Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
bioRxiv ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38979231

ABSTRACT

Traumatic brain injury (TBI) is a significant public health concern characterized by a complex cascade of cellular events. TBI induces adenosine monophosphate-activated protein kinase (AMPK) dysfunction impairs energy balance activates inflammatory cytokines and leads to neuronal damage. AMPK is a key regulator of cellular energy homeostasis during inflammatory responses. Recent research has revealed its key role in modulating the inflammatory process in TBI. Following TBI the activation of AMPK can influence various important pathways and mechanisms including metabolic pathways and inflammatory signaling. Our study investigated the effects of post-TBI loss of AMPK function on functional outcomes inflammasome activation, and inflammatory cytokine production. Male C57BL/6 adult wild-type (WT) and AMPK knockout (AMPK-KO) mice were subjected to a controlled cortical impact (CCI) model of TBI or sham surgery. The mice were tested for behavioral impairment at 24 h post-TBI thereafter, mice were anesthetized, and their brains were quickly removed for histological and biochemical evaluation. In vitro we investigated inflammasome activation in mixed glial cells stimulated with lipopolysaccharides+ Interferon-gamma (LI) (0.1 µg/20 ng/ml LPS/IFNg) for 6 h to induce an inflammatory response. Estimating the nucleotide-binding domain, leucine-rich-containing family pyrin domain containing western blotting ELISA and qRT-PCR performed 3 (NLRP3) inflammasome activation and cytokine production. Our findings suggest that TBI leads to reduced AMPK phosphorylation in WT mice and that the loss of AMPK correlates with worsened behavioral deficits at 24 h post-TBI in AMPK-KO mice as compared to WT mice. Moreover compared with the WT mice AMPK-KO mice exhibit exacerbated NLRP3 inflammasome activation and increased expression of proinflammatory mediators such as IL-1b IL-6 TNF-a iNOS and Cox 2. These results align with the in vitro studies using brain glial cells under inflammatory conditions, demonstrating greater activation of inflammasome components in AMPK-KO mice than in WT mice. Our results highlighted the critical role of AMPK in TBI outcomes. We found that the absence of AMPK worsens behavioral deficits and heightens inflammasome-mediated inflammation thereby exacerbating brain injury after TBI. Restoring AMPK activity after TBI could be a promising therapeutic approach for alleviating TBI-related damage.

2.
Antioxidants (Basel) ; 11(10)2022 Oct 18.
Article in English | MEDLINE | ID: mdl-36290774

ABSTRACT

The restoration of cerebral blood flow (CBF) to achieve brain tissue oxygenation (PbtO2) is the primary treatment for ischemic stroke, a significant cause of adult mortality and disability worldwide. Nitric oxide (NO) and its bioactive s-nitrosylated (SNO) reservoirs, such as s-nitrosoglutathione (GSNO), induce hypoxic vasodilation to enhance CBF during ischemia. The endogenous pool of SNOs/GSNO is enhanced via the activation of endothelial NO synthase (eNOS/NOS3) and by the suppression of class III alcohol dehydrogenase 5 (ADH5), also known as GSNO reductase (GSNOR). Remote ischemic conditioning (RIC), which augments NOS3 activity and SNO, is an emerging therapy in acute stroke. However, RIC has so far shown neutral effects in stroke clinical trials. As the majority of stroke patients are presented with endothelial dysfunctions and comorbidities, we tested the hypothesis that NOS3 dysfunction and diabetes will abolish the protective effects of RIC therapy in stroke, and the prior inhibition of GSNOR will turn RIC protective. Our data demonstrate that RIC during thrombotic stroke failed to enhance the CBF and the benefits of thrombolysis in NOS3 mutant (NOS3+/-) mice, a genetic model of NOS3 dysfunction. Interestingly, thrombotic stroke in diabetic mice enhanced the activity of GSNOR as early as 3 h post-stroke without decreasing the plasma nitrite (NO2-). In thrombotic stroke, neither a pharmacological inhibitor of GSNOR (GRI) nor RIC therapy alone was protective in diabetic mice. However, prior treatment with GRI followed by RIC enhanced the CBF and improved recovery. In a reperfused stroke model, the GRI-RIC combination therapy in diabetic mice augmented PbtO2, a translatory signature of successful microvascular reflow. In addition, RIC therapy unexpectedly increased the inflammatory markers at 6 h post-stroke in diabetic stroke that were downregulated in combination with GRI while improving the outcomes. Thus, we conclude that preexisting NOS3 dysfunctions due to comorbidities may neutralize the benefits of RIC in stroke, which can be turned protective in combination with GRI. Our findings may support the future clinical trial of RIC in comorbid stroke. Further studies are warranted to test and develop SNO reservoirs as the blood-associated biomarker to monitor the response and efficacy of RIC therapy in stroke.

