Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Physiol ; 14: 1165379, 2023.
Article in English | MEDLINE | ID: mdl-37324380

ABSTRACT

Proper vascular formation is regulated by multiple signaling pathways. The vascular endothelial growth factor (VEGF) signaling promotes endothelial proliferation. Notch and its downstream targets act to lead endothelial cells toward an arterial fate through regulation of arterial gene expression. However, the mechanisms of how endothelial cells (ECs) in the artery maintain their arterial characteristics remain unclear. Here, we show that PRDM16 (positive regulatory domain-containing protein 16), a zinc finger transcription factor, is expressed in arterial ECs, but not venous ECs in developing embryos and neonatal retinas. Endothelial-specific deletion of Prdm16 induced ectopic venous marker expression in the arterial ECs and reduced vascular smooth muscle cell (vSMC) recruitment around arteries. Whole-genome transcriptome analysis using isolated brain ECs show that the expression of Angpt2 (encoding ANGIOPOIETIN2, which inhibits vSMC recruitment) is upregulated in the Prdm16 knockout ECs. Conversely, forced expression of PRDM16 in venous ECs is sufficient to induce arterial gene expression and repress the ANGPT2 level. Together, these results reveal an arterial cell-autonomous function for PRDM16 in suppressing venous characteristics in arterial ECs.

2.
J Neuroinflammation ; 19(1): 191, 2022 Jul 20.
Article in English | MEDLINE | ID: mdl-35858866

ABSTRACT

Sialic acid immunoglobulin-like lectin E (Siglec-E) is a subtype of pattern recognition receptors found on the surface of myeloid cells and functions as a key immunosuppressive checkpoint molecule. The engagement between Siglec-E and the ligand α2,8-linked disialyl glycans activates the immunoreceptor tyrosine-based inhibitory motif (ITIM) in its intracellular domain, mitigating the potential risk of autoimmunity amid innate immune attacks on parasites, bacteria, and carcinoma. Recent studies suggest that Siglec-E is also expressed in the CNS, particularly microglia, the brain-resident immune cells. However, the functions of Siglec-E in brain inflammation and injuries under many neurological conditions largely remain elusive. In this study, we first revealed an anti-inflammatory role for Siglec-E in lipopolysaccharide (LPS)-triggered microglial activation. We then found that Siglec-E was induced within the brain by systemic treatment with LPS in mice in a dose-dependent manner, while its ablation exacerbated hippocampal reactive microgliosis in LPS-treated animals. The genetic deficiency of Siglec-E also aggravated oxygen-glucose deprivation (OGD)-induced neuronal death in mouse primary cortical cultures containing both neurons and glial cells. Moreover, Siglec-E expression in ipsilateral brain tissues was substantially induced following middle cerebral artery occlusion (MCAO). Lastly, the neurological deficits and brain infarcts were augmented in Siglec-E knockout mice after moderate MCAO when compared to wild-type animals. Collectively, our findings suggest that the endogenous inducible Siglec-E plays crucial anti-inflammatory and neuroprotective roles following ischemic stroke, and thus might underlie an intrinsic mechanism of resolution of inflammation and self-repair in the brain.


Subject(s)
Encephalitis , Sialic Acid Binding Immunoglobulin-like Lectins , Animals , Encephalitis/pathology , Infarction, Middle Cerebral Artery/pathology , Lipopolysaccharides/pharmacology , Mice , Microglia/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
3.
Cell Rep ; 39(12): 111000, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35732130

ABSTRACT

Prostaglandin E2 (PGE2) promotes tumor cell proliferation, migration, and invasion, fostering an inflammation-enriched microenvironment that facilitates angiogenesis and immune evasion. However, the PGE2 receptor subtype (EP1-EP4) involved in neuroblastoma (NB) growth remains elusive. Herein, we show that the EP2 receptor highly correlates with NB aggressiveness and acts as a predominant Gαs-coupled receptor mediating PGE2-initiated cyclic AMP (cAMP) signaling in NB cells with high-risk factors, including 11q deletion and MYCN amplification. Knockout of EP2 in NB cells blocks the development of xenografts, and its conditional knockdown prevents established tumors from progressing. Pharmacological inhibition of EP2 by our recently developed antagonist TG6-129 suppresses the growth of NB xenografts in nude mice and syngeneic allografts in immunocompetent hosts, accompanied by anti-inflammatory, antiangiogenic, and apoptotic effects. This proof-of-concept study suggests that the PGE2/EP2 signaling pathway contributes to NB malignancy and that EP2 inhibition by our drug-like compounds provides a promising strategy to treat this deadly pediatric cancer.


