Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
1.
Respir Res ; 20(1): 222, 2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31623614

ABSTRACT

Lung cancer (LC) has the highest relative risk of development as a comorbidity of chronic obstructive pulmonary disease (COPD). The molecular mechanisms that mediate chronic inflammation and lung function impairment in COPD have been identified in LC. This suggests the two diseases are more linked than once thought. Emerging data in relation to a key phosphatase, protein phosphatase 2A (PP2A), and its regulatory role in inflammatory and tumour suppression in both disease settings suggests that it may be critical in the progression of COPD to LC. In this review, we uncover the importance of the functional and active PP2A holoenzyme in the context of both diseases. We describe PP2A inactivation via direct and indirect means and explore the actions of two key PP2A endogenous inhibitors, cancerous inhibitor of PP2A (CIP2A) and inhibitor 2 of PP2A (SET), and the role they play in COPD and LC. We explain how dysregulation of PP2A in COPD creates a favourable inflammatory micro-environment and promotes the initiation and progression of tumour pathogenesis. Finally, we highlight PP2A as a druggable target in the treatment of COPD and LC and demonstrate the potential of PP2A re-activation as a strategy to halt COPD disease progression to LC. Although further studies are required to elucidate if PP2A activity in COPD is a causal link for LC progression, studies focused on the potential of PP2A reactivating agents to reduce the risk of LC formation in COPD patients will be pivotal in improving clinical outcomes for both COPD and LC patients in the future.


Subject(s)
Disease Progression , Lung Neoplasms/diagnosis , Lung Neoplasms/enzymology , Protein Phosphatase 2/metabolism , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/enzymology , Animals , Autoantigens/administration & dosage , Humans , Intracellular Signaling Peptides and Proteins/administration & dosage , Lung Neoplasms/drug therapy , Membrane Proteins/administration & dosage , Protein Phosphatase 2/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/drug therapy
2.
Exp Physiol ; 104(12): 1892-1910, 2019 12.
Article in English | MEDLINE | ID: mdl-31631436

ABSTRACT

NEW FINDINGS: • What is the central question of this study? We hypothesized that central inflammatory processes that involve activation of microglia and astrocytes contribute to the development of Gαi2 protein-dependent, salt-sensitive hypertension. • What is the main finding and its importance? The main finding is that PVN-specific inflammatory processes, driven by microglial activation, appear to be linked to the development of Gαi2 protein-dependent, salt-sensitive hypertension in Sprague-Dawley rats. This finding might reveal new mechanistic targets in the treatment of hypertension. ABSTRACT: The central mechanisms underlying salt-sensitive hypertension, a significant public health issue, remain to be established. Researchers in our laboratory have reported that hypothalamic paraventricular nucleus (PVN) Gαi2 proteins mediate the sympathoinhibitory and normotensive responses to high sodium intake in salt-resistant rats. Given the recent evidence of central inflammation in animal models of hypertension, we hypothesized that PVN inflammation contributes to Gαi2 protein-dependent, salt-sensitive hypertension. Male Sprague-Dawley rats received chronic intracerebroventricular infusions of a targeted Gαi2 or control scrambled oligodeoxynucleotide (ODN) and were maintained for 7 days on a normal-salt (NS; 0.6% NaCl) or high-salt (HS; 4% NaCl) diet; in subgroups on HS, intracerebroventricular minocycline (microglial inhibitor) was co-infused with ODNs. Radiotelemetry was used in subgroups of rats to measure mean arterial pressure (MAP) chronically. In a separate group of rats, plasma noradrenaline, plasma renin activity, urinary angiotensinogen and mRNA levels of the PVN pro-inflammatory cytokines TNFα, IL-1ß and IL-6 and the anti-inflammatory cytokine IL-10 were assessed. In additional groups, immunohistochemistry was performed for markers of PVN and subfornical organ microglial activation and cytokine levels and PVN astrocyte activation. High salt intake evoked salt-sensitive hypertension, increased plasma noradrenaline, PVN pro-inflammatory cytokine mRNA upregulation, anti-inflammatory cytokine mRNA downregulation and PVN-specific microglial activation in rats receiving a targeted Gαi2 but not scrambled ODN. Minocycline co-infusion significantly attenuated the increase in MAP and abolished the increase in plasma noradrenaline and inflammation in Gαi2 ODN-infused animals on HS. Our data suggest that central Gαi2 protein prevents microglial-mediated PVN inflammation and the development of salt-sensitive hypertension.


