evidence shows that impairment of adult neurogenesis by perturbed lipid metabolism predisposes patients to AD. However, it remains unknown what causes aberrant expression of Mgll in AD and how Mgll-regulated lipid metabolism impacts adult neurogenesis, thus predisposing to AD during aging. Here, we identify Mgll as an aging-induced factor that impairs adult neurogenesis and spatial memory in AD, and show that metformin, an FDA-approved anti-diabetic drug, can reduce the expression of Mgll to reverse impaired adult neurogenesis, prevent spatial memory decline and reduce ß-amyloid accumulation. Methods: Mgll expression was assessed in both human AD patient post-mortem hippocampal tissues and 3xTg-AD mouse model. In addition, we used both the 3xTg-AD animal model and the CbpS436A genetic knock-in mouse model to identify that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway, involving atypical protein kinase C (aPKC)-stimulated Ser436 phosphorylation of histone acetyltransferase CBP through biochemical methods. Furthermore, we performed in vivo adult neurogenesis assay with BrdU/EdU labelling and Morris water maze task in both animal models following pharmacological treatments to show the key role of Mgll in metformin-corrected neurogenesis and spatial memory deficits of AD through reactivating the aPKC-CBP pathway. Finally, we performed in vitro adult neurosphere assays using both animal models to study the role of the aPKC-CBP mediated Mgll repression in determining adult neural stem/progenitor cell (NPC) fate. Results: Here, we demonstrate that aging-dependent induction of Mgll is observed in the 3xTg-AD model and human AD patient post-mortem hippocampal tissues. Importantly, we discover that elevated Mgll expression is caused by the attenuation of the aPKC-CBP pathway. The accumulation of Mgll in the 3xTg-AD mice reduces the genesis of newborn neurons and perturbs spatial memory. However, we find that metformin-stimulated aPKC-CBP pathway decreases Mgll expression to recover these deficits in 3xTg-AD. In addition, we reveal that elevated Mgll levels in cultured adult NPCs from both 3xTg-AD and CbpS436A animal models are responsible for their NPC neuronal differentiation deficits. Conclusion: Our findings set the stage for development of a clinical protocol where Mgll would serve as a biomarker in early stages of AD to identify potential metformin-responsive AD patients to restore their neurogenesis and spatial memory.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Metformin/pharmacology , Monoacylglycerol Lipases/metabolism , Neurogenesis/drug effects , Spatial Memory/drug effects , Alzheimer Disease/pathology , Animals , Biomarkers/metabolism , CREB-Binding Protein/metabolism , Disease Models, Animal , Female , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Transgenic , Protein Kinase C/metabolism
4.
Neurosci Lett ; 687: 207-215, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30278248

ABSTRACT

Neuroinflammation has been observed in post-mortem Alzheimer's disease (AD) brains which could be due to Aß interacting with microglia and astrocytes. SLOH, a carbazole-based fluorophore, was shown to bind to Aß peptides. Herein, we investigated the anti-neuroinflammatory effects of SLOH using a BV-2 microglial cell model and a triple transgenic AD (3xTg-AD) mouse model. BV-2 cells were incubated with Aß in the presence of SLOH for 24 h. The levels of pro-inflammatory and anti-inflammatory cytokines were determined. Moreover, 3xTg-AD mice were administrated with SLOH (2 mg kg-1) for one month. The mice were then sacrificed and the brains were used to assess the levels of pro-inflammatory, anti-inflammatory cytokines and the activation of ionized calcium-binding adapter molecule 1 (Iba1). BV-2 cell studies suggested that SLOH reduced the production and mRNA levels of pro-inflammatory cytokines TNF-α, IL-1ß, COX-2, iNOS, and increased IL-10. Animal study confirmed that SLOH reduced the production of pro-inflammatory cytokines and increased the level of anti-inflammatory cytokine. Moreover, SLOH inhibited the activity of GSK-3ß. In 3xTg-AD mouse model, SLOH treatment significantly decreased the number of Iba1-positive cells in mouse brains. Our results demonstrated that SLOH significantly attenuated the neuroinflammation through down-regulating the activity of GSK-3ß.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/pharmacology , Glycogen Synthase Kinase 3 beta/drug effects , Microglia/drug effects , Alzheimer Disease/genetics , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Mice, Transgenic , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , tau Proteins/metabolism
5.
Neuropharmacology ; 131: 351-363, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29309769

