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











Publication year range
1.
Pharmaceutics ; 15(5)2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37242599

ABSTRACT

Our lab previously established that metformin, a first-line type two diabetes treatment, activates the Nrf2 pathway and improves post-stroke recovery. Metformin's brain permeability value and potential interaction with blood-brain barrier (BBB) uptake and efflux transporters are currently unknown. Metformin has been shown to be a substrate of organic cationic transporters (Octs) in the liver and kidneys. Brain endothelial cells at the BBB have been shown to express Octs; thus, we hypothesize that metformin uses Octs for its transport across the BBB. We used a co-culture model of brain endothelial cells and primary astrocytes as an in vitro BBB model to conduct permeability studies during normoxia and hypoxia using oxygen-glucose deprivation (OGD) conditions. Metformin was quantified using a highly sensitive LC-MS/MS method. We further checked Octs protein expression using Western blot analysis. Lastly, we completed a plasma glycoprotein (P-GP) efflux assay. Our results showed that metformin is a highly permeable molecule, uses Oct1 for its transport, and does not interact with P-GP. During OGD, we found alterations in Oct1 expression and increased permeability for metformin. Additionally, we showed that selective transport is a key determinant of metformin's permeability during OGD, thus, providing a novel target for improving ischemic drug delivery.

2.
Fluids Barriers CNS ; 20(1): 17, 2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36899432

ABSTRACT

Electronic nicotine delivery systems (ENDS), also commonly known as electronic cigarettes (e-cigs) are considered in most cases as a safer alternative to tobacco smoking and therefore have become extremely popular among all age groups and sex. It is estimated that up to 15% of pregnant women are now using e-cigs in the US which keeps increasing at an alarming rate. Harmful effects of tobacco smoking during pregnancy are well documented for both pregnancy and postnatal health, however limited preclinical and clinical studies exist to evaluate the long-term effects of prenatal e-cig exposure on postnatal health. Therefore, the aim of our study is to evaluate the effect of maternal e-cig use on postnatal blood-brain barrier (BBB) integrity and behavioral outcomes of mice of varying age and sex. In this study, pregnant CD1 mice (E5) were exposed to e-Cig vapor (2.4% nicotine) until postnatal day (PD) 7. Weight of the offspring was measured at PD0, PD7, PD15, PD30, PD45, PD60 and PD90. The expression of structural elements of the BBB, tight junction proteins (ZO-1, claudin-5, occludin), astrocytes (GFAP), pericytes (PDGFRß) and the basement membrane (laminin α1, laminin α4), neuron specific marker (NeuN), water channel protein (AQP4) and glucose transporter (GLUT1) were analyzed in both male and female offspring using western blot and immunofluorescence. Estrous cycle was recorded by vaginal cytology method. Long-term motor and cognitive functions were evaluated using open field test (OFT), novel object recognition test (NORT) and morris water maze test (MWMT) at adolescence (PD 40-45) and adult (PD 90-95) age. In our study, significantly reduced expression of tight junction proteins and astrocyte marker were observed in male and female offspring until PD 90 (P < 0.05). Additionally, prenatally e-cig exposed adolescent and adult offspring showed impaired locomotor, learning, and memory function compared to control offspring (P < 0.05). Our findings suggest that prenatal e-cig exposure induces long-term neurovascular changes of neonates by disrupting postnatal BBB integrity and worsening behavioral outcomes.


Subject(s)
Electronic Nicotine Delivery Systems , Vaping , Pregnancy , Female , Animals , Male , Mice , Humans , Blood-Brain Barrier , Nicotine , Tight Junction Proteins
3.
Cell Mol Neurobiol ; 43(5): 2105-2127, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36201091