Subject(s)
Hypertension/metabolism , Intracellular Signaling Peptides and Proteins/administration & dosage , Microglia/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Sodium Chloride, Dietary/administration & dosage , Animals , Hypertension/chemically induced , Hypertension/pathology , Infusions, Intraventricular , Male , Microglia/drug effects , Oligodeoxyribonucleotides/administration & dosage , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/pathology , Rats , Rats, Sprague-Dawley
3.
Reproduction ; 158(1): 95-107, 2019 07.
Article in English | MEDLINE | ID: mdl-31022702

ABSTRACT

Inflammation plays a pivotal role in the terminal process of human labor and delivery, including myometrial contractions and membrane rupture. TNF-alpha-induced protein 8-like-2 (TIPE2) is a novel inflammation regulator; however, there are no studies on the role of TIPE2 in human labor. We report that in myometrium, there is decreased TIPE2 mRNA expression during late gestation which was further decreased in labor. In fetal membranes, TIPE2 mRNA expression was decreased with both term and preterm labor compared to no labor samples. Knockdown of TIPE2 by siRNA in primary myometrium and amnion cells was associated with an augmentation of IL1B and TNF-induced expression of pro-inflammatory cytokines and chemokines; expression of contraction-associated proteins and secretion of the uterotonic prostaglandin PGF2α and expression of extracellular matrix degrading enzymes. In TIPE2-deficient myometrial cells treated with inhibitors of NF-κB or ERK1/2, the secretion of pro-labor mediators was reduced back to control levels. In conclusion, these in vitro experiments indicate that loss of TIPE2 exacerbates the inflammatory response.


Subject(s)
Amnion/drug effects , Anti-Inflammatory Agents/administration & dosage , Inflammation/drug therapy , Intracellular Signaling Peptides and Proteins/administration & dosage , Labor, Obstetric/drug effects , MAP Kinase Signaling System/drug effects , Myometrium/drug effects , Adult , Amnion/immunology , Amnion/metabolism , Cytokines/metabolism , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Labor, Obstetric/immunology , Labor, Obstetric/metabolism , Myometrium/immunology , Myometrium/metabolism , NF-kappa B/metabolism , Pregnancy
4.
J Neuropathol Exp Neurol ; 78(5): 426-435, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30888409

ABSTRACT

We have previously demonstrated that R18 and its d-enantiomer, R18D, are neuroprotective at 24 hours following intraluminal filament occlusion of the middle cerebral artery (MCAO) in the rat. This study examined R18 and R18D effectiveness in improving functional outcomes at up to 56 days poststroke following endothelin-1-induced MCAO. Peptides were administered intravenously at doses of 100, 300, or 1000 nmol/kg, 60 minutes after MCAO. Functional recovery poststroke was assessed using multiple forelimb placing tests and horizontal ladder test, and NA-1 (TAT-NR2B9c), a neuroprotective currently in phase 3 clinical stroke trials, was used as a benchmark. The study demonstrated that R18 (300 and 1000 nmol/kg) was the most effective peptide in improving functional outcomes, followed by R18D (300 and 1000 nmol/kg), and NA-1 (300 and 100 nmol/kg). Furthermore, R18 at doses of 300 and 1000 nmol/kg was the most effective agent in restoring pre-stroke body weight, while R18 and R18D at doses of 300 and 1000 nmol/kg, but not NA-1 also significantly reduced the number of animals requiring hand feeding 48 hours after stroke. This study confirms that R18 and R18D are effective in improving long-term functional outcomes after stroke, and suggests that R18 may be more effective than NA-1.


Subject(s)
Endothelin-1/toxicity , Intracellular Signaling Peptides and Proteins/administration & dosage , Peptides/administration & dosage , Recovery of Function/physiology , Stroke/chemically induced , Stroke/drug therapy , Animals , Infusions, Intravenous , Male , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Stroke/physiopathology , Treatment Outcome
5.
Curr Drug Deliv ; 16(3): 226-232, 2019.
Article in English | MEDLINE | ID: mdl-30381074

ABSTRACT

BACKGROUND: Humanin is a novel neuronal peptide that has displayed potential in the treatment of Alzheimer's Disease through the suppression of inflammatory IL-6 cytokine receptors. Such receptors are found throughout the body, including the eye, suggesting its other potential applications. Age-related Macular Degeneration (AMD) is the leading cause of blindness in the developing world. There is no cure for this disease, and current treatments have several negative side effects associated with them, making finding other treatment options desirable. OBJECTIVE: In this study, the potential applications in treating AMD for a more potent humanin derivative, AGA-HNG, were studied. METHODS: AGA-HNG was synthesized and encapsulated in chitosan Nanoparticles (NPs), which were then characterized for their size, Encapsulation Efficiency (EE), and drug release. Their ability to suppress VEGF secretion and protect against oxidative apoptosis was studied in vitro using ARPE-19 cells. The chitosan NPs exhibited similar anti-VEGF properties and oxidative protection as the free protein while exhibiting superior pharmaceutical characteristics including biocompatibility and drug release. RESULTS: Drug-loaded NPs exhibited a radius of 346nm with desirable pharmacokinetic properties including a stable surface charge (19.5 ± 3.7 mV) and steady drug release capacity. AGA-HNG showed great promise in mediating apoptosis in hypoxic cells. They were also able to significantly reduce VEGF expression in vitro with reduced cellular toxicity compared to the free drug. CONCLUSION: The ability of this drug delivery system to reduce retinal apoptosis with desirable pharmacokinetic and biocompatible properties makes this a promising therapeutic option for AMD.