Subject(s)
Neuroblastoma , Receptors, Prostaglandin E, EP2 Subtype , Animals , Dinoprostone/metabolism , Humans , Mice , Mice, Knockout , Mice, Nude , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Tumor Microenvironment
4.
Mol Neurobiol ; 59(7): 4593-4611, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35581521

ABSTRACT

Pathological changes in the brain endothelium play an important role in the progression of ischemic stroke and the compromised BBB under ischemic stroke conditions cause neuronal damage. However, the pathophysiological mechanisms of the BBB under normal conditions and under ischemic stroke conditions have not been fully elucidated. The present study demonstrated that knockdown of TAR DNA-binding protein 43 (TDP-43) or overexpression of TDP43-CTFs35 inhibited tight junction protein expression, and mammalian sterile-20-like 1/2 (MST1/2) and YES-associated protein (YAP) phosphorylation in brain ECs and suppressed brain EC migration in vitro. The cytoplasmic TDP43-CTFs35 level was increased in brain ECs 24 h and 72 h after MCAO, but it disappeared 1 week after cerebral ischemia. The expression of tight junction proteins was also significantly deceased 24 h after MCAO and then gradually recovered at 72 h and 1 week after MCAO. The level of YAP phosphorylation was first significantly decreased 24 h after MCAO and then increased 72 h and 1 week after MCAO, accompanied by nuclear YAP translocation. The underlying mechanism is TDP43-CTFs35-mediated inhibition of Hippo signaling pathway activity through the dephosphorylation of MST1/2, which leads to the inhibition of YAP phosphorylation and the subsequent impairment of brain EC migration and tight junction protein expression. This study provides new insights into the mechanisms of brain vascular EC regulation, which may impact on BBB integrity after cerebral ischemic injury.


Subject(s)
Brain Injuries , Brain Ischemia , Ischemic Stroke , Animals , Blood-Brain Barrier/pathology , Brain/metabolism , Brain Injuries/pathology , Brain Ischemia/pathology , DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Infarction, Middle Cerebral Artery/pathology , Mammals/metabolism , Tight Junction Proteins/metabolism
5.
Bioorg Med Chem ; 58: 116645, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35151118

ABSTRACT

The nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome plays an important role in microglia-mediated inflammation. Dysregulation of NLRP3 signaling results in microglial activation and triggers inflammatory responses contributing to the development of neurological disorders including ischemic stroke, schizophrenia, Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Inhibition of the NLRP3-linked inflammatory pathways reduces microglia-induced inflammation and is considered as a promising therapeutic approach for neuro-inflammatory diseases. In the present study, we report the development of AMS-17, a rationally-designed tertiary sulfonylurea compound for inhibition of inflammation in microglia. AMS-17 inhibited expression of the NLRP3, and its downstream components and cytokines such as caspase-1, tumor necrosis factor-α (TNF-α), IL-1ß and inducible nitric oxide synthase (iNOS). It also suppressed lipopolysaccharide (LPS)-induced N9 microglial cell phagocytosis in vitro and activation of the microglia in mouse brain in vivo. Together, these results provide promising evidences for the inhibitory effects of AMS-17 in inflammation. This proof-of-concept study provides a new chemical scaffold, designed with the aid of pharmacophore modeling, with NLRP3 inhibitory activity which can be further developed for the treatment of inflammation-associated neurological disorders.


Subject(s)
Inflammation/drug therapy , Microglia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfonylurea Compounds/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Inflammation/metabolism , Mice , Microglia/metabolism , Models, Molecular , Molecular Structure , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Sulfonylurea Compounds/chemical synthesis , Sulfonylurea Compounds/chemistry
6.
Neurotherapeutics ; 19(1): 366-385, 2022 01.
Article in English | MEDLINE | ID: mdl-35099767

