Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Oncol Res ; 31(2): 125-139, 2023.
Article in English | MEDLINE | ID: mdl-37304233

ABSTRACT

This research aimed to explore the influence of Src homology-2 containing protein tyrosine phosphatase (SHP-2) on the functions of tyrosine kinase receptors with immunoglobulin and EGF homology domains 2 (Tie2)-expressing monocyte/macrophages (TEMs) and the influence of the angiopoietin(Ang)/Tie2-phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) (Ang/Tie2-PI3K/Akt/mTOR) signaling pathway on the tumor microvascular remodeling in an immunosuppressive microenvironment. In vivo, SHP-2-deficient mice were used to construct colorectal cancer (CRC) liver metastasis models. SHP-2-deficient mice had significantly more metastatic cancer and inhibited nodules on the liver surface than wild-type mice, and the high-level expression of p-Tie2 was found in the liver tissue of the macrophages' specific SHP-2-deficient mice (SHP-2MAC-KO) + planted tumor mice. Compared with the SHP-2 wild type mice (SHP-2WT) + planted tumor group, the SHP-2MAC-KO + planted tumor group experienced increased expression of p-Tie2, p-PI3K, p-Akt, p-mTOR, vascular endothelial growth factor (VEGF), cyclooxygenase-2 (COX-2), matrix metalloproteinase 2 (MMP2), and MMP9 in the liver tissue. TEMs selected by in vitro experiments were co-cultured with remodeling endothelial cells and tumor cells as carriers. It was found that when Angpt1/2 was used for stimulation, the SHP-2MAC-KO + Angpt1/2 group displayed evident increases in the expression of the Ang/Tie2-PI3K/Akt/mTOR pathway. The number of cells passing through the lower chamber and the basement membrane and the number of blood vessels formed by cells compared with the SHP-2WT + Angpt1/2 group, while these indexes were subjected to no changes under the simultaneous stimulation of Angpt1/2 + Neamine. To sum up, the conditional knockout of SHP-2 can activate the Ang/Tie2-PI3K/Akt/mTOR pathway in TEMs, thereby strengthening tumor micro angiogenesis in the microenvironment and facilitating CRC liver metastasis.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Animals , Humans , Mice , Colorectal Neoplasms/genetics , Endothelial Cells , Liver Neoplasms/genetics , Matrix Metalloproteinase 2 , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases , Tumor Microenvironment , Vascular Endothelial Growth Factor A , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Receptor, TIE-2
2.
Front Oncol ; 13: 1192192, 2023.
Article in English | MEDLINE | ID: mdl-37274227

ABSTRACT

Objective: The intent of this study was to investigate the relationship between oxidative stress and treatment response in gastric cancer patients undergoing neoadjuvant chemotherapy. Methods: Blood samples from 108 patients and 108 healthy subjects were collected, and all patients were enrolled in SOX chemotherapy. The patients received four cycles of neoadjuvant chemotherapy. Blood samples were collected to determine oxidative stress levels at baseline prior to beginning chemotherapy, and at the end of cycles 2 and 4. The patients receiving neoadjuvant chemotherapy were followed up for several months to years. A survival curve was created according to the follow-up information from the patients. In addition, the correlation between oxidative stress level and treatment effect was evaluated and ROC curves were plotted according to the final collected data. Results: Compared with the normal group, the levels of the antioxidant index decreased while the peroxide index increased in the patients. Conversely, when patients were compared before and after chemotherapy, the antioxidant index increased but the peroxide index decreased. Furthermore, the antioxidant index increased in the response group while the peroxide index decreased in the non-response group. Conclusion: Patients with an increased antioxidant index after chemotherapy have good treatment responsiveness. These indicators can also be used as predictors to judge the patients' response to chemotherapy.