Subject(s)
Chitosan/administration & dosage , Intracellular Signaling Peptides and Proteins/administration & dosage , Nanoparticles/administration & dosage , Cell Line , Cell Survival/drug effects , Chitosan/chemistry , Drug Delivery Systems , Drug Liberation , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Macular Degeneration , Nanoparticles/chemistry , Vascular Endothelial Growth Factor A/metabolism
6.
Am J Physiol Heart Circ Physiol ; 315(3): H634-H643, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29775411

ABSTRACT

The chemotherapeutic effect of doxorubicin (Dox) is limited by cumulative dose-dependent cardiotoxicity in cancer survivors. Dexrazoxane (DRZ) is approved to prevent Dox-induced cardiotoxicity. Humanin and its synthetic analog HNG have a cytoprotective effect on the heart. To investigate the cardioprotective efficacy of HNG alone or in combination with DRZ against Dox-induced cardiotoxicity, 80 adult male mice were randomly divided into 8 groups to receive the following treatments via intraperitoneal injection: saline dailym HNG (5 mg/kg) daily, DRZ (60 mg/kg) weekly, Dox (3 mg/kg) weekly, DRZ + HNG, Dox + HNG, Dox + DRZ, and Dox + HNG + DRZ. Echocardiograms were performed before and at 4, 8, and 9.5 wk after the beginning of treatment. All mice were euthanized at 10 wk. In the absence of Dox, HNG, DRZ, or DRZ + HNG had no adverse effect on the heart. Dox treatment caused decreases in ejection fraction and cardiac mass and increases in cardiomyocyte apoptosis and intracardiac fibrosis. HNG or DRZ alone blunted the Dox-induced decrease in left ventricle posterior wall thickness and modestly ameliorated the Dox-induced decrease in ejection fraction. HNG + DRZ significantly ameliorated Dox-induced decreases in ejection function, cardiac fibrosis, and cardiac mass. Using a targeted analysis for the mitochondrial gene array and protein expression in heart tissues, we demonstrated that HNG + DRZ reversed DOX-induced altered transcripts that were biomarkers of cardiac damage and uncoupling protein-2. We conclude that HNG enhances the cardiac protective effect of DRZ against Dox-induced cardiotoxicity. HNG + DRZ protects mitochondria from Dox-induced cardiac damage and blunts the onset of cardiac dysfunction. Thus, HNG may be an adjuvant to DRZ in preventing Dox-induced cardiotoxicity. NEW & NOTEWORTHY Doxorubicin (Dox) is commonly used for treating a wide range of human cancers. However, cumulative dosage-dependent carditoxicity often limits its clinical applications. We demonstrated in this study that treating young adult male mice with synthetic humanin analog enhanced the cardiac protective effect of dexrazoxane against chemotherapeutic agent Dox-induced cardiac dysfunction. Thus, humanin analog can potentially serve as an adjuvant to dexrazoxane in more effectively preventing Dox-induced cardiac dysfunction and cardiomyopathy.


Subject(s)
Cardiotonic Agents/pharmacology , Dexrazoxane/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Myocytes, Cardiac/drug effects , Animals , Cardiotonic Agents/administration & dosage , Cardiotoxicity , Dexrazoxane/administration & dosage , Doxorubicin/toxicity , Drug Synergism , Intracellular Signaling Peptides and Proteins/administration & dosage , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism
7.
Redox Biol ; 16: 146-156, 2018 06.
Article in English | MEDLINE | ID: mdl-29502044

ABSTRACT

The endonuclease G gene (Endog), which codes for a mitochondrial nuclease, was identified as a determinant of cardiac hypertrophy. How ENDOG controls cardiomyocyte growth is still unknown. Thus, we aimed at finding the link between ENDOG activity and cardiomyocyte growth. Endog deficiency induced reactive oxygen species (ROS) accumulation and abnormal growth in neonatal rodent cardiomyocytes, altering the AKT-GSK3ß and Class-II histone deacethylases (HDAC) signal transduction pathways. These effects were blocked by ROS scavengers. Lack of ENDOG reduced mitochondrial DNA (mtDNA) replication independently of ROS accumulation. Because mtDNA encodes several subunits of the mitochondrial electron transport chain, whose activity is an important source of cellular ROS, we investigated whether Endog deficiency compromised the expression and activity of the respiratory chain complexes but found no changes in these parameters nor in ATP content. MtDNA also codes for humanin, a micropeptide with possible metabolic functions. Nanomolar concentrations of synthetic humanin restored normal ROS levels and cell size in Endog-deficient cardiomyocytes. These results support the involvement of redox signaling in the control of cardiomyocyte growth by ENDOG and suggest a pathway relating mtDNA content to the regulation of cell growth probably involving humanin, which prevents reactive oxygen radicals accumulation and hypertrophy induced by Endog deficiency.