ABSTRACT

As the inducible terminal enzyme for prostaglandin E2 (PGE2) synthesis, microsomal PGE synthase-1 (mPGES-1) contributes to neuroinflammation and secondary brain injury after cerebral ischemia via producing excessive PGE2. However, a proof of concept that mPGES-1 is a therapeutic target for ischemic stroke has not been established by a pharmacological strategy mainly due to the lack of drug-like mPGES-1 inhibitors that can be used in relevant rodent models. To this end, we recently developed a series of novel small-molecule compounds that can inhibit both human and rodent mPGES-1. In this study, blockade of mPGES-1 by our several novel compounds abolished the lipopolysaccharide (LPS)-induced PGE2 and pro-inflammatory cytokines interleukin 1ß (IL-1ß), IL-6, and tumor necrosis factor α (TNF-α) in mouse primary brain microglia. Inhibition of mPGES-1 also decreased PGE2 produced by neuronal cells under oxygen-glucose deprivation (OGD) stress. Among the five enzymes for PGE2 biosynthesis, mPGES-1 was the most induced one in cerebral ischemic lesions. Systemic treatment with our lead compound MPO-0063 (5 or 10 mg/kg, i.p.) in mice after transient middle cerebral artery occlusion (MCAO) improved post-stroke well-being, decreased infarction and edema, suppressed induction of brain cytokines (IL-1ß, IL-6, and TNF-α), alleviated locomotor dysfunction and anxiety-like behavior, and reduced the long-term cognitive impairments. The therapeutic effects of MPO-0063 in this proof-of-concept study provide the first pharmacological evidence that mPGES-1 represents a feasible target for delayed, adjunct treatment - along with reperfusion therapies - for acute brain ischemia.


Subject(s)
Brain Ischemia , Ischemic Stroke , Nervous System Diseases , Animals , Brain Ischemia/drug therapy , Cytokines , Dinoprostone , Interleukin-6 , Mice , Prostaglandin-E Synthases , Tumor Necrosis Factor-alpha
7.
Brain Res Bull ; 179: 74-82, 2022 02.
Article in English | MEDLINE | ID: mdl-34942325

ABSTRACT

As major immune responsive cells in the central nervous system (CNS), activated microglia can present pro-inflammatory M1 phenotype aggravating the neuronal injury or anti-inflammatory M2 phenotype providing neuroprotection and promoting neuronal survival in neurodegenerative diseases. In this study, we demonstrated that a compound, 4R-cembranoid (4R, 1S, 2E, 4R, 6R,-7E, 11E-2, 7, 11-cembratriene-4, 6-diol cembranoids) promoted M2 phenotype while attenuated M1 phenotype in N9 cells, a microglial cell line. Following Lipopolysaccharides (LPS) or Oxygen-glucose deprivation (OGD) treatment, the N9 cells treated by 1 µM 4R showed an increased Arginase-1 (Arg1, a M2 marker) expression and a reduced inducible nitric oxide synthase (iNOS, M1 marker) expression. In addition, the conditioned medium of 4R-treated post-OGD N9 cells protected neuro2a cells, a neuronal cell line, from OGD-induced injury. The viability of neuro2a cells in OGD condition was increased by 54.5% after treated with the conditioned medium of 4R-treated post-OGD N9 cells. Furthermore, we demonstrated the protective mechanism of 4R was associated with a decreased TNF-α release and an increased IL-10 release from N9 cells. In conclusion, our study demonstrated that the neuroprotective effects of 4R were through the regulation of microglial activation by promoting the protective M2 activation and inhibiting the damaging M1 activation. Therefore, the findings of this study suggest that 4R could be a promising lead structure for the development of drugs for the treatment of ischemic stroke and other neurodegenerative diseases with an inflammatory component involved.


Subject(s)
Cell Hypoxia/drug effects , Diterpenes/pharmacology , Microglia/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Cells, Cultured , Glucose/metabolism , Humans
8.
Front Aging Neurosci ; 13: 716917, 2021.
Article in English | MEDLINE | ID: mdl-34489683

ABSTRACT

Growing evidence suggests that hypertension and aging are prominent risk factors for the development of late-onset Alzheimer's disease (LOAD) by inducement of neuroinflammation. Recent study showed that neuroinflammation via activated microglia induces reactive astrocytes, termed A1 astrocytes, that highly upregulate numerous classical complement cascade genes that are destructive to neurons in neurodegeneration diseases. Moreover, triggering receptor expressed on myeloid cells 2 (TREM2) is considered as one of the strongest single-allele genetic risk factors and plays important roles in neuroinflammation for LOAD. However, the mechanisms of microglia in the regulation of A1 astrocytic activation are still not clear. We introduced angiotensin II-induced hypertension in middle-aged mice and found that hypertension-upregulated TREM2 expression and A1 astrocytic activation were involved in neuroinflammation in the animal models used in this study. The in vitro results revealed that overexpression of microglial TREM2 not only mitigated microglial inflammatory response but also had salutary effects on reverse A1 astrocytic activation and neuronal toxicity.