3.
Proc Natl Acad Sci U S A ; 119(25): e2123265119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35700359

ABSTRACT

Metabolic aberrations impact the pathogenesis of multiple sclerosis (MS) and possibly can provide clues for new treatment strategies. Using untargeted metabolomics, we measured serum metabolites from 35 patients with relapsing-remitting multiple sclerosis (RRMS) and 14 healthy age-matched controls. Of 632 known metabolites detected, 60 were significantly altered in RRMS. Bioinformatics analysis identified an altered metabotype in patients with RRMS, represented by four changed metabolic pathways of glycerophospholipid, citrate cycle, sphingolipid, and pyruvate metabolism. Interestingly, the common upstream metabolic pathway feeding these four pathways is the glycolysis pathway. Real-time bioenergetic analysis of the patient-derived peripheral blood mononuclear cells showed enhanced glycolysis, supporting the altered metabolic state of immune cells. Experimental autoimmune encephalomyelitis mice treated with the glycolytic inhibitor 2-deoxy-D-glucose ameliorated the disease progression and inhibited the disease pathology significantly by promoting the antiinflammatory phenotype of monocytes/macrophage in the central nervous system. Our study provided a proof of principle for how a blood-based metabolomic approach using patient samples could lead to the identification of a therapeutic target for developing potential therapy.


Subject(s)
Drug Development , Glycolysis , Metabolomics , Multiple Sclerosis, Relapsing-Remitting , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antimetabolites/pharmacology , Antimetabolites/therapeutic use , Deoxyglucose/pharmacology , Deoxyglucose/therapeutic use , Drug Development/methods , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Glycolysis/drug effects , Humans , Leukocytes, Mononuclear/metabolism , Mice , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/metabolism
4.
Biomolecules ; 11(11)2021 10 27.
Article in English | MEDLINE | ID: mdl-34827584

ABSTRACT

The COVID-19 pandemic has escalated the occurrence of hypoxia including thrombotic stroke worldwide, for which nitric oxide (NO) therapy seems very promising and translatable. Therefore, various modes/routes of NO-delivery are now being tested in different clinical trials for safer, faster, and more effective interventions against ischemic insults. Intravenous (IV) infusion of S-Nitrosoglutathione (GSNO), the major endogenous molecular pool of NO, has been reported to protect against mechanical cerebral ischemia-reperfusion (IR); however, it has been never tested in any kind of "clinically" relevant thromboembolic stroke models with or without comorbidities and in combination with the thrombolytic reperfusion therapy. Moreover, "IV-effects" of higher dose of GSNO following IR-injury have been contradicted to augment stroke injury. Herein, we tested the hypothesis that nebulization of low-dose GSNO will not alter blood pressure (BP) and will mitigate stroke injury in diabetic mice via enhanced cerebral blood flow (CBF) and brain tissue oxygenation (PbtO2). GSNO-nebulization (200 µg/kgbwt) did not alter BP, but augmented the restoration of CBF, improved behavioral outcomes and reduced stroke injury. Moreover, GSNO-nebulization increased early reoxygenation of brain tissue/PbtO2 as measured at 6.5 h post-stroke following thrombolytic reperfusion, and enervated unwanted effects of late thrombolysis in diabetic stroke. We conclude that the GSNO-nebulization is safe and effective for enhancing collateral microvascular perfusion in the early hours following stroke. Hence, nebulized-GSNO therapy has the potential to be developed and translated into an affordable field therapy against ischemic events including strokes, particularly in developing countries with limited healthcare infrastructure.


Subject(s)
Diabetes Complications/drug therapy , Diabetes Mellitus/drug therapy , Hemorrhage/prevention & control , S-Nitrosoglutathione/administration & dosage , Stroke/complications , Thrombolytic Therapy/adverse effects , Animals , Behavior, Animal , Blood Pressure , Blood-Brain Barrier , COVID-19/epidemiology , Hemorrhage/complications , Hypoxia , Infusions, Intravenous , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred C57BL , Microcirculation , Nebulizers and Vaporizers , Neuroprotective Agents/pharmacology , Perfusion , Reperfusion Injury/drug therapy , Risk , Stress, Mechanical
5.
Cond Med ; 4(3): 124-129, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34414362

ABSTRACT

Remote ischemic conditioning (RIC) is a promising safe, feasible, and inexpensive treatment for acute stroke, both ischemic and hemorrhagic. It is applied with a blood pressure cuff on the limbs and is ideal for the prehospital setting. RIC is a form of preconditioning with similarities to physical exercise. Its mechanisms of action are multiple and include improvement of collateral cerebral blood flow (CBF) and RIC acts as a "collateral therapeutic". The increased CBF is likely related to nitric oxide synthase 3 in the endothelium and more importantly in circulating blood cells like the red blood cell. The RESIST clinical trial is a 1500 subject multicenter, randomized, sham-controlled trial of RIC in the prehospital setting in Denmark and should address the questions of whether RIC is safe and effective in acute stroke and whether the effect is mediated by an effect on nitric oxide/nitrite metabolism.