Subject(s)
Endodeoxyribonucleases/genetics , Hypertrophy/genetics , Intracellular Signaling Peptides and Proteins/administration & dosage , Mitochondria/genetics , Animals , Apoptosis/drug effects , DNA, Mitochondrial/drug effects , Endodeoxyribonucleases/deficiency , Endodeoxyribonucleases/metabolism , Humans , Hypertrophy/drug therapy , Hypertrophy/enzymology , Hypertrophy/metabolism , Mice , Mitochondria/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
8.
Biochem Biophys Res Commun ; 497(1): 292-297, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29432738

ABSTRACT

Mitochondrial dysfunction is associated with ß-cell failure and insulin resistance in diabetes. Humanin is an endogenous cytoprotective peptide. In the current study, we aimed to define the effects of Humanin on mitochondrial biogenesis in pancreatic ß-cells. Our findings demonstrated that Humanin treatment significantly increased the expression of PGC-1α and its downstream target genes NRF1 and TFAM in MIN6 ß-cells. Notably, Humanin treatment significantly promoted mitochondrial biogenesis by increasing mitochondrial mass, elevating mtDNA/nDNA ratio, and stimulating the expression of cytochrome B, which were suppressed by the specific AMPK inhibitor compound C. Indeed, Humanin treatment caused the phosphorylation of AMPK, which was involved in the induction of PGC-1α, NRF1, and TFAM by Humanin. Importantly, our findings indicate that Humanin treatment led to a possible functional gain of the mitochondria by increasing ATP levels and respiratory rate. Our findings provided a new insight into the molecular mechanisms of action by which Humanin improves pancreatic ß-cell function via enhanced mitochondrial mass and performance.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/drug effects , Mitochondria/physiology , Organelle Biogenesis , Animals , Cell Line , Insulin-Secreting Cells/cytology , Mice , Mitochondria/ultrastructure , Treatment Outcome
9.
J Alzheimers Dis ; 61(4): 1343-1353, 2018.
Article in English | MEDLINE | ID: mdl-29376862

ABSTRACT

Cardiac ischemia-reperfusion (I/R) injury has been shown to impair brain function. Humanin analogue (HNG) given prior to cardiac ischemia has been shown to attenuate both heart and brain mitochondrial dysfunction caused by cardiac I/R injury. In a clinical setting, patients received medical treatment for acute myocardial infarction either during or after the onset of myocardial ischemia; thus, in this study, we tested the hypothesis that the administration of HNG during cardiac I/R injury has therapeutic potential for brain protection. Thirty-six male Wistar rats were divided into two groups: a cardiac I/R group (n = 30), and a sham group (n = 6). The I/R rats were then divided into five subgroups to receive: 1) vehicle; 2) HNG (84 µg/kg); 3) HNG (168 µg/kg); 4) HNG (252 µg/kg) intravenously administered during the cardiac-ischemia; and 5) HNG at 252 µg/kg given at the onset of reperfusion. At the end of treatment, brains were removed for determination of blood-brain barrier (BBB) breakdown, oxidative stress, brain mitochondrial function, brain mitochondrial dynamics, p-tau, amyloid-ß (Aß) and apoptosis. HNG at a dose of 168 and 252 µg/kg administered during ischemia, and 252 µg/kg given at the onset of reperfusion effectively attenuated the brain mitochondrial dysfunction, tau hyperphosphorylation and Aß accumulation, and apoptosis, without reducing BBB breakdown, brain oxidative stress, or mitochondrial dynamic, caused by cardiac I/R injury. In conclusion, humanin exerted neuroprotection during induced cardiac I/R injury via improvement in brain mitochondrial function, and the reduction of Alzheimer's disease pathology and apoptosis.


Subject(s)
Apoptosis/drug effects , Intracellular Signaling Peptides and Proteins/administration & dosage , Mitochondria/drug effects , Myocardial Reperfusion Injury/prevention & control , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Animals , Blood-Brain Barrier/metabolism , Brain/pathology , Male , Microscopy, Electron , Mitochondria/ultrastructure , Myocardial Reperfusion Injury/physiopathology , Neuroprotection , Oxidative Stress/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects
10.
Food Funct ; 9(1): 397-406, 2018 Jan 24.
Article in English | MEDLINE | ID: mdl-29215104

ABSTRACT

Insulin resistance caused by the overexpression of protein tyrosine phosphatase 1 B (PTP1B) as well as the dephosphorylation of its target is one of the main causes of type 2 diabetes (T2D). A newly discovered proteoglycan, Fudan-Yueyang Ganoderma lucidum (FYGL) extracted from Ganoderma lucidum, was first reported to be capable of competitively inhibiting PTP1B activity in vitro in our previous work. In the present study, we sought to reveal the mechanism of PTP1B inhibition by FYGL at the animal and cellular levels. We found that FYGL can decrease blood glucose, reduce body weight and ameliorate insulin resistance in ob/ob mice. Decrease of PTP1B expression and increase of the phosphorylation of PTP1B targets in the insulin signaling pathway of skeletal muscles were observed. In order to clearly reveal the underlying mechanism of the hypoglycemic effect caused by FYGL, we further investigated the effects of FYGL on the PTP1B-involved insulin signaling pathway in rat myoblast L6 cells. We demonstrated that FYGL had excellent cell permeability by using a confocal laser scanning microscope and a flow cytometer. We found that FYGL had a positive effect on insulin-stimulated glucose uptake by using the 2-deoxyglucose (2-DG) method. FYGL could inhibit PTP1B expression at the mRNA level, phosphorylating insulin receptor substrate-1 (IRS1), as well as activating phosphatidylinositol-3 kinase (PI3K) and protein kinase B (Akt). Finally, FYGL increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and consequently up-regulated the expression of glucose transporter type 4 (GLUT4), promoting GLUT4 transportation to the plasma membrane in PTP1B-transfected L6 cells. Our study provides theoretical evidence for FYGL to be potentially used in T2D management.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucose Transporter Type 4/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Intracellular Signaling Peptides and Proteins/administration & dosage , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Reishi/chemistry , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucose Transporter Type 4/genetics , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/isolation & purification , Mice , Mice, Inbred C57BL , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Proteoglycans/administration & dosage , Proteoglycans/chemistry , Proteoglycans/isolation & purification , Rats , Signal Transduction/drug effects
11.
J Recept Signal Transduct Res ; 37(6): 543-549, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28840751