9.
ACS Pharmacol Transl Sci ; 3(4): 635-643, 2020 Aug 14.
Article in English | MEDLINE | ID: mdl-32832866

ABSTRACT

Prostaglandin E2 (PGE2) is elevated in the brain by excitotoxic insults and, in turn, aggravates the neurotoxicity mainly through acting on its Gαs-coupled receptor EP2, inspiring a therapeutic strategy of targeting this key proinflammatory pathway. Herein, we investigated the effects of several highly potent and selective small-molecule antagonists of the EP2 receptor on neuronal excitotoxicity both in vitro and in vivo. EP2 inhibition by these novel compounds largely decreased the neuronal injury in rat primary hippocampal cultures containing both neurons and glia that were treated with N-methyl-d-aspartate and glycine. Using a bioavailable and brain-permeant analogue TG6-10-1 that we recently developed to target the central EP2 receptor, we found that the poststroke EP2 inhibition in mice decreased the neurological deficits and infarct volumes as well as downregulated the prototypic inflammatory cytokines in the brain after a transient ischemia. Our preclinical findings together reinforced the notion that targeting the EP2 receptor represents an emerging therapeutic strategy to prevent the neuronal injury and inflammation following ischemic stroke.

10.
Proc Natl Acad Sci U S A ; 114(41): 10918-10923, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973878

ABSTRACT

Angiogenesis and vascular remodeling are essential for the establishment of vascular networks during organogenesis. Here we show that the Hippo signaling pathway effectors YAP and TAZ are required, in a gene dosage-dependent manner, for the proliferation and migration of vascular endothelial cells (ECs) during retinal angiogenesis. Intriguingly, nuclear translocation of YAP and TAZ induced by Lats1/2-deletion blocked endothelial migration and phenocopied Yap/Taz-deficient mutants. Furthermore, overexpression of a cytoplasmic form of YAP (YAPS127D) partially rescued the migration defects caused by loss of YAP and TAZ function. Finally, we found that cytoplasmic YAP positively regulated the activity of the small GTPase CDC42, deletion of which caused severe defects in endothelial migration. These findings uncover a previously unrecognized role of cytoplasmic YAP/TAZ in promoting cell migration by activating CDC42 and provide insight into how Hippo signaling in ECs regulates angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Movement , Endothelium, Vascular/cytology , Neovascularization, Physiologic , Phosphoproteins/physiology , Transcription Factors/physiology , cdc42 GTP-Binding Protein/physiology , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Proliferation , Endothelium, Vascular/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
11.
Sci Rep ; 7(1): 9459, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28842681

ABSTRACT

Cyclooxygenase-2 (COX-2) triggers pro-inflammatory processes that can aggravate neuronal degeneration and functional impairments in many neurological conditions, mainly via producing prostaglandin E2 (PGE2) that activates four membrane receptors, EP1-EP4. However, which EP receptor is the culprit of COX-2/PGE2-mediated neuronal inflammation and degeneration remains largely unclear and presumably depends on the insult types and responding components. Herein, we demonstrated that COX-2 was induced and showed nuclear translocation in two neuronal cell lines - mouse Neuro-2a and human SH-SY5Y - after treatment with neurotoxin 6-hydroxydopamine (6-OHDA), leading to the biosynthesis of PGE2 and upregulation of pro-inflammatory cytokine interleukin-1ß. Inhibiting COX-2 or microsomal prostaglandin E synthase-1 suppressed the 6-OHDA-triggered PGE2 production in these cells. Treatment with PGE2 or EP2 selective agonist butaprost, but not EP4 agonist CAY10598, increased cAMP response in both cell lines. PGE2-initiated cAMP production in these cells was blocked by our recently developed novel selective EP2 antagonists - TG4-155 and TG6-10-1, but not by EP4 selective antagonist GW627368X. The 6-OHDA-promoted cytotoxicity was largely blocked by TG4-155, TG6-10-1 or COX-2 selective inhibitor celecoxib, but not by GW627368X. Our results suggest that PGE2 receptor EP2 is a key mediator of COX-2 activity-initiated cAMP signaling in Neuro-2a and SH-SY5Y cells following 6-OHDA treatment, and contributes to oxidopamine-mediated neurotoxicity.