6.
JCI Insight ; 6(1)2021 01 11.
Article in English | MEDLINE | ID: mdl-33427206

ABSTRACT

The CNS is regarded as an immunoprivileged organ, evading routine immune surveillance; however, the coordinated development of immune responses profoundly influences outcomes after brain injury. Innate lymphoid cells (ILCs) are cytokine-producing cells that are critical for the initiation, modulation, and resolution of inflammation, but the functional relevance and mechanistic regulation of ILCs are unexplored after acute brain injury. We demonstrate increased proliferation of all ILC subtypes within the meninges for up to 1 year after experimental traumatic brain injury (TBI) while ILCs were present within resected dura and elevated within cerebrospinal fluid (CSF) of moderate-to-severe TBI patients. In line with energetic derangements after TBI, inhibition of the metabolic regulator, AMPK, increased meningeal ILC expansion, whereas AMPK activation suppressed proinflammatory ILC1/ILC3 and increased the frequency of IL-10-expressing ILC2 after TBI. Moreover, intracisternal administration of IL-33 activated AMPK, expanded ILC2, and suppressed ILC1 and ILC3 within the meninges of WT and Rag1-/- mice, but not Rag1-/- IL2rg-/- mice. Taken together, we identify AMPK as a brake on the expansion of proinflammatory, CNS-resident ILCs after brain injury. These findings establish a mechanistic framework whereby immunometabolic modulation of ILCs may direct the specificity, timing, and magnitude of cerebral immunity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain Injuries, Traumatic/enzymology , Brain Injuries, Traumatic/immunology , Immunity, Innate , Lymphocytes/immunology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , Adolescent , Adult , Aged , Animals , Brain Injuries, Traumatic/cerebrospinal fluid , Disease Models, Animal , Female , Humans , Lymphocytes/classification , Lymphocytes/pathology , Male , Meninges/immunology , Meninges/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Young Adult
7.
Neuromolecular Med ; 23(3): 371-382, 2021 09.
Article in English | MEDLINE | ID: mdl-33185833

ABSTRACT

Traumatic optic neuropathy (TON) is characterized by visual dysfunction after indirect or direct injury to the optic nerve following blunt head trauma. TON is associated with increased oxidative stress and inflammation resulting in retinal ganglion cell (RGC) death. Remote ischemic post-conditioning (RIC) has been shown to enhance endogenous protective mechanisms in diverse disease models including stroke, vascular cognitive impairment (VCI), retinal injury and optic nerve injury. However, the protective mechanisms underlying the improvement of retinal function and RGC survival after RIC treatment remain unclear. Here, we hypothesized that RIC therapy may be protective following TON by preventing RGC death, oxidative insult and inflammation in the mouse retina. To carry out the study, mice were divided in three different groups (Control, TON and TON + RIC). We harvested retinal tissue 5 days after TON induction for western blotting and histochemical analysis. We observed increased TON-induced retinal cell death compared with controls by cleaved caspase-3 immunohistochemistry. Furthermore, the TON cohort demonstrated increased TUNEL positive cells which were significantly attenuated by RIC. Immunofluorescence data showed that oxidative stress markers dihydroethidium (DHE), NOX-2 and nitrotyrosine expression were elevated in the TON group relative to controls and RIC therapy significantly reduced the expression level of these markers. Next, we found that the proinflammatory cytokine TNF-α was increased and anti-inflammatory IL-10 was decreased in plasma of TON animals, and RIC therapy reversed this expression level. Interestingly, western blotting of retinal tissue showed that RGC marker Brn3a and tight junction proteins (ZO-1 and Occludin), and AMPKα1 expression were downregulated in the TON group compared to controls. However, RIC significantly increased the expression levels of these proteins. Together these data suggest that RIC therapy activates endogenous protective mechanisms which may attenuate TON-induced oxidative stress and inflammation, and improves BRB integrity.


Subject(s)
Ischemic Postconditioning , Optic Nerve Injuries/therapy , Adenylate Kinase/biosynthesis , Adenylate Kinase/genetics , Animals , Blood-Retinal Barrier , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Death , Eye Proteins/biosynthesis , Eye Proteins/genetics , Hindlimb/blood supply , Interleukin-10/blood , Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Microglia/physiology , Models, Animal , NADPH Oxidase 2/analysis , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/prevention & control , Oxidative Stress , Retinal Ganglion Cells/pathology , Superoxides/analysis , Transcription Factor Brn-3A/biosynthesis , Transcription Factor Brn-3A/genetics , Tumor Necrosis Factor-alpha/blood , Tyrosine/analogs & derivatives , Tyrosine/analysis
8.
Br J Pharmacol ; 177(11): 2466-2477, 2020 06.
Article in English | MEDLINE | ID: mdl-31975437