ABSTRACT

Gastrodin, one of the major components extracted from the Chinese herb Gastrodia elata Bl., has been widely used as an anticonvulsant, sedative, analgesic and hypotensive. In our study, we aimed to investigate the effects and possible mechanisms of gastrodin on vascular KATP channels. Tension experiments were used on rat mesenteric artery rings without an endothelium. Patch clamp experiments were executed to investigate the influences of gastrodin on the membrane current in mesenteric artery smooth muscle cells. Gastrodin induced vasorelaxation in a concentration dependent manner when rat mesenteric artery rings were pre-contracted with Phenylephrine. The vasorelaxation effect was partially diminished by pre-treating with a KATP channel inhibitor, or a PKA inhibitor. With whole-cell patch-clamp recording techniques, we found that gastrodin is a activator of KATP in rat mesenteric artery smooth muscle cells, and this effect was eliminate by pre-treating with H89or PKI, PKA inhibitor. In addition, when rat vascular smooth muscle cells were treated with 100 µM gastrodin for 24 h, maximum KATP current density increased by 28.1%. The results indicate that gastrodin exerts vasorelaxation effect through activation of PKA and subsequent opening of smooth muscle KATP channels.


Subject(s)
Benzyl Alcohols/administration & dosage , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Glucosides/administration & dosage , KATP Channels/genetics , Muscle, Smooth, Vascular/metabolism , Vasodilation/drug effects , Animals , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/antagonists & inhibitors , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/chemistry , Gastrodia/chemistry , Humans , Intracellular Signaling Peptides and Proteins/administration & dosage , Isoquinolines/administration & dosage , KATP Channels/metabolism , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Muscle, Smooth, Vascular/drug effects , Organ Culture Techniques , Patch-Clamp Techniques , Phenylephrine/administration & dosage , Rats , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Vasodilation/genetics
12.
Saudi J Gastroenterol ; 23(2): 105-111, 2017.
Article in English | MEDLINE | ID: mdl-28361841

ABSTRACT

BACKGROUND/AIM: The excessive apoptosis of intestinal epithelial cells (IECs) partly accounts for the development of colonic inflammation and eventually results in ulcerative colitis (UC). Humanin, an endogenous anti-apoptotic peptide, has previously been shown to protect against Alzheimer's disease and a variety of cellular insults. The present study aimed to investigate the effects of glysin variant of humanin (HNG) on 2,4,6-trinitrobenzene sulphonic acid (TNBS)-induced colitis in rats. MATERIALS AND METHODS: Rats were divided into four groups as follows: Group 1 (n = 8): control; isotonic saline solution 0.1 ml/rat rectally, Group 2 (n = 8): TNBS colitis; 0.1 ml of a 2.5% (w/v) TNBS solution in 50% ethanol rectally, Group 3 (n = 8): 10 µM HNG, and Group 4 (n = 8): 20 µM HNG intraperitoneal (ip) on day 2 and 6 after rectal TNBS administration. Rats were sacrificed 7 days after the induction of colitis. Blood and tissue samples were harvested for biochemical and histopathological analysis. RESULTS: HNG treatment significantly ameliorated weight loss and macroscopic and microscopic scores. TNBS-induced colitis significantly increased the colonic mRNA expression of tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1ß), and caspase-3 activities in group II in comparison to the group I. HNG treatment was associated with an inhibition of mRNA expression of TNF-α and IL-1ß, and a decrease in caspase-3 activities in colon tissues in group III and IV when compared to group II. CONCLUSION: The results of this study indicate that HNG treatment may exert beneficial effects in UC by decreasing inflammatory reactions and apoptosis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Colitis, Ulcerative/drug therapy , Intracellular Signaling Peptides and Proteins/administration & dosage , Trinitrobenzenesulfonic Acid/adverse effects , Animals , Anti-Inflammatory Agents/pharmacology , Caspase 3/genetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Gene Expression Regulation/drug effects , Interleukin-1beta/genetics , Intestinal Mucosa/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Rats , Treatment Outcome , Tumor Necrosis Factor-alpha/genetics , Weight Loss/drug effects
13.
Peptides ; 83: 16-20, 2016 09.
Article in English | MEDLINE | ID: mdl-27475912