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative dysfunction characterized by memory impairment and brings a heavy burden to old people both in developing and developed countries. Amyloid hypothesis reveals that aggregation and deposition of amyloid plaques are the cause of AD neurodegeneration. SLOH, a carbazole-based fluorophore, is reported to inhibit amyloid beta (Aß) aggregation in vitro. In the current study, we intended to evaluate the protective effect of SLOH in a triple transgenic AD mouse model (3xTg-AD). 3xTg-AD (10-month-old) were treated with SLOH (0.5, 1 and 2 mg kg-1) for one month via intraperitoneal injection. After treatment, cognitive function was assessed by Morris Water Maze (MWM) and Y-maze tasks. In addition, biochemical estimations were used to examine the degree of Aß deposition, tau hyperphosphorylation and neuroinflammation in the brains of 3xTg-AD mice. An in vitro study was conducted on human neuroblastoma (SH-SY5Y) cells to determine the activity of SLOH on tau and GSK-3ß using western blot and immunofluorescence staining. One month treatment with SLOH significantly ameliorated memory impairments in 3xTg-AD mice in MWM and Y-maze tests. Moreover, SLOH treatment mitigated the level of amyloid plaques, tau hyperphosphorylation and neuroinflammation in the mouse brain. SLOH also reduced tau hyperphosphorylation and down-regulated GSK-3ß activity in Aß induced neurotoxic SH-SY5Y cells. The promising results in mitigating amyloid plaques, tau hyperphosphorylation, neuroinflammation and ameliorating cognitive deficits following one-month treatment suggest that SLOH could be a potential multi-target molecule for the AD treatment.


Subject(s)
Alzheimer Disease/drug therapy , Carbazoles/pharmacology , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Dose-Response Relationship, Drug , Female , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Maze Learning/drug effects , Maze Learning/physiology , Mice, Inbred C57BL , Mice, Transgenic , Molecular Structure , Random Allocation , tau Proteins/genetics , tau Proteins/metabolism
6.
Eur J Pharm Sci ; 110: 101-108, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28359686

ABSTRACT

Phosphorylated tau dissociates from microtubules and aggregates to form neurofibrillary tangles resulting in neuronal toxicity and cognitive deficits. Attenuating tau hyperphosphorylation is considered as an effective therapeutic approach for Alzheimer's disease (AD). From our previous study, SLM, a carbazole-based fluorophore prevents Aß aggregation, reduced glycogen synthase kinase-3ß (GSK-3ß) activity and tau hyperphosphorylation in triple transgenic mouse model of AD. However, the mechanism by which SLM attenuates tau hyperphosphorylation warrants further investigation. In the current study, we intend to evaluate the effects of SLM against okadaic acid (OA)-induced tau hyperphosphorylation and microtubules instability in human neuroblastoma (SH-SY5Y) cells. The results showed that, SLM reduced the OA-induced cell neurotoxicity and tau hyperphosphorylation in SH-SY5Y cells. SLM treatment down-regulated GSK-3ß activity. However, in the presence of GSK-3ß inhibitor (SB216763, 10µM), SLM treatment could not reduce GSK-3ß activity and tau hyperphosphorylation as compared with SB216763 treatment alone. Furthermore, SLM treatment also ameliorated OA-induced microtubules instability and cytoskeleton damage. Collectively, SLM attenuated OA-induced tau hyperphosphorylation via down-regulating GSK-3ß activity in SH-SY5Y cells. Therefore, this study supports SLM as a potential compound for AD and other tau pathology-related neurodegenerative disorders.