ABSTRACT

BACKGROUND AND PURPOSE: Intravenous thrombolysis (IVT) after stroke enhances C3a generation, which may abrogate the benefits of reperfusion. The C3aR antagonist SB290157 is neuroprotective following transient but not permanent middle cerebral artery occlusion (MCAo). SB290157 remains untested in thromboembolic (TE) models, which better approximate human stroke and also facilitate testing in combination with IVT. We hypothesized SB290157 would confer neuroprotection in TE stroke with and without "late" IVT. EXPERIMENTAL APPROACH: We used two different models of TE stroke to examine the efficacy of SB290157 alone and in combination with late IVT. We evaluated the benefit of SB290157 in attenuating post-ischaemic behavioural deficits, infarction, brain oedema and haemorrhage. KEY RESULTS: Plasma C3a was elevated 6 hr after TE stroke alongside increased cerebrovascular C3aR expression, which was sustained to 4 weeks. Increased C3aR expression also was visualized in human ischaemic brain. In a photothrombotic (PT) stroke model, which exhibits rapid spontaneous reperfusion, SB290157 given at 1 hr post-PT significantly improved neurofunction and reduced infarction at 48 hr. In an embolic (eMCAo) model, SB290157 administered at 2 hr improved histological and functional outcomes. Conversely, late IVT administered 4.5 hr post-eMCAo was ineffective likely due to increased haemorrhage and brain oedema. However, SB290157 administered prior to late IVT ameliorated haemorrhage and oedema and improved outcomes. CONCLUSIONS AND IMPLICATIONS: We conclude that SB290157 is safe and effective with and without late IVT following TE stroke. Therefore, C3a receptor antagonist therapy represents a promising candidate for clinical translation in stroke, particularly as an adjuvant to IVT.


Subject(s)
Brain Ischemia , Stroke , Animals , Brain Ischemia/drug therapy , Fibrinolytic Agents/therapeutic use , Humans , Mice , Stroke/drug therapy , Thrombolytic Therapy , Treatment Outcome
9.
Neuromolecular Med ; 21(4): 505-516, 2019 12.
Article in English | MEDLINE | ID: mdl-31515728

ABSTRACT

Brain endothelial cells play an important role in maintaining blood flow homeostasis in the brain. Cerebral ischemia is a major cause of endothelial dysfunction which can disrupt the blood-brain barrier (BBB). Oxygen-glucose deprivation (OGD)/reperfusion promote cell death and BBB breakdown in brain endothelial cells. Acetyl-11-keto-ß-boswellic acid (AKBA), a biologically active phytoconstituent of the medicinal plant Boswellia serrata, has been shown to be protective against various inflammatory diseases as well as ischemic brain injury. The molecular mechanisms underlying these beneficial characteristics of AKBA are poorly understood. We subjected bEND.3 cells to OGD/reperfusion to investigate the protective role of AKBA in this model. We found that AKBA treatment attenuated endothelial cell death and oxidative stress assessed by means of TUNEL assay, cleaved-caspase-3, and dihydroethidium (DHE) staining. Furthermore, OGD downregulated tight junction proteins ZO-1 and Occludin levels, and increased the expressions of inflammatory cytokines TNF-α, ICAM-1, and complement C3a receptor (C3aR). We also noticed the increased phosphorylation of ERK 1/2 in bEND.3 cells in OGD group. AKBA treatment significantly attenuated expression levels of these inflammatory proteins and prevented the degradation of ZO-1 and Occludin following OGD. In conclusion, AKBA treatment provides protection against endothelial cell dysfunction following OGD by attenuating oxidative stress and inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Boswellia/chemistry , Complement Activation/drug effects , Complement Inactivating Agents/pharmacology , Endothelial Cells/drug effects , Hypoxia-Ischemia, Brain/metabolism , Oxidative Stress/drug effects , Plants, Medicinal/chemistry , Triterpenes/pharmacology , Animals , Cell Hypoxia , DNA Damage , Endothelial Cells/cytology , Endothelial Cells/metabolism , Glucose/pharmacology , Inflammation , MAP Kinase Signaling System/drug effects , Mice , Oxygen/pharmacology , Plant Extracts/chemistry , Reactive Oxygen Species/analysis
10.
Exp Neurobiol ; 28(2): 216-228, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31138990

ABSTRACT

The complement cascade is a central component of innate immunity which plays a critical role in brain inflammation. Complement C3a receptor (C3aR) is a key mediator of post-ischemic cerebral injury, and pharmacological antagonism of the C3a receptor is neuroprotective in stroke. Cerebral ischemia injures brain endothelial cells, causing blood brain barrier (BBB) disruption which further exacerbates ischemic neuronal injury. In this study, we used an in vitro model of ischemia (oxygen glucose deprivation; OGD) to investigate the protective effect of a C3aR antagonist (C3aRA, SB290157) on brain endothelial cells (bEnd.3). Following 24 hours of reperfusion, OGD-induced cell death was assessed by TUNEL and Caspase-3 staining. Western blot and immunocytochemistry were utilized to demonstrate that OGD upregulates inflammatory, oxidative stress and antioxidant markers (ICAM-1, Cox-2, Nox-2 and MnSOD) in endothelial cells and that C3aRA treatment significantly attenuate these markers. We also found that C3aRA administration restored the expression level of the tight junction protein occludin in endothelial cells following OGD. Interestingly, OGD/reperfusion injury increased the phosphorylation of ERK1/2 and C3aR inhibition significantly reduced the activation of ERK suggesting that endothelial C3aR may act via ERK signaling. Furthermore, exogenous C3a administration stimulates these same inflammatory mechanisms both with and without OGD, and C3aRA suppresses these C3a-mediated responses, supporting an antagonist role for C3aRA. Based on these results, we conclude that C3aRA administration attenuates inflammation, oxidative stress, ERK activation, and protects brain endothelial cells following experimental brain ischemia.