ABSTRACT

Astrocytes have been implicated in the onset and complication of various central nervous system (CNS) injuries and disorders. Uncontrolled astrogliosis (gliosis), while a necessary process for recovery after CNS trauma, also causes impairments in CNS performance and functions. The ability to preserve astrocyte health and better regulate the gliosis process could play a major role in controlling damage in the aftermath of acute insults and during chronic dysfunction. Here in, we demonstrate the ability of dental pulp-derived stem cells (DPSCs) in protecting the health of astrocytes during induced gliosis. First of all, we have characterized the expression of genes in primary astrocytes that are relevant to the pathological conditions of CNS by inducing gliosis. Subsequently, we found that astrocytes co-cultured with DPSCs reduced ROS production, NRF2 and GCLM expressions, mitochondrial membrane potential, and mitochondrial functions compared to the astrocytes that were not co-cultured with DPSCs in gliosis condition. In addition, hyperactive autophagy was also decreased in astrocytes that were co-cultured with DPSCs compared to the astrocytes that were not co-cultured with DPSCs during gliosis. This reversal and mitigation of gliosis in astrocytes were partly due to induction of neurogenesis in DPSCs through enhanced expressions of the neuronal genes like GFAP, NeuN, and Synapsin in DPSCs and by secretion of higher amounts of neurotropic factors, such as BDNF, GDNF, and TIMP-2. Protein-Protein docking analysis suggested that BDNF and GDNF can bind with CSPG4 and block the downstream signaling. Together these findings demonstrate novel functions of DPSCs to preserve astrocyte health during gliosis.


Subject(s)
Astrocytes , Gliosis , Humans , Brain-Derived Neurotrophic Factor , Dental Pulp , Glial Cell Line-Derived Neurotrophic Factor , Cells, Cultured
4.
Fluids Barriers CNS ; 19(1): 74, 2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36085043

ABSTRACT

BACKGROUND: The short and long-term health effects of JUUL electronic cigarette (e-Cig) are largely unknown and warrant extensive research. We hypothesized that JUUL exposure could cause cerebrovascular toxicities impacting the progression and outcome of ischemic stroke comparable to tobacco smoke (TS) exposure. METHODS: We exposed male C57 mice to TS/JUUL vapor for 14 days. LCMS/MS was used to measure brain and plasma nicotine and cotinine level. Transient middle cerebral artery occlusion (tMCAO) followed by reperfusion was used to mimic ischemic stroke. Plasma levels of IL-6 and thrombomodulin were assessed by enzyme-linked immunosorbent assay. At the same time, western blotting was used to study blood-brain barrier (BBB) tight junction (TJ) proteins expression and key inflammatory and oxidative stress markers. RESULTS: tMCAO upregulated IL-6 and decreased plasma thrombomodulin levels. Post-ischemic brain injury following tMCAO was significantly worsened by JUUL/TS pre-exposure. TJ proteins expression was also downregulated by JUUL/TS pre-exposure after tMCAO. Like TS, exposure to JUUL downregulated the expression of the antioxidant Nrf2. ICAM-1 was upregulated in mice subjected to tMCAO following pre-exposure to TS or JUUL, with a greater effect of TS than JUUL. CONCLUSIONS: These results suggest that JUUL exposure could negatively impact the cerebrovascular system, although to a lesser extent than TS exposure.


Subject(s)
Electronic Nicotine Delivery Systems , Ischemic Stroke , Animals , Blood-Brain Barrier , Interleukin-6 , Male , Mice , Thrombomodulin , Tight Junction Proteins
5.
Pharm Res ; 39(7): 1587-1598, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35239135

ABSTRACT

PURPOSE: Neurolysin (Nln) is a peptidase that functions to preserve the brain following ischemic stroke by hydrolyzing various neuropeptides. Nln activation has emerged as an attractive drug discovery target for treatment of ischemic stroke. Among first-in-class peptidomimetic Nln activators, we selected three lead compounds (9d, 10c, 11a) for quantitative pharmacokinetic analysis to provide valuable information for subsequent preclinical development. METHODS: Pharmacokinetic profile of these compounds was studied in healthy and ischemic stroke-induced mice after bolus intravenous administration. Brain concentration and brain uptake clearance (Kin) was calculated from single time point analysis. The inter-relationship between LogP with in-vitro and in-vivo permeability was studied to determine CNS penetration. Brain slice uptake method was used to study tissue binding, whereas P-gp-mediated transport was evaluated to understand the potential brain efflux of these compounds. RESULTS: According to calculated parameters, all three compounds showed a detectable amount in the brain after intravenous administration at 4 mg/kg; however, 11a had the highest brain concentration and brain uptake clearance. A strong correlation was documented between in-vitro and in-vivo permeability data. The efflux ratio of 10c was ~6-fold higher compared to 11a and correlated well with its lower Kin value. In experimental stroke animals, the Kin of 11a was significantly higher in ischemic vs. contralateral and intact hemispheres, though it remained below its A50 value required to activate Nln. CONCLUSIONS: Collectively, these preclinical pharmacokinetic studies reveal promising BBB permeability of 11a and indicate that it can serve as an excellent lead for developing improved drug-like Nln activators.