Subject(s)
Cyclooxygenase 2/metabolism , Neurons/physiology , Oxidopamine/metabolism , Parkinson Disease/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Cytokines/metabolism , Humans , Inflammation , Inflammation Mediators/metabolism , Mice , Parkinson Disease/immunology , Signal Transduction , Up-Regulation
12.
Front Neurosci ; 11: 272, 2017.
Article in English | MEDLINE | ID: mdl-28611572

ABSTRACT

(1S, 2E, 4R, 6R,-7E, 11E)-2, 7, 11-cembratriene-4, 6-diol (4R) is one of the cembranoids found in tobacco leaves. Previous studies have found that 4R protected acute rat hippocampal slices against neurotoxicity induced by N-methyl-D-aspartate (NMDA) and against the toxic organophosphorus compounds paraoxon and diisopropylfluorophosphate (DFP). Furthermore, in vivo, 4R reduced the infarct size in a rodent ischemic stroke model and neurodegeneration caused by DFP. The present study expanded our previous study by focusing on the effect of 4R in Parkinson's disease (PD) and elucidating its underlying mechanisms using 6-hydroxydopamine (6-OHDA)-induced injury models. We found that 4R exhibited significant neuroprotective activity in the rat unilateral 6-OHDA-induced PD model in vivo. The therapeutic effect was evident both at morphological and behavioral levels. 4R (6 and 12 mg/kg) treatments significantly improved outcomes of 6-OHDA-induced PD in vivo as indicated by reducing forelimb asymmetry scores and corner test scores 4 weeks after injection of 6-OHDA (p < 0.05). The therapeutic effect of 4R was also reflected by decreased depletion of tyrosine hydroxylase (TH) in the striatum and substantia nigra (SN) on the side injected with 6-OHDA. TH expression was 70.3 and 62.8% of the contralateral side in striatum and SN, respectively, after 6 mg/kg 4R treatment; furthermore, it was 80.1 and 79.3% after treatment with 12 mg/kg of 4R. In the control group, it was 51.9 and 23.6% of the contralateral striatum and SN (p < 0.05). Moreover, 4R also protected differentiated neuro-2a cells from 6-OHDA-induced cytotoxicity in vitro. The activation of p-AKT and HAX-1, and inhibition of caspase-3 and endothelial inflammation, were involved in 4R-mediated protection against 6-OHDA-induced injury. In conclusion, the present study indicates that 4R shows a therapeutic effect in the rat 6-OHDA-induced PD model in vivo and in 6-OHDA-challenged neuro-2a cells in vitro.

13.
Neuroscience ; 329: 30-42, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27132231

ABSTRACT

The present study generated a novel DNA complex to specifically target endothelial NF-κB to inhibit cerebral vascular inflammation. This DNA complex (GS24-NFκB) contains a DNA decoy which inhibits NF-κB activity, and a DNA aptamer (GS-24), a ligand of transferrin receptor (TfR), which allows for targeted delivery of the DNA decoy into cells. The results indicate that GS24-NFκB was successfully delivered into a murine brain-derived endothelial cell line, bEND5, and inhibited inflammatory responses induced by tumor necrosis factor α (TNF-α) or oxygen-glucose deprivation/re-oxygenation (OGD/R) via down-regulation of the nuclear NF-κB subunit, p65, as well as its downstream inflammatory cytokines, inter-cellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule (VCAM-1). The inhibitory effect of the GS24-NFκB was demonstrated by a significant reduction in TNF-α or OGD/R induced monocyte adhesion to the bEND5 cells after GS24-NFκB treatment. Intravenous (i.v.) injection of GS24-'NFκB (15mg/kg) was able to inhibit the levels of phoseph-p65 and VCAM-1 in brain endothelial cells in a mouse lipopolysaccharide (LPS)-induced inflammatory model in vivo. In conclusion, our approach using DNA nanotechnology for DNA decoy delivery could potentially be utilized for inhibition of inflammation in ischemic stroke and other neuro-inflammatory diseases affecting cerebral vasculature.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aptamers, Nucleotide/pharmacology , Brain/drug effects , Endothelial Cells/drug effects , Inflammation/drug therapy , Oligodeoxyribonucleotides/pharmacology , Vasculitis, Central Nervous System/drug therapy , Animals , Anti-Inflammatory Agents/blood , Anti-Inflammatory Agents/pharmacokinetics , Aptamers, Nucleotide/blood , Aptamers, Nucleotide/pharmacokinetics , Brain/blood supply , Brain/immunology , Cell Hypoxia/drug effects , Cell Line , Disease Models, Animal , Drug Design , Drug Evaluation, Preclinical , Endothelial Cells/immunology , Glucose/deficiency , Goats , Inflammation/metabolism , Lipopolysaccharides , Male , Mice , Neuroprotective Agents/blood , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Oligodeoxyribonucleotides/blood , Oligodeoxyribonucleotides/pharmacokinetics , Tumor Necrosis Factor-alpha , Vasculitis, Central Nervous System/metabolism
14.
Biochim Biophys Acta ; 1852(11): 2362-71, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26300484