11.
Br J Pharmacol ; 176(13): 2250-2263, 2019 07.
Article in English | MEDLINE | ID: mdl-30931525

ABSTRACT

BACKGROUND AND PURPOSE: Inactivation of the gene for adenosine A2A receptors (ADORA2A for humans and Adora2a for rodents) protects against brain injury in experimental stroke. However, the cell-specific pathogenic effects of A2A receptors in thromboembolic stroke and the underlying mechanisms remain undefined. Here, we tested the hypothesis that inhibition of endothelial A2A receptors after thromboembolic stroke improves post-stroke outcomes via down-regulation of inflammation. EXPERIMENTAL APPROACH: Thromboembolic stroke was induced by embolic middle cerebral artery occlusion in mice. Post-stroke outcomes were determined with neurological deficit scoring, infarct volume, inflammatory marker expression, brain leukocyte infiltration, blood-brain barrier (BBB) leakage, and oedema assessment. Anti-inflammatory effects of silencing the gene for A2A receptors or pharmacological antagonism of these receptors were assessed in vitro. KEY RESULTS: Thromboembolic stroke induced Adora2a expression in the brain. Mice globally deficient in Adora2a (Adora2a-/- ) were resistant to stroke injury. Mice specifically deficient in endothelial Adora2a (Adora2aΔVEC ) showed reduced leukocyte infiltration, BBB leakage, and oedema after stroke, along with attenuated downstream proinflammatory markers, both in vivo and in vitro. The A2A receptor antagonist, KW 6002, also reduced brain injury and inflammation after stroke. Inactivation of ADORA2A inhibited endothelial inflammation via suppression of the NLRP3 inflammasome, down-regulating cleaved caspase 1 and IL-1ß expression. CONCLUSIONS AND IMPLICATIONS: Specific inactivation of endothelial A2A receptors mitigated ischaemic brain injury and improved post-stroke outcomes, at least partly, through anti-inflammatory effects via blockade of NLRP3 inflammasome activity. Our findings may open new approaches to vascular protection after ischaemic stroke.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neuroprotection , Receptor, Adenosine A2A/metabolism , Thromboembolism/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Brain/physiology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Purines/pharmacology , Receptor, Adenosine A2A/genetics , Stroke/physiopathology , THP-1 Cells
12.
Invest Ophthalmol Vis Sci ; 59(13): 5548-5557, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30480743

ABSTRACT

Purpose: Traumatic optic neuropathy (TON) is the most feared visual consequence of head and ocular trauma in both military and civilian communities, for which standard treatment does not exist. Animal models are critical for the development of novel TON therapies as well as the understanding of TON pathophysiology. However, the models currently used for TON have some limitations regarding consistency and mirroring the exact pathological progression of TON in closed ocular trauma. In this study, we modified the model of controlled cortical impact and adapted it for studying TON. Methods: We defined new standardized procedures to induce TON in mice, wherein the optic nerve is reproducibly exposed to a graded controlled impact of known velocity to produce a graded deficit in retinal ganglion cell (RGC) electrophysiological functions. Results: The key results of validating this newly modified model, "controlled orbital impact (COI)," included (1) the injury parameters (velocity as well as contusion depth and time), which were quantifiable and manageable to generate a wide range of TON severities; (2) a reproducible endpoint of diminished positive scotopic threshold response (pSTR) has been achieved without the interference of surgical variability and destruction of surrounding tissues; (3) the contralateral eyes showed no significant difference to the eyes of naïve mice, allowing them to be used as an internal control to minimize interindividual variability among mice; and (4) the occurrence of injury-associated mortality and/or ocular comorbidity was rare. Conclusions: Taken together, this model overcomes some limitations of prior TON mouse models and provides an innovative platform to identify therapeutic targets for neuroprotection and/or neurorestoration following traumatic ocular injury.