Subject(s)
Ischemic Stroke , Peptidomimetics , Stroke , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Metalloendopeptidases , Mice , Peptidomimetics/metabolism , Stroke/drug therapy
6.
J Pharmacol Exp Ther ; 375(3): 498-509, 2020 12.
Article in English | MEDLINE | ID: mdl-33033170

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 virus, is turning out to be one of the most devastating global pandemics in the history of humankind. There is a shortage of effective therapeutic strategies or preventative vaccines for this disease to date. A rigorous investigation is needed for identifying and developing more effective therapeutic strategies for COVID-19. Angiotensin-converting enzyme 2 (ACE2), a crucial factor in COVID-19 pathogenesis, has been identified as a potential target for COVID-19 treatment. Smoking and vaping are potential risk factors for COVID-19 that are also shown to upregulate ACE2 expression. In this review, we have discussed the pathobiology of COVID-19 in the lungs and brain and the role of ACE2 in the transmission and pathobiology of this disease. Furthermore, we have shown possible interactions between nicotine/smoking and ACE2 in the lungs and brain, which could aggravate the transmission and pathobiology of COVID-19, resulting in a poor disease outcome. SIGNIFICANCE STATEMENT: This review addresses the present global pandemic of coronavirus disease 2019 (COVID-19) with respect to its pathobiology in the lungs and brain. It focuses on the potential negative impact of tobacco and nicotine exposure on the outcomes of this disease by interaction with the angiotensin-converting enzyme 2 receptor. It adds to the time-sensitive and critically important growing knowledge about the risk factors, transmission, pathobiology, and prognosis of COVID-19.


Subject(s)
COVID-19/epidemiology , Smoking/epidemiology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/virology , COVID-19/etiology , COVID-19/transmission , Humans , Lung/drug effects , Lung/metabolism , Lung/virology , Nicotine/metabolism , Nicotine/toxicity , SARS-CoV-2/pathogenicity , Smoking/adverse effects
7.
J Neurochem ; 153(1): 63-79, 2020 04.
Article in English | MEDLINE | ID: mdl-31883376

ABSTRACT

It has been shown that prenatal nicotine and tobacco smoke exposure can cause different neurobehavioral disorders in the offspring. We hypothesize that prenatal exposure to nicotine-containing electronic cigarette (e-Cig) vapor can predispose newborn to enhanced sensitivity to hypoxic-ischemic (HI) brain injury and impaired motor and cognitive functions. In this study, pregnant CD1 mice were exposed to e-Cig vapor (2.4% nicotine). Primary cortical neurons isolated from e-Cig exposed fetus were exposed to oxygen-glucose deprivation followed by reoxygenation (OGD/R) to mimic HI brain injury. Cell viability and glucose utilization were analyzed in these neurons. HI brain injury was induced in 8-9-day-old pups. Short-term brain injury was evaluated by triphenyltetrazolium chloride staining. Long-term motor and cognitive functions were evaluated by open field, novel object recognition, Morris water maze, and foot fault tests. Western blotting and immunofluorescence were done to characterize glucose transporters in offspring brain. We found that e-Cig exposed neurons demonstrated decreased cell viability and glucose utilization in OGD/R. Prenatally e-Cig exposed pups also had increased brain injury and edema 24 hr after HI brain injury. Further, in utero e-Cig exposed offspring with HI brain injury displayed impaired memory, learning, and motor coordination at adolescence. Additionally, the expression of glucose transporters decreased in e-Cig exposed offspring brain after HI brain injury. These results indicate that reduced glucose utilization can contribute to prenatal e-Cig exposure induced worsened HI brain injury in offspring. This study is instrumental in elucidating the possible deleterious effects of e-Cig use in the general population.