ABSTRACT

Sepsis is an infection-induced severe inflammatory disorder that leads to multiple organ failure. Amongst organs affected, myocardial depression is believed to be a major contributor to septic death. While it has been identified that large amounts of circulating pro-inflammatory cytokines are culprit for triggering cardiac dysfunction in sepsis, the underlying mechanisms remain obscure. Additionally, recent studies have shown that exosomes released from bacteria-infected macrophages are pro-inflammatory. Hence, we examined in this study whether blocking the generation of exosomes would be protective against sepsis-induced inflammatory response and cardiac dysfunction. To this end, we pre-treated RAW264.7 macrophages with GW4869, an inhibitor of exosome biogenesis/release, followed by endotoxin (LPS) challenge. In vivo, we injected wild-type (WT) mice with GW4869 for 1h prior to endotoxin treatment or cecal ligation/puncture (CLP) surgery. We observed that pre-treatment with GW4869 significantly impaired release of both exosomes and pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6) in RAW264.7 macrophages. At 12h after LPS treatment or CLP surgery, WT mice pre-treated with GW4869 displayed lower amounts of exosomes and pro-inflammatory cytokines in the serum than control PBS-injected mice. Accordingly, GW4869 treatment diminished the sepsis-induced cardiac inflammation, attenuated myocardial depression and prolonged survival. Together, our findings indicate that blockade of exosome generation in sepsis dampens the sepsis-triggered inflammatory response and thereby, improves cardiac function and survival.

15.
Neuroscience ; 291: 250-259, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25677097

ABSTRACT

(1S,2E,4R,6R,-7E,11E)-2,7,11-cembratriene-4,6-diol (4R) is a precursor to key flavor ingredients in leaves of Nicotiana species. The present study shows 4R decreased brain damage in rodent ischemic stroke models. The 4R-pretreated mice had lower infarct volumes (26.2±9.7 mm3) than those in control groups (untreated: 63.4±4.2 mm3, DMSO: 60.2±14.2 mm3). The 4R-posttreated rats also had less infarct volumes (120±65 mm3) than those in the rats of the DMSO group (291±95 mm3). The results from in vitro experiments indicate that 4R decreased neuro2a cell (neuroblastoma cells) apoptosis induced by oxygen-glucose deprivation (OGD), and improved the population spikes' (PSs) recovery in rat acute hippocampal slices under OGD; a phosphatidylinositol 3-kinase (PI3K) inhibitor, wortmannin, abolished the effect of 4R on PSs recovery. Furthermore, 4R also inhibited monocyte adhesion to murine brain-derived endothelial (bEND5) cells and upregulation of intercellular adhesion molecule-1(ICAM-1) induced by OGD/reoxygenation (OGD/R), and restored the p-Akt level to pre-OGD/R values in bEND5 cells. In conclusion, the present study indicates that 4R has a protective effect in rodent ischemic stroke models. Inhibition of ICAM-1 expression and restoration of Akt phosphorylation are the possible mechanisms involved in cellular protection by 4R.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Diterpenes/pharmacology , Neuroprotective Agents/pharmacology , Stroke/drug therapy , Animals , Brain/pathology , Brain/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line , Cell Line, Tumor , Disease Models, Animal , Female , Glucose/deficiency , Male , Mice , Rats , Rats, Sprague-Dawley , Stroke/pathology , Stroke/physiopathology
16.
Mol Ther Nucleic Acids ; 3: e209, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25368913