ABSTRACT

Rubimetide (Met-Arg-Trp), which had been isolated as an antihypertensive peptide from an enzymatic digest of spinach ribulose-bisphosphate carboxylase/oxygenase (Rubisco), showed anxiolytic-like activity prostaglandin (PG) D2-dependent manner in the elevated plus-maze test after administration at a dose of 0.1mg/kg (ip.) or 1mg/kg (p.o.) in male mice of ddY strain. In this study, we found that rubimetide has weak affinities for the FPR1 and FPR2, subtypes of formyl peptide receptor (FPR). The anxiolytic-like activity of rubimetide (0.1mg/kg, ip.) was blocked by WRW4, an antagonist of FPR2, but not by Boc-FLFLF, an antagonist of FPR1, suggesting that the anxiolytic-like activity was mediated by the FPR2. Humanin, an endogenous agonist peptide of the FPR2, exerted an anxiolytic-like activity after intracerebroventricular (icv) administration, which was also blocked by WRW4. MMK1, a synthetic agonist peptide of the FPR2, also exerted anxiolytic-like activity. Thus, FPR2 proved to mediate anxiolytic-like effect as the first example of central effect exerted by FPR agonists. As well as the anxiolytic-like activity of rubimetide, that of MMK1 was blocked by BW A868C, an antagonist of the DP1-receptor. Furthermore, anxiolytic-like activity of rubimetide was blocked by SCH58251 and bicuculline, antagonists for adenosine A2A and GABAA receptors, respectively. From these results, it is concluded that the anxiolytic-like activities of rubimetide and typical agonist peptides of the FPR2 were mediated successively by the PGD2-DP1 receptor, adenosine-A2A receptor, and GABA-GABAA receptor systems downstream of the FPR2.


Subject(s)
Anxiety/drug therapy , Peptide Fragments/administration & dosage , Receptor, Adenosine A2A/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, GABA-A/metabolism , Ribulose-Bisphosphate Carboxylase/administration & dosage , Adenosine A2 Receptor Antagonists/administration & dosage , Animals , Anti-Anxiety Agents/administration & dosage , Anxiety/metabolism , Bicuculline/administration & dosage , GABA-A Receptor Antagonists/administration & dosage , Humans , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Peptides/administration & dosage , Peptides/chemical synthesis , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Formyl Peptide/genetics , Receptors, GABA-A/genetics , Ribulose-Bisphosphate Carboxylase/chemistry , Ribulose-Bisphosphate Carboxylase/genetics , Ribulose-Bisphosphate Carboxylase/isolation & purification , Spinacia oleracea/chemistry , Transcription Factor DP1/antagonists & inhibitors , Transcription Factor DP1/metabolism
14.
Oncotarget ; 7(30): 46899-46912, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27384491

ABSTRACT

Humanin is a small secreted peptide that is encoded in the mitochondrial genome. Humanin and its analogues have a protective role in multiple age-related diseases including type 2 diabetes and Alzheimer's disease, through cytoprotective and neuroprotective effects both in vitro and in vivo. However, the humanin-mediated signaling pathways are not well understood. In this paper, we demonstrate that humanin acts through the GP130/IL6ST receptor complex to activate AKT, ERK1/2, and STAT3 signaling pathways. Humanin treatment increases phosphorylation in AKT, ERK 1/2, and STAT3 where PI3K, MEK, and JAK are involved in the activation of those three signaling pathways, respectively. Furthermore, old mice, but not young mice, injected with humanin showed an increase in phosphorylation in AKT and ERK1/2 in the hippocampus. These findings uncover a key signaling pathway of humanin that is important for humanin's function and also demonstrates an age-specific in vivo effect in a region of the brain that is critical for memory formation in an age-dependent manner.


Subject(s)
Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Signal Transduction , Age Factors , Alzheimer Disease/metabolism , Animals , Cell Line, Tumor , Cytokine Receptor gp130/metabolism , Diabetes Mellitus, Type 2/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/administration & dosage , Janus Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism
15.
Cardiovasc Ther ; 34(6): 404-414, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27434747

ABSTRACT

AIM: Myocardial reperfusion via the re-canalization of occluded coronary arteries is gold standard for the treatment of acute myocardial infarction. However, reperfusion itself can cause myocardial damage due to increased reactive oxygen species (ROS) production, a process known as ischemia/reperfusion (I/R) injury. Cardiac mitochondria are the major organelle of ROS production in the heart. Cardiac mitochondrial dysfunction caused by an increased ROS production can increase cardiac arrhythmia incidence, myocardial infarct size, and cardiac dysfunction. Thus, preservation of cardiac mitochondrial function is a promising pharmacological approach to reduce cardiac I/R injury. Humanin (HN), a newly discovered 24-amino acid polypeptide, has been shown to exert antioxidative stress and antiapoptotic effects. Although the cardioprotective effects of HN against I/R injury has been reported, the effect of HN on cardiac mitochondrial function has not yet been investigated. Thus, we tested the hypothesis that HN exerts its cardioprotective effects against I/R injury through the attenuation of cardiac mitochondrial dysfunction. METHODS: I/R protocol was carried out using a 30-minutes occlusion of a left anterior descending coronary artery followed by a 120-minutes of reperfusion. The plasma HN level, infarct size, arrhythmia incidence, left ventricular function, and cardiac mitochondrial function were determined. RESULTS: Endogenous HN level before I/R injury showed no difference between groups, but was markedly decreased after I/R injury. HN analogue pretreatment decreased arrhythmia incidence and infarct size, improved cardiac mitochondrial function, and attenuated cardiac dysfunction. CONCLUSIONS: Humanin analogue pretreatment exerted cardioprotective effects against I/R injury through the attenuation of cardiac mitochondrial dysfunction.