Subject(s)
Disease Models, Animal , Optic Nerve Injuries/physiopathology , Optic Nerve/physiopathology , Retina/physiopathology , Animals , Axons/pathology , Blotting, Western , Electroretinography , Mice , Mice, Inbred C57BL , Night Vision/physiology , Retinal Ganglion Cells/pathology
13.
J Exp Med ; 215(10): 2636-2654, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30190288

ABSTRACT

Spontaneous intracerebral hemorrhage (ICH) produces the highest acute mortality and worst outcomes of all stroke subtypes. Hematoma volume is an independent determinant of ICH patient outcomes, making clot resolution a primary goal of clinical management. Herein, remote-limb ischemic post-conditioning (RIC), the repetitive inflation-deflation of a blood pressure cuff on a limb, accelerated hematoma resolution and improved neurological outcomes after ICH in mice. Parabiosis studies revealed RIC accelerated clot resolution via a humoral-mediated mechanism. Whereas RIC increased anti-inflammatory macrophage activation, myeloid cell depletion eliminated the beneficial effects of RIC after ICH. Myeloid-specific inactivation of the metabolic regulator, AMPKα1, attenuated RIC-induced anti-inflammatory macrophage polarization and delayed hematoma resolution, providing a molecular link between RIC and immune activation. Finally, chimera studies implicated myeloid CD36 expression in RIC-mediated neurological recovery after ICH. Thus, RIC, a clinically well-tolerated therapy, noninvasively modulates innate immune responses to improve ICH outcomes. Moreover, immunometabolic changes may provide pharmacodynamic blood biomarkers to clinically monitor the therapeutic efficacy of RIC.


Subject(s)
AMP-Activated Protein Kinases/immunology , Hematoma/immunology , Ischemic Postconditioning , Macrophage Activation , Macrophages/immunology , Stroke/immunology , AMP-Activated Protein Kinases/genetics , Animals , Hematoma/pathology , Hematoma/therapy , Macrophages/pathology , Mice , Mice, Knockout , Stroke/pathology , Stroke/therapy
14.
Transl Stroke Res ; 9(1): 51-63, 2018 02.
Article in English | MEDLINE | ID: mdl-28755277

ABSTRACT

Vascular contributions to cognitive impairment and dementia (VCID) make up 50% of the cases of dementia. The purpose of this study was to determine the effect of chronic remote ischemic conditioning (C-RIC) on improving long-term (6 months) outcomes and cerebral blood flow (CBF) and collateral formation in a mouse model of VCID. Adult C57BL/6J male mice (10 weeks) were randomly assigned to four different groups: (1) sham-bilateral carotid artery stenosis (BCAS), (2) BCAS + sham RIC, (3) BCAS+C-RIC for 1 month (1MO), and (4) BCAS+C-RIC-4 months (4MO). CBF, cognitive impairment, and functional outcomes were performed up for 6 months after BCAS surgery. The expression of CD31, α-SMA, and myelin basic protein (MBP) was assessed by immunohistochemistry (IHC). Additional set of mice were randomized to sham, BCAS, and BCAS+C-RIC. The cerebrovascular angioarchitecture was studied with micro-CT. RIC therapy for either 1 or 4 months significantly improved CBF, new collateral formation, functional and cognitive outcomes, and prevented white matter damage. There was no difference between C-RIC for 1 or 4 months; IHC studies at 6 months showed an increase in brain CD31 and α-SMA expression indicating increased angiogenesis and MBP indicating preservation of white matter in animals receiving RIC. One month of daily RIC is as effective as 4 months of daily RIC in improving CBF, angiogenesis, and long-term functional outcomes (6 months) in a VCID model. This suggests that 1 month of RIC is sufficient to reduce cognitive impairment and induce beneficial cerebrovascular remodeling.


Subject(s)
Cerebrovascular Circulation/physiology , Cognitive Dysfunction/therapy , Dementia, Vascular/therapy , Ischemic Preconditioning/methods , Vascular Remodeling/physiology , Actins/metabolism , Angiography , Animals , Cognitive Dysfunction/physiopathology , Cytokines/blood , Dementia, Vascular/physiopathology , Disease Models, Animal , Gene Expression Regulation/physiology , Macrophages/pathology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/prevention & control , Nitrites/blood , Random Allocation , Statistics, Nonparametric , Time Factors
15.
Brain Behav Immun ; 68: 224-237, 2018 02.
Article in English | MEDLINE | ID: mdl-29079445

ABSTRACT

Inflammation is an important mediator of secondary neurological injury after traumatic brain injury (TBI). Endocannabinoids, endogenously produced arachidonate based lipids, have recently emerged as powerful anti-inflammatory compounds, yet the molecular and cellular mechanisms underlying these effects are poorly defined. Endocannabinoids are physiological ligands for two known cannabinoid receptors, CB1R and CB2R. In the present study, we hypothesized that selective activation of CB2R attenuates neuroinflammation and reduces neurovascular injury after TBI. Using a murine controlled cortical impact (CCI) model of TBI, we observed a dramatic upregulation of CB2R within infiltrating myeloid cells beginning at 72 h. Administration of the selective CB2R agonist, GP1a (1-5 mg/kg), attenuated pro-inflammatory M1 macrophage polarization, increased anti-inflammatory M2 polarization, reduced edema development, enhanced cerebral blood flow, and improved neurobehavioral outcomes after TBI. In contrast, the CB2R antagonist, AM630, worsened outcomes. Taken together, our findings support the development of selective CB2R agonists as a therapeutic strategy to improve TBI outcomes while avoiding the psychoactive effects of CB1R activation.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Indenes/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB2/metabolism , Animals , Brain Injuries/complications , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Cannabinoids/therapeutic use , Cannabis , Disease Models, Animal , Endocannabinoids/therapeutic use , Inflammation/complications , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroimmunomodulation/physiology , Receptor, Cannabinoid, CB2/physiology , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/physiology
16.
Front Immunol ; 8: 922, 2017.
Article in English | MEDLINE | ID: mdl-28861078