Subject(s)
Brain/drug effects , Brain/metabolism , Electronic Nicotine Delivery Systems , Glucose/metabolism , Hypoxia-Ischemia, Brain/etiology , Nicotine/toxicity , Animals , Animals, Newborn , Brain Chemistry , Cells, Cultured , Cerebral Cortex/embryology , Cognition/drug effects , Female , Glucose/administration & dosage , Glucose Transporter Type 1/analysis , Male , Maternal-Fetal Exchange , Mice , Neurons/drug effects , Neurons/metabolism , Oxygen/administration & dosage , Pregnancy , Prenatal Exposure Delayed Effects , Prognosis
8.
J Neurochem ; 153(1): 120-137, 2020 04.
Article in English | MEDLINE | ID: mdl-31486527

ABSTRACT

Previous studies documented up-regulation of peptidase neurolysin (Nln) after brain ischemia, however, the significance of Nln function in the post-stroke brain remained unknown. The aim of this study was to assess the functional role of Nln in the brain after ischemic stroke. Administration of a specific Nln inhibitor Agaricoglyceride A (AgaA) to mice after stroke in a middle cerebral artery occlusion model, dose-dependently aggravated injury measured by increased infarct and edema volumes, blood-brain barrier disruption, increased levels of interleukin 6 and monocyte chemoattractant protein-1, neurological and motor deficit 24 h after stroke. In this setting, AgaA resulted in inhibition of Nln in the ischemic hemisphere leading to increased levels of Nln substrates bradykinin, neurotensin, and substance P. AgaA lacked effects on several physiological parameters and appeared non-toxic to mice. In a reverse approach, we developed an adeno-associated viral vector (AAV2/5-CAG-Nln) to overexpress Nln in the mouse brain. Applicability of AAV2/5-CAG-Nln to transduce catalytically active Nln was confirmed in primary neurons and in vivo. Over-expression of Nln in the mouse brain was also accompanied by decreased levels of its substrates. Two weeks after in vivo transduction of Nln using the AAV vector, mice were subjected to middle cerebral artery occlusion and the same outcome measures were evaluated 72 h later. These experiments revealed that abundance of Nln in the brain protects animals from stroke. This study is the first to document functional significance of Nln in pathophysiology of stroke and provide evidence that Nln is an endogenous mechanism functioning to preserve the brain from ischemic injury.


Subject(s)
Brain/physiopathology , Metalloendopeptidases/physiology , Stroke/physiopathology , Animals , Edema , Gene Expression Regulation , Glycerides/pharmacology , Infarction, Middle Cerebral Artery , Male , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Mice , Recombinant Proteins/drug effects , Stroke/etiology , Stroke/pathology , Transfection
9.
Pharmaceutics ; 11(9)2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31509975

ABSTRACT

Transporters (expressed) at the blood-brain barrier (BBB) can play an essential role in the treatment of brain injury by transporting neuroprotective substance to the central nervous system. The goal of this study was to understand the role of organic anion transporting polypeptide (OATP1; OATP1A2 in humans and oatp1a4 in rodents) in the transport of a potent opioid receptor agonist, biphalin, across the BBB during ischemic stroke. Brain microvascular endothelial cells (BMECs) that were differentiated from human induced pluripotent stem cells (iPSCs) were used in the present study. The effect of oxygen-glucose deprivation (OGD) and reperfusion on the OATP1 expression, uptake, and transport of biphalin was measured in induced pluripotent stem cells differentiated brain microvascular endothelial cells (iPSC-BMECs) in the presence and absence of an OATP1 substrate, estrone-3-sulfate (E3S). Biphalin brain permeability was quantified while using a highly sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. It was found that iPSC-BMECs expressed OATP1. In vitro studies showed that biphalin BBB uptake and transport decreased in the presence of an OATP1 specific substrate. It was also observed that OGD and reperfusion modulate the expression and function of OATP1 in BMECs. This study strongly demonstrates that OATP1 contributes to the transport of biphalin across the BBB and increased expression of OATP1 during OGD-reperfusion could provide a novel target for improving ischemic brain drug delivery of biphalin or other potential neurotherapeutics that have affinity to this BBB transporter.