Subject(s)
Carbazoles/pharmacology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Neuroprotective Agents/pharmacology , Okadaic Acid/pharmacology , tau Proteins/metabolism , Animals , Cell Line, Tumor , Down-Regulation , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice , Microtubules/metabolism , Neurons , Phosphorylation , Protein Stability
7.
ACS Chem Neurosci ; 8(3): 676-685, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28032988

ABSTRACT

Amyloid ß (Aß) peptide aggregating to form a neurotoxic plaque, leading to cognitive deficits, is believed to be one of the plausible mechanisms for Alzheimer's disease (AD). Inhibiting Aß aggregation is supposed to offer a neuroprotective effect to ameliorate AD. A previous report has shown that SLM, a carbazole-based fluorophore, binds to Aß to inhibit the aggregation. However, it is not entirely clear whether the inhibition of Aß aggregation alone would lead to the anticipated neuroprotective effects. In the current study, we intended to examine the protective action of SLM against Aß-induced neurotoxicity in vitro and to evaluate if SLM can decrease the cognitive and behavioral deficits observed in triple transgenic AD mouse model (3xTg-AD). In the in vitro study, neurotoxicity induced by Aß42 in human neuroblastoma (SH-SY5Y) cells was found to be reduced through the treatment with SLM. In the in vivo study, following one month SLM intraperitoneal injection (1, 2, and 4 mg/kg), 3xTg-AD mice were tested on Morris water maze (MWM) and Y-maze for their cognitive ability and sacrificed for biochemical estimations. Results show that SLM treatment improved the learning and memory ability in 3xTg-AD mice in MWM and Y-maze tasks. SLM also mitigated the amyloid burden by decreasing brain Aß40 and Aß42 levels and reduced tau phosphorylation, glycogen synthase kinase-3ß activity, and neuro-inflammation. From our observations, SLM shows neuroprotection in SH-SY5Y cells against Aß42 and also in 3xTg-AD mouse model by mitigating the pathological features and behavioral impairments.


Subject(s)
Alzheimer Disease/drug therapy , Carbazoles/chemistry , Carbazoles/therapeutic use , Cell Death/drug effects , Gene Expression Regulation/drug effects , Neuroprotective Agents/therapeutic use , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Gene Expression Regulation/genetics , Humans , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/pathology , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Plaque, Amyloid/etiology , Plaque, Amyloid/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , tau Proteins/genetics , tau Proteins/metabolism
8.
Mol Neurobiol ; 54(8): 6074-6084, 2017 Oct.
Article in English | MEDLINE | ID: mdl-27699599

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an incretin hormone shown to be active in the treatment of type-2 diabetes (T2D) and has also been shown as efficacious in Alzheimer's disease (AD). Dipeptidyl peptidase-4 (DPP-4), an enzyme that is expressed in numerous cells, rapidly inactivates endogenous GLP-1. Therefore, DPP-4 inhibition is employed as a therapeutic avenue to increase GLP-1 levels in the management of T2D. The effectiveness of DPP-4 inhibitors in the treatment of AD has been reported in various animal models of AD. With this background, the present study was designed to examine the effectiveness of linagliptin, a DPP-4 inhibitor in the 3xTg-AD mouse model of Alzheimer's disease. Nine-month-old 3xTg-AD mice were administered linagliptin orally (5, 10, and 20 mg/kg) for 8 weeks. At the end of the linagliptin treatment, mice were evaluated for cognitive ability on the Morris Water Maze and Y-maze. Following cognitive evaluation, mice were sacrificed to determine the effect of the linagliptin on brain incretin levels, amyloid burden, tau phosphorylation, and neuroinflammation. We confirm that linagliptin treatment for 8 weeks mitigates the cognitive deficits present in 3xTg-AD mice. Moreover, linagliptin also improves brain incretin levels and attenuates amyloid beta, tau phosphorylation as well as neuroinflammation. In conclusion, linagliptin possesses neuroprotective properties that may be attributed to the improvement of incretin levels in the brain.