3.
Front Endocrinol (Lausanne) ; 13: 887238, 2022.
Article in English | MEDLINE | ID: mdl-35712239

ABSTRACT

Background: Chronic pain is defined as pain that persists typically for a period of over six months. Chronic pain is often accompanied by an anxiety disorder, and these two tend to exacerbate each other. This can make the treatment of these conditions more difficult. Glucose-dependent insulinotropic polypeptide (GIP) is a member of the incretin hormone family and plays a critical role in glucose metabolism. Previous research has demonstrated the multiple roles of GIP in both physiological and pathological processes. In the central nervous system (CNS), studies of GIP are mainly focused on neurodegenerative diseases; hence, little is known about the functions of GIP in chronic pain and pain-related anxiety disorders. Methods: The chronic inflammatory pain model was established by hind paw injection with complete Freund's adjuvant (CFA) in C57BL/6 mice. GIP receptor (GIPR) agonist (D-Ala2-GIP) and antagonist (Pro3-GIP) were given by intraperitoneal injection or anterior cingulate cortex (ACC) local microinjection. Von Frey filaments and radiant heat were employed to assess the mechanical and thermal hypersensitivity. Anxiety-like behaviors were detected by open field and elevated plus maze tests. The underlying mechanisms in the peripheral nervous system and CNS were explored by GIPR shRNA knockdown in the ACC, enzyme-linked immunosorbent assay, western blot analysis, whole-cell patch-clamp recording, immunofluorescence staining and quantitative real-time PCR. Results: In the present study, we found that hind paw injection with CFA induced pain sensitization and anxiety-like behaviors in mice. The expression of GIPR in the ACC was significantly higher in CFA-injected mice. D-Ala2-GIP administration by intraperitoneal or ACC local microinjection produced analgesic and anxiolytic effects; these were blocked by Pro3-GIP and GIPR shRNA knockdown in the ACC. Activation of GIPR inhibited neuroinflammation and activation of microglia, reversed the upregulation of NMDA and AMPA receptors, and suppressed the enhancement of excitatory neurotransmission in the ACC of model mice. Conclusions: GIPR activation was found to produce analgesic and anxiolytic effects, which were partially due to attenuation of neuroinflammation and inhibition of excitatory transmission in the ACC. GIPR may be a suitable target for treatment of chronic inflammatory pain and pain-related anxiety.


Subject(s)
Chronic Pain , Receptors, Gastrointestinal Hormone , Animals , Chronic Pain/drug therapy , Chronic Pain/metabolism , Freund's Adjuvant , Gastric Inhibitory Polypeptide/physiology , Gyrus Cinguli/metabolism , Mice , Mice, Inbred C57BL , RNA, Small Interfering , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/metabolism
4.
Oxid Med Cell Longev ; 2020: 6576718, 2020.
Article in English | MEDLINE | ID: mdl-31998440

ABSTRACT

BACKGROUND: Infrasound is a major threat to global health by causing injuries of the central nervous system (CNS). However, there remains no effective therapeutic agent for preventing infrasound-caused CNS injury. 2,3,5,4'-Tetrahydroxystilbene-2-O-ß-D-glycoside (THSG) exerts protective function against CNS injuries and may have beneficial effects on infrasound-induced CNS impairment. METHODS: A mouse model with CNS (oxidative stress-induced inflammation and neuronal apoptosis) injuries was established when the mouse was exposed to the infrasound of 16 Hz at 130 dB for 2 h each day and the duration of treatment was 8 d. The mice were divided into the control (CG, healthy mice), the model (MG, model mice), and the THSG (EG, experimental group, model mice treated with THSG) groups. The learning and memory impairments caused by infrasound were examined using a Morris water maze test. Lipid profiles, antioxidant biomarkers, and inflammatory cytokines in hippocampus tissue were measured by using corresponding ELISA kits. Meanwhile, BCL-2/BAX/caspase-3 signaling pathway was measured in the hippocampi and prefrontal cortex of the mouse brain using real-time qPCR and Western blot. Nissl's stain was used to measure neuronal necrosis in the hippocampi and prefrontal cortex of the mouse brain. RESULTS: THSG significantly ameliorated the learning and memory impairments caused by infrasound. On the other hand, THSG improved lipid profiles, increased antioxidant properties by affecting the levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), and malondialdehyde (MDA), and displayed anti-inflammatory action via the downregulation of IL- (interleukin-) 6, IL-8, IL-10, TNF- (tumor necrosis factor-) α, and hs-CRP (high-sensitivity C-reactive protein) in the hippocampal tissues of the mouse model (P < 0.05). Additionally, Nissl's stain showed that THSG inhibited infrasound-induced neuronal necrosis in the hippocampi and prefrontal cortex. Besides, THSG exerted antiapoptosis function by upregulating the level of Bcl-2 and downregulating the levels of BAX and caspase-3 in the hippocampi. CONCLUSION: THSG may be an effective anti-infrasound drug against CNS injury by improving antioxidant, anti-inflammatory, antiapoptosis, and antinecrosis capacities. Further research is still needed to confirm the exact molecular mechanism.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Central Nervous System , Glucosides/pharmacology , Stilbenes/pharmacology , Ultrasonic Waves/adverse effects , Animals , Central Nervous System/injuries , Central Nervous System/metabolism , Central Nervous System/pathology , Cytokines/metabolism , Disease Models, Animal , Inflammation/drug therapy , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Mice
6.
J Clin Invest ; 129(6): 2333-2350, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063987