ABSTRACT

Innate lymphoid cells (ILCs) are master regulators of immune and inflammatory responses, but their own regulatory mechanisms and functional roles of their subtypes (i.e., ILC1s-ILC3s) remain largely unresolved. Interestingly, AMP-activated protein kinase (AMPK), influences inflammatory responses, but its role in modulation of ILCs is not known. Periodontitis is a prevalent disorder with impairment of immune and inflammatory responses contributing importantly to its pathogenesis; however, neither the role of ILCs nor AMPK has been explored in this condition. We tested the hypotheses that (a) periodontitis increases ILCs and expression of relevant cytokines thereby contributing to inflammation and (b) knockdown of AMPK worsens indices of periodontitis in association with further increases in subtypes of ILCs and cytokine expression. The studies utilized wild-type (WT) and AMPK knockout (KO) mice, subjected to ligature-induced periodontitis or sham operation, in association with the use of micro-CT for assessment of bone loss, immunogold electron microscopy to show presence of ILCs in periodontal tissues, flow cytometry for quantitative assessment of subtypes of ILCs and RT-polymerase chain reaction analyses to measure mRNA expression of several relevant cytokines. The results for the first time show (a) presence of each subtype of ILCs in periodontal tissues of sham control and periodontitis animals, (b) that periodontitis is associated with increased frequencies of ILC1s-ILC3s with the effect more marked for ILC2s and differential phenotypic marker expression for ILC3s, (c) that AMPK KO mice display exacerbation of indices of periodontitis in association with further increases in the frequency of subtypes of ILCs with persistence of ILC2s effect, and (d) that periodontitis increased mRNA for interleukin (IL)-33, but not IL-5 or IL-13, in WT mice but expression of these cytokines was markedly increased in AMPK KO mice with periodontitis. Subsequently, we showed that human periodontitis is associated with increases in each ILCs subtype with the effect more marked for ILC2s and that mRNA expressions for IL-33 and IL-5 are markedly greater for sites affected by periodontitis than healthy sites. Collectively, these novel observations indicate a pivotal role for ILCs in pathogenesis of periodontitis and that AMPK is a regulator of their phenotype expression in this condition.

17.
EMBO Mol Med ; 9(9): 1263-1278, 2017 09.
Article in English | MEDLINE | ID: mdl-28751580

ABSTRACT

The nucleoside adenosine is a potent regulator of vascular homeostasis, but it remains unclear how expression or function of the adenosine-metabolizing enzyme adenosine kinase (ADK) and the intracellular adenosine levels influence angiogenesis. We show here that hypoxia lowered the expression of ADK and increased the levels of intracellular adenosine in human endothelial cells. Knockdown (KD) of ADK elevated intracellular adenosine, promoted proliferation, migration, and angiogenic sprouting in human endothelial cells. Additionally, mice deficient in endothelial ADK displayed increased angiogenesis as evidenced by the rapid development of the retinal and hindbrain vasculature, increased healing of skin wounds, and prompt recovery of arterial blood flow in the ischemic hindlimb. Mechanistically, hypomethylation of the promoters of a series of pro-angiogenic genes, especially for VEGFR2 in ADK KD cells, was demonstrated by the Infinium methylation assay. Methylation-specific PCR, bisulfite sequencing, and methylated DNA immunoprecipitation further confirmed hypomethylation in the promoter region of VEGFR2 in ADK-deficient endothelial cells. Accordingly, loss or inactivation of ADK increased VEGFR2 expression and signaling in endothelial cells. Based on these findings, we propose that ADK downregulation-induced elevation of intracellular adenosine levels in endothelial cells in the setting of hypoxia is one of the crucial intrinsic mechanisms that promote angiogenesis.