Subject(s)
Alzheimer Disease/drug therapy , Cognition Disorders/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Linagliptin/therapeutic use , Maze Learning/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Blood Glucose/metabolism , Cognition Disorders/genetics , Cognition Disorders/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Female , Glucagon-Like Peptide 1/metabolism , Linagliptin/pharmacology , Mice , Mice, Transgenic , tau Proteins/metabolism
9.
Indian J Pharmacol ; 46(2): 176-80, 2014.
Article in English | MEDLINE | ID: mdl-24741189

ABSTRACT

OBJECTIVE: The present study investigates the neuroprotective activity of ethanol extract of Tinospora cordifolia aerial parts against 6-hydroxy dopamine (6-OHDA) lesion rat model of Parkinson's disease (PD). MATERIALS AND METHODS: T. cordifolia ethanol extract (TCEE) was standardized with high performance thin layer chromatography using berberine. Experimental PD was induced by intracerebral injection of 6-OHDA (8 µg). Animals were divided into five groups: sham operated, negative control, positive control (levodopa 6 mg/kg) and two experimental groups (n = 6/group). Experimental groups received 200 and 400 mg/kg of TCEE once daily for 30 days by oral gavage. Biochemical parameters including dopamine level, oxidative stress, complex I activity and brain iron asymmetry ratio and locomotor activity including skeletal muscle co-ordination and degree of catatonia were assessed. RESULTS: TCEE exhibited significant neuroprotection by increasing the dopamine levels (1.96 ± 0.20 and 2.45 ± 0.40 ng/mg of protein) and complex I activity (77.14 ± 0.89 and 78.50 ± 0.96 nmol/min/mg of protein) at 200 and 400 mg/kg respectively when compared with negative control group. Iron asymmetry ratio was also significantly attenuated by TCEE at 200 (1.57 ± 0.18) and 400 mg/kg (1.11 ± 0.15) when compared with negative control group. Neuroprotection by TCEE was further supported by reduced oxidative stress and restored locomotor activity in treatment groups. CONCLUSION: Results show that TCEE possess significant neuroprotection in 6-OHDA induced PD by protecting dopaminergic neurons and reducing the iron accumulation.


Subject(s)
Neuroprotective Agents/therapeutic use , Oxidopamine/pharmacology , Parkinson Disease/prevention & control , Plant Extracts/therapeutic use , Tinospora/chemistry , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Electron Transport Complex I/metabolism , Ethanol/chemistry , Iron/metabolism , Male , Motor Activity/drug effects , Neuroprotective Agents/isolation & purification , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Plant Components, Aerial/chemistry , Plant Extracts/isolation & purification , Rats, Wistar , Rotarod Performance Test
10.
Phytother Res ; 28(11): 1731-4, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24590863

ABSTRACT

The fruit of Eugenia jambolana Lam. is very popular for its anti-diabetic property. Previous studies on the crude extract of E. jambolana (EJE) have successfully explored the scientific basis for some of its traditional medicinal uses. Considering its wide use and consumption as a seasonal fruit, the present study investigates the ability of E. jambolana to interact with cytochrome P450 enzymes. The standardized EJE was incubated with pooled human liver microsomes to assess the CYP2C9-, CYP2D6-, and CYP3A4-mediated metabolism of diclofenac, dextromethorphan, and testosterone, respectively. The metabolites formed after the enzymatic reactions were quantified by high performance liquid chromatography. EJE showed differential effect on cytochrome P450 activities with an order of inhibitory potential as CYP2C9 > CYP3A4 > CYP2D6 having IC50 of 76.69, 359.02, and 493.05 µg/mL, respectively. The selectivity of EJE for CYP2C9 rather than CYP3A4 and CYP2D6 led to perform the enzyme kinetics to explicate the mechanism underlying the inhibition of CYP2C9-mediated diclofenac 4'-hydroxylation. EJE was notably potent in inhibiting the reaction in a non-competitive manner with Ki of 84.85 ± 5.27 µg/mL. The results revealed the CYP2C9 inhibitory potential of EJE with lower Ki value suggesting that EJE should be examined for its potential pharmacokinetic and pharmacodynamic interactions when concomitantly administered with other drugs.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/drug effects , Plant Extracts/pharmacology , Syzygium/chemistry , Chromatography, High Pressure Liquid , Ellagic Acid/analysis , Fruit/chemistry , Humans , Microsomes, Liver/enzymology , Polyphenols/analysis
11.
Behav Brain Res ; 267: 55-65, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24667360