ABSTRACT

Hormone therapy (HT) is reported to be deficient in improving learning and memory in older postmenopausal women according to recent clinical studies; however, the reason for failure is unknown. A "window of opportunity" for estrogen treatment is proposed to explain this deficiency. Here, we found that facilitation of memory extinction and long-term depression by 17ß-estradiol (E2) was normal in mice 1 week after ovariectomy (OVXST), but it was impaired in mice 3 months after ovariectomy (OVXLT). High-throughput sequencing revealed a decrease of miR-221-5p, which promoted cannabinoid receptor 1 (CB1) ubiquitination by upregulation of Neurl1a/b in E2-treated OVXLT mice. Blood samples from postmenopausal women aged 56-65 indicated decreases of miR-221-5p and 2-arachidonoylglycerol compared with samples from perimenopausal women aged 46-55. Replenishing of miR-221-5p or treatment with a CB1 agonist rescued the impairment of fear extinction in E2-treated OVXLT mice. The present study demonstrates that an HT time window in mice can be prolonged by cotreatment with a CB1 agonist, implying a potential strategy for HT in long-term menopausal women.


Subject(s)
Hormone Replacement Therapy , Ovariectomy , Postmenopause/drug effects , Receptor, Cannabinoid, CB1/agonists , Aged , Animals , Estradiol/pharmacology , Female , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Postmenopause/genetics , Postmenopause/metabolism , Receptor, Cannabinoid, CB1/biosynthesis , Up-Regulation/drug effects
7.
Brain Res Bull ; 149: 75-85, 2019 07.
Article in English | MEDLINE | ID: mdl-31005665

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) inhibitors increase the incidence of pain in patients, and this finding has been replicated in animal models. However, reports on possible analgesics for this condition are scant. Accumulating evidence finds that nicotinic acetylcholine receptors (nAChRs) are involved in mediating pain. However, whether nicotine, a full agonist of nAChRs, alleviates mTORC1 inhibition-induced pain and its underlying mechanisms remain unknown. In this study, pain was induced in naïve male C57BL/6J mice by intraperitoneally injecting rapamycin acutely or repeatedly. Subsequently, pain thresholds, including mechanical and thermal pain, were measured. The involving signaling pathway was tested using western blot analysis and immunofluorescent assay. Changes in neuronal excitability caused by different treatments were also analyzed using whole-cell recording. Microinjection into the anterior cingulate cortex (ACC) was used to test the role of nAChRs containing the α4ß2 or α7 subtype in this brain region in pain modulation. Our results showed that nicotine significantly reduced hyperalgesia in mice that received acute or repeated rapamycin injections, and reversed the effects of rapamycin on the phosphorylation of S6K, 4E-BP1, insulin receptor substrate-1 (IRS-1) at Ser636/639, AKT at Ser473, and ERK at Thr202/Tyr204. Whole-cell recording results showed that nicotine reduced the firing rates of pyramidal neurons in the ACC, and a pharmacological blockade of nAChRs containing the α4ß2 or α7 subtype in ACC inhibited the antinociceptive effects of nicotine in mice with rapamycin-induced pain. Our findings indicate that analgesics targeting nAChRs can be developed to help patients with rapamycin-induced pain.