Subject(s)
Adenosine/metabolism , Epigenesis, Genetic , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Adenosine Kinase/genetics , Adenosine Kinase/metabolism , Animals , Aorta/metabolism , DNA Methylation , Humans , Mice , Promoter Regions, Genetic , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
18.
Biochim Biophys Acta Mol Basis Dis ; 1863(10 Pt B): 2614-2626, 2017 10.
Article in English | MEDLINE | ID: mdl-28533056

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of mortality and long-term morbidity worldwide. Despite decades of pre-clinical investigation, therapeutic strategies focused on acute neuroprotection failed to improve TBI outcomes. This lack of translational success has necessitated a reassessment of the optimal targets for intervention, including a heightened focus on secondary injury mechanisms. Chronic immune activation correlates with progressive neurodegeneration for decades after TBI; however, significant challenges remain in functionally and mechanistically defining immune activation after TBI. In this review, we explore the burgeoning evidence implicating the acute release of damage associated molecular patterns (DAMPs), such as adenosine 5'-triphosphate (ATP), high mobility group box protein 1 (HMGB1), S100 proteins, and hyaluronic acid in the initiation of progressive neurological injury, including white matter loss after TBI. The role that pattern recognition receptors, including toll-like receptor and purinergic receptors, play in progressive neurological injury after TBI is detailed. Finally, we provide support for the notion that resident and infiltrating macrophages are critical cellular targets linking acute DAMP release with adaptive immune responses and chronic injury after TBI. The therapeutic potential of targeting DAMPs and barriers to clinical translational, in the context of TBI patient management, are discussed.


Subject(s)
Brain Injuries, Traumatic/metabolism , White Matter/metabolism , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/therapy , HMGB1 Protein/immunology , HMGB1 Protein/metabolism , Humans , Hyaluronic Acid/immunology , Hyaluronic Acid/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , S100 Proteins/immunology , S100 Proteins/metabolism , White Matter/immunology , White Matter/pathology
19.
J Immunol ; 198(9): 3615-3626, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28341672

ABSTRACT

Traumatic brain injury (TBI) is a major public health issue, producing significant patient mortality and poor long-term outcomes. Increasing evidence suggests an important, yet poorly defined, role for the immune system in the development of secondary neurologic injury over the days and weeks following a TBI. In this study, we tested the hypothesis that peripheral macrophage infiltration initiates long-lasting adaptive immune responses after TBI. Using a murine controlled cortical impact model, we used adoptive transfer, transgenic, and bone marrow chimera approaches to show increased infiltration and proinflammatory (classically activated [M1]) polarization of macrophages for up to 3 wk post-TBI. Monocytes purified from the injured brain stimulated the proliferation of naive T lymphocytes, enhanced the polarization of T effector cells (TH1/TH17), and decreased the production of regulatory T cells in an MLR. Similarly, elevated T effector cell polarization within blood and brain tissue was attenuated by myeloid cell depletion after TBI. Functionally, C3H/HeJ (TLR4 mutant) mice reversed M1 macrophage and TH1/TH17 polarization after TBI compared with C3H/OuJ (wild-type) mice. Moreover, brain monocytes isolated from C3H/HeJ mice were less potent stimulators of T lymphocyte proliferation and TH1/TH17 polarization compared with C3H/OuJ monocytes. Taken together, our data implicate TLR4-dependent, M1 macrophage trafficking/polarization into the CNS as a key mechanistic link between acute TBI and long-term, adaptive immune responses.


Subject(s)
Brain Injuries, Traumatic/immunology , Macrophages/physiology , Th1 Cells/immunology , Th17 Cells/immunology , Toll-Like Receptor 4/genetics , Adaptive Immunity , Adoptive Transfer , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Phenotype
20.
Chin J Integr Med ; 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28197933

ABSTRACT

OBJECTIVE: To investigate the oxidative stress induced by 6 h of immobilization stress in Albino Wistar rats. Further, the pre- and post-treatment of aqueous garlic extract was studied to evaluate its preventive and curative efficacy on stress-induced altered oxidative parameters in rats. METHODS: Albino Wistar rats were exposed to 6 h of immobilization stress, and received garlic extract (100 mg/kg body weight) treatment pre- or post-stress exposure. The oxidative status of plasma after various treatments were evaluated by determining the levels of reduced glutathione, glucose, uric acid, thiobarbituric acid reactive substances, aspartate aminotransferase (AST), alanine transaminase (ALT), alkaline phosphatase (ALP) and the activities of superoxide dismutase, catalase and glutathione-S-transferase by standardized procedures. RESULTS: Immobilization of rats generated oxidative stress in rat plasma, by decreasing the activities of antioxidant enzymes, glutathione levels and glucose, while increasing the lipid peroxidation, serum glutamic oxaloacetic transaminase, serum glutamic pyruvic transaminase, ALP and uric acid compared to the non-stressed controls (P<0.01). The garlic extract administration both pre- and post-stress exposure significantly prevented the rise in the diagnostic liver enzymes and reverted the decrease of antioxidant enzymatic activities compared to the stressed group (P<0.05 or P <0.01). Post-stress treatment of extract was found more effective than pre-stress treatment in reverting the values back to normal (P<0.01). CONCLUSION: Garlic extract seems promising as a nutritional supplement for scavenging free radicals generated in the plasma and to prevent resulting oxidative stress.

SELECTION OF CITATIONS
SEARCH DETAIL
...