ABSTRACT

Because adhesion of leukocytes to endothelial cells is the first step of vascular-neuronal inflammation, inhibition of adhesion and recruitment of leukocytes to vascular endothelial cells will have a beneficial effect on neuroinflammatory diseases. In this study, we used the pRNA of bacteriophage phi29 DNA packaging motor to construct a novel RNA nanoparticle for specific targeting to transferrin receptor (TfR) on the murine brain-derived endothelial cells (bEND5) to deliver ICAM-1 siRNA. This RNA nanoparticle (FRS-NPs) contained a FB4 aptamer targeting to TfR and a siRNA moiety for silencing the intercellular adhesion molecule-1 (ICAM-1). Our data indicated that this RNA nanoparticle was delivered into murine brain-derived endothelial cells. Furthermore, the siRNA was released from the FRS-NPs in the cells and knocked down ICAM-1 expression in the TNF-α-stimulated cells and in the cells under oxygen-glucose deprivation/reoxygenation (OGD/R) condition. The functional end points of the study indicated that FRS-NPs significantly inhibited monocyte adhesion to the bEND5 cells induced by TNF-α and OGD/R. In conclusion, our approach using RNA nanotechnology for siRNA delivery could be potentially applied for inhibition of inflammation in ischemic stroke and other neuroinflammatory diseases, or diseases affecting endothelium of vasculature.

17.
J Pharm Pharm Sci ; 16(4): 541-50, 2013.
Article in English | MEDLINE | ID: mdl-24210062

ABSTRACT

PURPOSE: Drug delivery by transferrin receptor-mediated transport at the blood-brain barrier has shown beneficial effects in animal models of stroke, but it is unclear whether receptor mediated uptake remains functional in the ischemic tissue. The present study addressed that question in a mouse model of brain focal ischemia, permanent or transient middle cerebral artery occlusion (MCAO). METHODS: Brain accumulation of 125I-labeled 8D3, a mouse-specific transferrin receptor antibody, or of the isotype control UPC-10 used as vascular marker, was measured autoradiographically by phosphorimaging in the core ischemic region on cryostat brain sections up to 24h after ischemia or reperfusion. Cerebral blood flow was quantitatively determined in the same animals after administration of 99mTc-ECD (Neurolite). RESULTS: Apparent volume of distribution obtained with UPC-10 indicated no significant nonspecific leakage of the blood-brain barrier at any time point. Although brain uptake of 8D3 gradually declined compared to healthy tissue under MCAO, VD remained significantly higher than VD of UPC-10 up to 5h. In transient MCAO the brain uptake recovered to levels as in healthy tissue immediately after reperfusion. CONCLUSION: Transferrin receptor-mediated brain uptake, which is an energy dependent vesicular transport process, is sensitive to reduction in blood supply but remains partially functional for several hours after onset of ischemia. The uptake shows complete recovery after reperfusion. These results support the use of transferrin receptor-mediated brain drug delivery in the early phase of ischemia and in the phase when blood flow is restored. This article is open to POST-PUBLICATION REVIEW. Registered readers (see "For Readers") may comment by clicking on ABSTRACT on the issue's contents page.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Drug Delivery Systems , Receptors, Transferrin/metabolism , Animals , Brain/blood supply , Brain Ischemia/drug therapy , Cerebrovascular Circulation , Female , Infarction, Middle Cerebral Artery , Mice , Reperfusion
18.
J Microencapsul ; 30(7): 701-8, 2013.
Article in English | MEDLINE | ID: mdl-23517066

ABSTRACT

Novel aptamer-functionalized polyethylene glycol-polylactic acid (PEG-PLA) (APP) micelles were developed with the objective to target the transferrin receptor on brain endothelial cells. Flurbiprofen, a potential drug for therapeutic management of Alzheimer's disease (AD), was loaded into the APP micelles using the co-solvent evaporation method. Results indicated that 9.03% (w/w) of flurbiprofen was entrapped in APP with good retention capacity in vitro. Targeting potential of APPs was investigated using the transferring receptor-expressing murine brain endothelial bEND5 cell line. APPs significantly enhanced surface association of micelles to bEND5 cells as quantified by fluorescence spectroscopy. Most importantly, APPs significantly enhanced intracellular flurbiprofen delivery when compared to unmodified micelles. These results suggest that APP micelles may offer an effective strategy to deliver therapeutically effective flurbiprofen concentrations into the brain for AD patients.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aptamers, Nucleotide/chemistry , Brain/metabolism , Drug Delivery Systems , Flurbiprofen/administration & dosage , Micelles , Polyethylene Glycols/chemistry , Alzheimer Disease/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Aptamers, Nucleotide/metabolism , Base Sequence , Brain/cytology , Cell Line , Drug Carriers/chemistry , Drug Carriers/metabolism , Endothelial Cells/metabolism , Flurbiprofen/pharmacokinetics , Mice , Polyethylene Glycols/metabolism , Receptors, Transferrin/metabolism
19.
Neurosci Lett ; 534: 338-43, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23262083