Subject(s)
Nicotine/pharmacology , Pain/drug therapy , Analgesics/pharmacology , Animals , Insulin Receptor Substrate Proteins/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Nicotine/metabolism , Pain/physiopathology , Pain Threshold/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
8.
Mol Brain ; 12(1): 36, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30961625

ABSTRACT

Chronic pain is commonly accompanied with anxiety disorder, which complicates treatment. In this study, we investigated the analgesic and anxiolytic effects of Formononetin (FMNT), an active component of traditional Chinese medicine red clover (Trifolium pratense L.) that is capable of protecting neurons from N-methyl-D-aspartate (NMDA)-evoked excitotoxic injury, on mice suffering from complete Freund's adjuvant (CFA)-induced chronic inflammatory pain. The results show that FMNT administration significantly reduces anxiety-like behavior but does not affect the nociceptive threshold in CFA-injected mice. The treatment reverses the upregulation of NMDA, GluA1, and GABAA receptors, as well as PSD95 and CREB in the basolateral amygdala (BLA). The effects of FMNT on NMDA receptors and CREB binding protein (CBP) were further confirmed by the potential structure combination between these compounds, which was analyzed by in silico docking technology. FMNT also inhibits the activation of the NF-κB signaling pathway and microglia in the BLA of mice suffering from chronic inflammatory pain. Therefore, the anxiolytic effects of FMNT are partially due to the attenuation of inflammation and neuronal hyperexcitability through the inhibition of NMDA receptor and CBP in the BLA.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety/therapy , Inflammation/pathology , Isoflavones/therapeutic use , Animals , Anti-Anxiety Agents/pharmacology , Basolateral Nuclear Complex/metabolism , Behavior, Animal/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Freund's Adjuvant , Isoflavones/chemistry , Isoflavones/pharmacology , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Models, Molecular , NF-kappa B/metabolism , NF-kappa B/pharmacokinetics , Pain/drug therapy , Receptors, GABA/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Up-Regulation/drug effects
9.
Mol Pain ; 14: 1744806918814367, 2018.
Article in English | MEDLINE | ID: mdl-30380983

ABSTRACT

Tetrahydroxystilbene glucoside (THSG) is one of the active ingredients of Polygonum multiflorum. It has been shown to exert a variety of pharmacological effects, including antioxidant, anti-aging, and anti-atherosclerosis. Because of its prominent anti-inflammatory effect, we explored whether THSG had analgesic effect. In this study, we used a model of chronic inflammatory pain caused by injecting complete Freund's adjuvant into the hind paw of mice. We found THSG relieved swelling and pain in the hind paw of mice on a dose-dependent manner. In the anterior cingulate cortex, THSG suppressed the upregulation of GluN2B-containing N-methyl-D-aspartate receptors and the downregulation of GluN2A-containing N-methyl-D-aspartate receptors caused by chronic inflammation. In addition, THSG increased Bcl-2 and decreased Bax and Caspase-3 expression by protecting neuronal survival. Furthermore, THSG inhibited the phosphorylation of p38 and the increase of nuclear factor κB (NF-κB) and tumor necrosis factor α (TNF-α). Immunohistochemical staining revealed that THSG blocked the activation of microglia and reduced the release of proinflammatory cytokines TNF-α, interleukin 1ß (IL-1ß), and interleukin 6 (IL-6). In conclusion, this study demonstrated that THSG had a certain effect on alleviating complete Freund's adjuvant-induced chronic inflammatory pain.