ABSTRACT

Alzheimer's disease (AD), the most common form of dementia, is characterized by the loss of normal functions of brain cells and neuronal death, ultimately leading to memory loss. Recent accumulating evidences have demonstrated the therapeutic potential of anti-diabetic agents, such as dipeptidyl peptidase-4 (DPP-4) inhibitors, for the treatment of Alzheimer's disease (AD), providing opportunities to explore and test the DPP-4 inhibitors for treating this fatal disease. Prior studies determining the efficacy of Pterocarpus marsupium (PM, Fabaceae) and Eugenia jambolana (EJ, Myrtaceae) extracts for ameliorating type 2 diabetes have demonstrated the DPP-4 inhibitory properties indicating the possibility of using of these extracts even for the treating AD. Therefore, in the present study, the neuroprotective roles of PM and EJ for ameliorating the streptozotocin (STZ) induced AD have been tested in rat model. Experimentally, PM and EJ extracts, at a dose range of 200 and 400mg/kg, were administered orally to STZ induced AD Wistar rats and cognitive evaluation tests were performed using radial arm maze and hole-board apparatus. Following 30 days of treatment with the extracts, a dose- and time-dependent attenuation of AD pathology, as evidenced by decreasing amyloid beta 42, total tau, phosphorylated tau and neuro-inflammation with an increase in glucagon-like peptide-1 (GLP-1) levels was observed. Therefore, PM and EJ extracts contain cognitive enhancers as well as neuroprotective agents against STZ induced AD.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/therapeutic use , Phytotherapy , Plant Preparations/therapeutic use , Pterocarpus , Syzygium , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Glucagon-Like Peptide 1/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Maze Learning/drug effects , Peptide Fragments/metabolism , Phosphorylation/drug effects , Plaque, Amyloid/drug therapy , Rats, Wistar , Streptozocin , Time Factors , tau Proteins/metabolism
12.
Pharm Biol ; 52(2): 268-71, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24074231

ABSTRACT

CONTEXT: Pterocarpus marsupium (PM) (Leguminosae), Eugenia jambolana (EJ) (Myrtaceae) and Gymnema sylvestre (GS) (Asclepiadaceae) are the most important medicinal plants in the Indian system of traditional medicine for the treatment of hyperglycemia. OBJECTIVES: Dipeptidyl peptidase-4 (DPP-4) inhibitors are the emerging class of anti-diabetic agents. However, only few compounds are commercially available. Therefore, in the present study we tried to explore the naturally occurring PM, EJ and GS semi-standardized extracts for their potential DPP-4 inhibition in vitro and in vivo. MATERIALS AND METHODS: DPP-4 inhibition was evaluated by in vitro inhibitory assay, and enzyme kinetics were calculated using one-phase exponential decay equation. Glucose load (2 g/kg) was administered to control and diabetic rats 30 min following extract administration (100, 200 and 400 mg/kg) orally once, and blood samples were withdrawn at 0, 0.5, 1, 1.5, 2 and 3 h to measure plasma active glucagon-like peptide-1 (GLP-1) levels. RESULTS: PM and EJ inhibit DPP-4 potently with IC50 values of 273.73 ± 2.96 and 278.94 ± 6.73 µg/mL, respectively, compared to GS (773.22 ± 9.21 µg/mL). PM, EJ and GS exhibit long duration of action with enzyme inhibitory half-lives of 462.3, 317.2 and 153.8 min, respectively. Extracts significantly increase GLP-1 levels compared to negative control groups and peak GLP-1 level was observed at 2 h for PM and EJ, whereas for GS it was at 1.5 h DISCUSSION AND CONCLUSION: Taken together, results suggest the extracts may have potent DPP-4 inhibitory action, and their hypoglycemic action attributed through an increase in plasma active GLP-1 levels.