ABSTRACT

Hax-1, a multi-functional protein, recently was found to be involved in apoptosis and nerve system development. The purpose of this study was to detect the effect of cerebral ischemia on Hax-1 expression. We have detected the expression of Hax-1 in normal brain tissue and in ischemic brain tissue. Hax-1 was expressed in all brain regions detected with a level similar to the level of ß-actin. There were no differences in the expression of Hax-1 in different brain regions detected. The confocal images confirmed that neurons expressed Hax-1. The results of ischemic stroke in vivo indicated that Hax-1 level was significantly reduced at 24h after ischemia in the ischemic hemisphere, which was only 37%±4.8 of healthy hemisphere (p<0.05), and there was a strong reverse correlation between the level of Hax-1 and infarct size indicated by the regress analysis (R(2)=0.84). The expression of Hax-1 was also reduced in the cells subjected to oxygen/glucose deprivation (OGD) (p<0.01). The expression of Hax-1 was 87%±4.6, 78%±4.9 and 54%±8.2 of control in the murine brain endothelial cell (bEND5 cell) at 1h, 2h and 16h OGD, respectively. The Hax-1 level was 82%±7.3 and 61%±8.1 of control in neuronal cell line (neuro-2a cells) at 5h and 12h OGD, respectively. The percentage of neuro-2a cell death was 40%±11 induced by a 5h of OGD compared to only 10%±4.2 cell death in the control group (p<0.01). Our present study provides preliminary evidence of the effect of cerebral ischemia on Hax-1 expression. The expression of Hax-1 in normal brain tissue and reduction of Hax-1 in ischemic brain tissue indicate its possible involvement in pathophysiological functions in the brain.


Subject(s)
Brain/metabolism , Ischemic Attack, Transient/metabolism , Proteins/metabolism , Animals , Brain/blood supply , Brain/pathology , Cell Death , Cell Hypoxia , Cell Line , Endothelial Cells/metabolism , Glucose/deficiency , Intracellular Signaling Peptides and Proteins , Ischemic Attack, Transient/pathology , Mice , Neurons/pathology
20.
AAPS PharmSciTech ; 13(4): 1309-18, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23054983

ABSTRACT

This study was performed to test the feasibility of chitosan and polylactic-co-glycolic acid (PLGA) incorporated nanoparticles as sustained-release carriers for the delivery of negatively charged low molecular weight heparin (LMWH). Fourier transform infrared (FTIR) spectrometry was used to evaluate the interactions between chitosan and LMWH. The shifts, intensity, and broadening of the characteristic peaks for the functional groups in the FTIR spectra indicated that strong interactions occur between the positively charged chitosans and the negatively charged LMWHs. Three types of LMWH nanoparticles (NP-1, NP-2, and NP-3) were prepared using chitosan with or without PLGA: NP-1 nanoparticles were formed by polyelectrolyte complexation after single mixing, NP-2 nanoparticles were prepared by polyelectrolyte complexation after single emulsion-diffusion-evaporation, and NP-3 nanoparticles were optimized by double emulsion-diffusion-evaporation. NP-3 nanoparticles of LMWH prepared by the emulsion-diffusion-evaporation method showed significant differences in particle morphology, size, zeta potential, and drug release profile compared to NP-1 nanoparticles formed by polyelectrolyte complexation. Another ionic complex of LMWH with chitosan-incorporated PLGA nanoparticles (NP-2) showed lower drug entrapment efficiency than that of NP-1 and NP-3. The drug release rate of NP-3 was slower than the release rates of NP-1 and NP-2, although particle morphology of NP-3 was similar to that of NP-2. Cell viability was not adversely affected when cells were treated with all three types of nanoparticles. The data presented in this study demonstrate that nanoparticles formulated with chitosan-PLGA could be a safe sustained-release carrier for the delivery of LMWH.


Subject(s)
Chitosan/chemistry , Heparin, Low-Molecular-Weight/chemistry , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Cell Line , Cell Survival/drug effects , Delayed-Action Preparations/chemistry , Drug Carriers/chemistry , Drug Delivery Systems/methods , Emulsions/chemistry , Epithelial Cells/drug effects , Feasibility Studies , Humans , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer
SELECTION OF CITATIONS
SEARCH DETAIL
...