Subject(s)
Apoptosis , Chronic Pain/drug therapy , Glucosides/therapeutic use , Gyrus Cinguli/metabolism , Gyrus Cinguli/pathology , Inflammation/drug therapy , Microglia/pathology , Receptors, N-Methyl-D-Aspartate/metabolism , Stilbenes/therapeutic use , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Chronic Pain/complications , Chronic Pain/pathology , Cytokines/metabolism , Edema/drug therapy , Freund's Adjuvant/administration & dosage , Glucosides/chemistry , Glucosides/pharmacology , Gyrus Cinguli/drug effects , Hyperalgesia/complications , Hyperalgesia/drug therapy , Inflammation/complications , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , NF-kappa B/metabolism , Signal Transduction , Stilbenes/chemistry , Stilbenes/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Mol Brain ; 10(1): 38, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28800762

ABSTRACT

The G protein-coupled receptor 55 (GPR55) is a novel cannabinoid receptor, whose exact role in anxiety remains unknown. The present study was conducted to explore the possible mechanisms by which GPR55 regulates anxiety and to evaluate the effectiveness of O-1602 in the treatment of anxiety-like symptoms. Mice were exposed to two types of acute stressors: restraint and forced swimming. Anxiety behavior was evaluated using the elevated plus maze and the open field test. We found that O-1602 alleviated anxiety-like behavior in acutely stressed mice. We used lentiviral shRNA to selective ly knockdown GPR55 in the medial orbital cortex and found that knockdown of GPR55 abolished the anxiolytic effect of O-1602. We also used Y-27632, a specific inhibitor of ROCK, and U73122, an inhibitor of PLC, and found that both inhibitors attenuated the effectiveness of O-1602. Western blot analysis revealed that O-1602 downregulated the expression of GluA1 and GluN2A in mice. Taken together, these results suggest that GPR55 plays an important role in anxiety and O-1602 may have therapeutic potential in treating anxiety-like symptoms.


Subject(s)
Anxiety/metabolism , Anxiety/psychology , Prefrontal Cortex/metabolism , Receptors, Cannabinoid/metabolism , Stress, Psychological/metabolism , Acute Disease , Amides/administration & dosage , Amides/pharmacology , Amides/therapeutic use , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Cannabidiol/analogs & derivatives , Chronic Disease , Cyclohexanes/pharmacology , Cyclohexanes/therapeutic use , Estrenes/pharmacology , Gene Knockdown Techniques , Injections, Intraperitoneal , Male , Mice, Inbred C57BL , Pyridines/administration & dosage , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrrolidinones/pharmacology , Resorcinols/pharmacology , Resorcinols/therapeutic use , Restraint, Physical , Signal Transduction , Stress, Psychological/drug therapy , Swimming
11.
Neuroreport ; 28(5): 259-267, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28240721

ABSTRACT

Cucurbitacin IIa (CuIIa) is the major active component of the Helmseya amabilis root and is known to have antiviral and anti-inflammatory effects. In this study, we examined the antidepressant-like effects of CuIIa in a mouse model of chronic unpredictable mild stress (CUMS) and investigated the possible underlying mechanisms. To evaluate the antidepressant-like effects of CuIIa on depression-like behaviors, mice were subjected to the open-field test, the elevated plus-maze test, the forced-swimming test, and the tail-suspension test. We found that CuIIa treatment reversed the CUMS-induced behavioral abnormalities. Western blot analyses showed that CUMS significantly decreased brain-derived neurotrophic factor (BDNF) levels, cAMP-response element binding protein (CREB), and calcium/calmodulin-dependent protein kinase II (CaMKII) phosphorylation, and N-methyl-D-aspartate receptor subtype GluN2B and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor GluA1 expression in the amygdala; in addition, the expression of gamma-aminobutyric acid receptor A subunit α2 was upregulated in CUMS mice. These CUMS-induced changes were all normalized by CuIIa treatment and administration of the BDNF antagonist ANA-12 can block the antidepressant effect of CuIIa. Our findings suggest that the antidepressant-like effects of CuIIa may be exerted by regulation of the CaMKIIα-CREB-BDNF pathway and the balance between excitatory and inhibitory synaptic transmission in the amygdala.