Subject(s)
Cardiotonic Agents/administration & dosage , Intracellular Signaling Peptides and Proteins/administration & dosage , Mitochondria, Heart/drug effects , Mitochondrial Diseases/prevention & control , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Animals , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Disease Models, Animal , Drug Administration Schedule , Male , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/drug effects , Rats, Wistar , Reactive Oxygen Species/metabolism , Time Factors , Ventricular Function, Left/drug effects
16.
Zhongguo Fei Ai Za Zhi ; 19(5): 241-6, 2016 May 20.
Article in Chinese | MEDLINE | ID: mdl-27215450

ABSTRACT

BACKGROUND: The curative effect of radiotherapy may be limited by the radioresistance of tumor. Mimetic compounds of Second mitochondria-derived activator of caspase (Smac) were hopeful to become new drugs of radiosensitization for tumor because they can increase radiation induced apoptosis in tumor cells. The aim of present study is to observe the radiosensitization effect of a new Smac mimetic ANTP-SmacN7 fusion peptide in H460 cell line. METHODS: In order to observe if the fusion peptide can enter into tumor cell, ANTP-SmacN7 fusion peptide was synthesized and linked by FITC. H460 cell was divided into control, radiation only, ANTP-SmacN7 only and ANTP-SmacN7 combined with radiation group. The cells were exposed by 0, 2, 4 and 6 Gy and the concentration of ANTP-SmacN7 was 20 µmol/L. Proliferation of H460 tumor cell was detected by WST-1 assay. There are four groups in the present study: control group, radiation group, ANTP-SmacN7 group and ANTP-SmacN7 combined with radiation group. Apoptosis was detected by flow cytometry at 24 and 48 hours after the treatment of all the groups. The level of caspase3 and cleaved caspase3 were detected by Western blot assay. RESULTS: ANTP-SmacN7 can enter into cells and promote the radiosensitization of H460 cell obviously (F=25.1, P<0.01, sensitivity enhancement ratio was 1.86). The treatment of ANTP-SmacN7 combined with radiation decreased the cloning forming efficiency (χ2=45.2, P<0.01; χ2=40.3, P<0.01), activated caspase3 by promoting the expression of cleaved caspase3 and increased the apoptosis of H460 cell line. CONCLUSIONS: ANTP-SmacN7 fusion peptide had remarkably radiosensitization effect on H460 cell line. ANTP-SmacN7 fusion peptide might be hopeful to be applied in radiosensitization therapy as a new Smac mimetic polypeptide in the future.


Subject(s)
Intracellular Signaling Peptides and Proteins/administration & dosage , Lung Neoplasms/radiotherapy , Mitochondrial Proteins/administration & dosage , Peptides/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Apoptosis/radiation effects , Apoptosis Regulatory Proteins , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/physiopathology , Mitochondrial Proteins/metabolism , Peptides/metabolism , Radiation-Sensitizing Agents/metabolism
17.
Sci Rep ; 6: 26510, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27211848

ABSTRACT

Aberrant activation of the NOTCH signaling pathway is crucial for the onset and progression of T cell leukemia. Yet recent studies also suggest a tumor suppressive role of NOTCH signaling in acute myeloid leukemia (AML) and reactivation of this pathway offers an attractive opportunity for anti-AML therapies. N-methylhemeanthidine chloride (NMHC) is a novel Amaryllidaceae alkaloid that we previously isolated from Zephyranthes candida, exhibiting inhibitory activities in a variety of cancer cells, particularly those from AML. Here, we report NMHC not only selectively inhibits AML cell proliferation in vitro but also hampers tumor development in a human AML xenograft model. Genome-wide gene expression profiling reveals that NMHC activates the NOTCH signaling. Combination of NMHC and recombinant human NOTCH ligand DLL4 achieves a remarkable synergistic effect on NOTCH activation. Moreover, pre-inhibition of NOTCH by overexpression of dominant negative MAML alleviates NMHC-mediated cytotoxicity in AML. Further mechanistic analysis using structure-based molecular modeling as well as biochemical assays demonstrates that NMHC docks in the hydrophobic cavity within the NOTCH1 negative regulatory region (NRR), thus promoting NOTCH1 proteolytic cleavage. Our findings thus establish NMHC as a potential NOTCH agonist that holds great promises for future development as a novel agent beneficial to patients with AML.