Subject(s)
Gymnema sylvestre/chemistry , Hypoglycemic Agents/pharmacology , Pterocarpus/chemistry , Syzygium/chemistry , Administration, Oral , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/enzymology , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/isolation & purification , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dose-Response Relationship, Drug , Glucagon-Like Peptide 1/blood , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/isolation & purification , India , Inhibitory Concentration 50 , Male , Medicine, Ayurvedic , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/pharmacology , Rats , Rats, Wistar , Time Factors
13.
J Pharm Pharmacol ; 65(12): 1773-84, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24117480

ABSTRACT

OBJECTIVES: Adults who develop type 2 diabetes (T2D) at later stages are at a higher risk of developing Alzheimer's disease (AD). Pharmacological agents such as dipeptidyl peptidase-4 (DPP-4) inhibitors that increase the levels of glucagon-like peptide-1 (GLP-1) and ameliorate T2D have also become promising candidates as disease-modifying agents in the treatment of AD. The present study investigates the efficacy of vildagliptin, a DPP-4 inhibitor in a streptozotocin (STZ)-induced rat model of AD. METHODS: Three months following the induction of AD by intracerebral injection of STZ, animals were orally administered with vildagliptin (2.5, 5 and 10 mg/kg) for 30 days. Dose-dependent and time-course effects of vildagliptin on memory retention were investigated during the course of treatment. Following treatment, the animals were sacrificed, and brain tissues were used to evaluate the effects of vildagliptin on hippocampal and cortical GLP-1 levels, amyloid beta (Aß) burden, tau phosphorylation and inflammatory markers. KEY FINDINGS: The results reveal a time-dependent improvement in memory retention and a dose-dependent attenuation of Aß, tau phosphorylation and inflammatory markers and increased GLP-1 levels. CONCLUSIONS: These robust therapeutic effects of vildagliptin demonstrate a unique mechanism for Aß and tau clearance and reverse the cognitive deficits and pathology observed in AD.


Subject(s)
Adamantane/analogs & derivatives , Alzheimer Disease/drug therapy , Cognition Disorders/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Nitriles/therapeutic use , Pyrrolidines/therapeutic use , Adamantane/therapeutic use , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides/analysis , Animals , Glucagon-Like Peptide 1/analysis , Hippocampus/chemistry , Interleukin-1beta/analysis , Male , Maze Learning/drug effects , Rats , Rats, Wistar , Streptozocin , Tumor Necrosis Factor-alpha/analysis , Vildagliptin , tau Proteins/analysis
14.
Neuropharmacology ; 72: 291-300, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23603201

ABSTRACT

Type 2 diabetes (T2D) is one of the major risk factors associated with Alzheimer's disease (AD). Recent studies have found similarities in molecular mechanisms that underlie the respective degenerative developments in the two diseases. Pharmacological agents, such as dipeptidyl peptidase-4 (DPP-4) inhibitors, which increase the level of glucagon-like peptide-1 (GLP-1) and ameliorate T2D, have become valuable candidates as disease modifying agents in the treatment of AD. In addition, endogenous GLP-1 levels decrease amyloid beta (Aß) peptide and tau phosphorylation in AD. The present study examines the efficacy of Saxagliptin, a DPP-4 inhibitor in a streptozotocin (STZ) induced rat model of AD. Three months following induction of AD by intracerebral administration of streptozotocin, animals were orally administered Saxagliptin (0.25, 0.5 and 1 mg/kg) for 60 days. The effect of the DPP-4 inhibitor on hippocampal GLP-1 levels, Aß burden, tau phosphorylation, inflammatory markers and memory retention were evaluated. The results reveal an attenuation of Aß, tau phosphorylation and inflammatory markers and an improvement in hippocampal GLP-1 and memory retention following treatment. This remarkable therapeutic effect of Saxagliptin mediated through DPP-4 inhibition demonstrates a unique mechanism for Aß and tau clearance by increasing GLP-1 levels and reverses the behavioural deficits and pathology observed in AD.


Subject(s)
Adamantane/analogs & derivatives , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Antibiotics, Antineoplastic/toxicity , Dipeptides/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Streptozocin/toxicity , Adamantane/therapeutic use , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Gene Expression Regulation/drug effects , Glucagon-Like Peptide 1/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Interleukin-1beta/metabolism , Male , Maze Learning/drug effects , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...