Subject(s)
Antidepressive Agents/therapeutic use , Cucurbitacins/therapeutic use , Depression/drug therapy , Depression/etiology , Stress, Psychological/complications , Amygdala/drug effects , Amygdala/metabolism , Animals , Azepines/therapeutic use , Benzamides/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , CREB-Binding Protein/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Hindlimb Suspension , Male , Maze Learning/drug effects , Mice , Mice, Inbred BALB C , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Psychological/drug therapy , Swimming/psychology
12.
Biomed Pharmacother ; 86: 81-87, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27939523

ABSTRACT

Huntington's disease (HD) is an autosomal dominant inherited disease characterized by movement, psychiatric, and cognitive disorders. Previous research suggests that Praeruptorin C (Pra-C), an effective component in the root of Peucedanum praeruptorum dunn, a traditional Chinese medicine, may function in neuroprotection. The present study was conducted to evaluate the effectiveness of Pra-C in the treatment of HD-like symptoms in a 3-nitropropionic acid (3-NP) mouse model, and to explore the possible mechanism of the drug's activity. We treated 3-NP-injected mice with two different doses of Pra-C (1.5 and 3.0mg/kg) for 3 days. Motor behavior was tested using the open field test (OFT) and rotarod test, while psychiatric symptoms were tested using the forced swimming test (FST) and tail suspension test (TST). We found that Pra-C alleviated the motor deficits and depression-like behavior in the 3-NP-treated mice, and protected neurons from excitotoxicity. Western blot analysis revealed that Pra-C upregulated BDNF, DARPP32, and huntingtin protein in the striatum of 3-NP mice. These results taken together suggest that Pra-C may have therapeutic potential with respect to the movement, psychiatric, and cognitive symptoms of HD.


Subject(s)
Coumarins/therapeutic use , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Huntington Disease/chemically induced , Huntington Disease/drug therapy , Nitro Compounds/toxicity , Propionates/toxicity , Animals , Dose-Response Relationship, Drug , Huntington Disease/metabolism , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Treatment Outcome
13.
PLoS One ; 11(2): e0148827, 2016.
Article in English | MEDLINE | ID: mdl-26863436

ABSTRACT

Diabetes is a major risk factor for the development of stroke. Glucagon-like peptide-1 receptor (GLP-1R) agonists have been in clinical use for the treatment of diabetes and also been reported to be neuroprotective in ischemic stroke. The quinoxaline 6,7-dichloro-2-methylsulfonyl-3-N-tert- butylaminoquinoxaline (DMB) is an agonist and allosteric modulator of the GLP-1R with the potential to increase the affinity of GLP-1 for its receptor. The aim of this study was to evaluate the neuroprotective effects of DMB on transient focal cerebral ischemia. In cultured cortical neurons, DMB activated the GLP-1R, leading to increased intracellular cAMP levels with an EC50 value about 100 fold that of exendin-4. Pretreatment of neurons with DMB protected against necrotic and apoptotic cell death was induced by oxygen-glucose deprivation (OGD). The neuroprotective effects of DMB were blocked by GLP-1R knockdown with shRNA but not by GLP-1R antagonism. In C57BL/6 mice, DMB was orally administered 30 min prior to middle cerebral artery occlusion (MCAO) surgery. DMB markedly reduced the cerebral infarct size and neurological deficits caused by MCAO and reperfusion. The neuroprotective effects were mediated by activation of the GLP-1R through the cAMP-PKA-CREB signaling pathway. DMB exhibited anti-apoptotic effects by modulating Bcl-2 family members. These results provide evidence that DMB, a small molecular GLP-1R agonist, attenuates transient focal cerebral ischemia injury and inhibits neuronal apoptosis induced by MCAO. Taken together, these data suggest that DMB is a potential neuroprotective agent against cerebral ischemia.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/administration & dosage , Quinoxalines/administration & dosage , Sulfones/administration & dosage , Administration, Oral , Allosteric Regulation , Animals , Apoptosis/drug effects , Blood Glucose , Brain/blood supply , Brain/drug effects , Brain/pathology , Cell Survival/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Exenatide , Infarction, Middle Cerebral Artery/blood , Inhibitory Concentration 50 , Insulin/blood , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Peptides/pharmacology , Primary Cell Culture , Reperfusion Injury/prevention & control , Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...