Subject(s)
Amaryllidaceae Alkaloids/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Intracellular Signaling Peptides and Proteins/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Membrane Proteins/administration & dosage , Receptor, Notch1/genetics , Amaryllidaceae Alkaloids/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Female , HL-60 Cells , Humans , Intracellular Signaling Peptides and Proteins/pharmacology , K562 Cells , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/pharmacology , Mice , Molecular Docking Simulation , Receptor, Notch1/chemistry , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
18.
Cell Physiol Biochem ; 38(3): 1138-46, 2016.
Article in English | MEDLINE | ID: mdl-26964005

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is characterized with progressive memory loss and severe cognitive impairments, which affect everyday life and human health in the elderly. It is required that an effective and safe protective reagent against AD should be developed. It has been reported that humanin (HN) exerts neuroprotective effects against AD. In this study, we investigated the effect of a novel and more effective HN derivative, Colivelin (CLN) on AD. METHODS: PDAPP(V717I) transgenic AD model mice (derived from parental C57/BL6 mice) were used in our study as AD model. Morris water maze test was used to test the memory impairment of AD mice and the levels of Aß40 and Aß42 were determined by an Elisa assay. We used an Immunohistochemistry and Immunofluorescence staining method to check the GFAP and MAP2 positive cells, and TUNEL to assess the apoptotic cells. Western blot assay was used to check the expression and phosphorylation level of p38. RESULTS: We found that CLN improved the memory impairment induced by AD and reduced the deposit of Aß40 and Aß42. CLN also inhibited cell apoptosis and activation of caspase 3 in brain tissues of AD mice. Inflammation in AD mice was alleviated by CLN treatment, including the accumulation of GFAP positive cells and the inflammatory cytokines. With both structure of AGA-HNG and ANDF, CLN exhibited significantly stronger effects than synchronously administration of AGA-HNG and ADNF, suggesting CLN as a novel potential effective therapeutic reagent for AD patients. Finally, we found that CLN inhibited phosphorylation of p38 in AD mice and p38 inhibitor, SB203580 weakened the therapeutic effect of CLN. CONCLUSION: CLN effectively improved the memory dysfunction in PDAPP mice, and our data suggests CLN as a novel and effective reagent which may have great potentials in AD therapy.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Intracellular Signaling Peptides and Proteins/administration & dosage , Memory/drug effects , Neuroprotective Agents/administration & dosage , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Animals , Apoptosis/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Intracellular Signaling Peptides and Proteins/pharmacology , Memory Disorders , Mice , Mice, Transgenic , Neuroprotective Agents/pharmacology , Phosphorylation , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Biochem J ; 473(1): 99-107, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26508734

ABSTRACT

Cancer cells are capable of sphere formation (transformation) through reactive oxygen species (ROS) and glycolysis shift. Transformation is linked to tumorigenesis and therapy resistance, hence targeting regulators of ROS and glycolysis is important for cancer therapeutic candidates. Here, we demonstrate that Smac mimetic AZ58 in combination with tumour necrosis factor-α (TNF-α) was able to inhibit the production of ROS, inhibit glycolysis through Pim-1 kinase-mediated Ser-112 phosphorylation of BAD, and increase depolarization of mitochondria. We also identified mitochondrial isoforms of Pim-1 kinase that were targeted for degradation by AZ58 in combination with TNF-α or AZ58 in combination with Fas ligand (FasL) plus cycloheximide (CHX) through caspase-3 to block transformation. Our study demonstrates that Smac mimetic in combination with TNF-α is an ideal candidate to target Pim-1 expression, inhibit ROS production and to block transformation from blebbishields.


Subject(s)
Biomimetic Materials/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Reactive Oxygen Species/metabolism , Transformation, Genetic/physiology , Tumor Necrosis Factor-alpha/metabolism , Apoptosis Regulatory Proteins , Biomimetic Materials/administration & dosage , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/administration & dosage , Mitochondrial Proteins/administration & dosage , Protein Isoforms/metabolism , Transformation, Genetic/drug effects , Tumor Necrosis Factor-alpha/administration & dosage
20.
Sci Rep ; 5: 14923, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26464223

ABSTRACT

Following root avulsion, spinal nerves are physically disconnected from the spinal cord. Severe motoneuron death and inefficient axon regeneration often result in devastating motor dysfunction. Newly formed axons need to extend through inhibitory scar tissue at the CNS-PNS transitional zone before entering into a pro-regenerative peripheral nerve trajectory. CSPGs are dominant suppressors in scar tissue and exert inhibition via neuronal receptors including PTPσ. Previously, a small peptide memetic of the PTPσ wedge region named ISP (Intracellular Sigma Peptide) was generated, and its capabilities to target PTPσ and relieve CSPG inhibition were validated. Here, we demonstrate that after ventral root avulsion and immediate re-implantation, modulation of PTPσ by systemic delivery of ISP remarkably enhanced regeneration. ISP treatment reduced motoneuron death, increased the number of axons regenerating across scar tissue, rebuilt healthy neuromuscular junctions and enhanced motor functional recovery. Our study shows that modulation of PTPσ is a potential therapeutic strategy for root avulsion.


Subject(s)
Intracellular Signaling Peptides and Proteins/administration & dosage , Radiculopathy/physiopathology , Radiculopathy/therapy , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Recovery of Function/physiology , Spinal Cord Regeneration/physiology , Animals , Central Nervous System Agents/administration & dosage , Dose-Response Relationship, Drug , Female , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord Regeneration/drug effects , Spinal Nerve Roots/drug effects , Spinal Nerve Roots/physiopathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...