10.
Am J Physiol Cell Physiol ; 316(1): C2-C15, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30207783

ABSTRACT

Transporters at the neurovascular unit (NVU) are vital for the regulation of normal brain physiology via ion, water, and nutrients movement. In ischemic stroke, the reduction of cerebral blood flow causes several complex pathophysiological changes in the brain, one of which includes alterations of the NVU transporters, which can exacerbate stroke outcome by increased brain edema (by altering ion, water, and glutamate transporters), altered energy metabolism (by altering glucose transporters), and enhanced drug toxicity (by altering efflux transporters). Smoking and diabetes are common risk factors as well as coexisting conditions in ischemic stroke that are also reported to change the expression and function of NVU transporters. Coexistence of these conditions could cause an additive effect in terms of the alterations of brain transporters that might lead to worsened ischemic stroke prognosis and recovery. In this review, we have discussed the effects of ischemic stroke, smoking, and diabetes on some essential NVU transporters and how the simultaneous presence of these conditions can affect the clinical outcome after an ischemic episode. Further scientific investigations are required to elucidate changes in NVU transport in cerebral ischemia, which can lead to better, personalized therapeutic interventions tailor-made for these comorbid conditions.


Subject(s)
Brain Ischemia/metabolism , Diabetes Mellitus/metabolism , Membrane Transport Proteins/metabolism , Neurovascular Coupling/physiology , Smoking/metabolism , Stroke/metabolism , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Brain/pathology , Brain Ischemia/pathology , Diabetes Mellitus/pathology , Glucose Transport Proteins, Facilitative/metabolism , Humans , Ion Transport/physiology , Neurons/metabolism , Neurons/pathology , Smoking/pathology , Stroke/pathology
11.
Brain Res ; 1699: 166-176, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30165043

ABSTRACT

Brain edema is one of the critical factors causing hightened disability and mortality in stroke patients, which is exaggerated further in diabetic patients. Organic osmolytes could play a critical role in the maintenance of cytotoxic edema. The present study was aimed to assess the role of myo-inositol, an organic osmolyte, on stroke outcome in diabetic and non-diabetic animals. In situ brain perfusion and acute brain slice methods were used to assess transport of myo-inositol across the blood-brain barrier and uptake by brain cells using non-diabetic (C57BL/6) and diabetic (streptozotocin-induced) mice, respectively. In vitro studies were conducted to assess the role of myo-inositol during and after ischemia utilizing oxygen glucose deprivation (OGD) and reperfusion. Further, the expression of transporters, such as SGLT6, SMIT1 and AQP4 were measured using immunofluorescence. Therapeutic efficacy of myo-inositol was evaluated in a transient middle cerebral artery occlusion (tMCAO) mouse model using non-diabetic (C57BL/6) and diabetic (db/db) mice. Myo-inositol release from and uptake in astrocytes and altered expression of myo-inositol transporters at different OGD timepoints revealed the role of myo-inositol and myo-inositol transporters during ischemia reperfusion. Further, hyperglycemic conditions reduced myo-inositol uptake in astrocytes. Interestingly, in in-vivo tMCAO, infarct and edema ratios following 24 h reperfusion decreased in myo-inositol treated mice. These results were supported by improvement in behavioral outcomes in open-field test, corner test and neurological score in both non-diabetic and db/db animals. Our data suggest that myo-inositol and myo-inositol transporters may provide neuroprotection during/following stroke both in non-diabetic and diabetic conditions.


Subject(s)
Brain Ischemia/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Inositol/pharmacology , Stroke/drug therapy , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Glucose/deficiency , Male , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Primary Cell Culture , Stroke/complications , Stroke/metabolism , Stroke/pathology , Tissue Culture Techniques
12.
Redox Biol ; 13: 353-362, 2017 10.
Article in English | MEDLINE | ID: mdl-28646795

ABSTRACT

Recently published in vitro and in vivo findings strongly suggest that BBB impairment and increased risk for stroke by tobacco smoke (TS) closely resemble that of type-2 diabetes (2DM) and develop largely in response to common key modulators such oxidative stress (OS), inflammation and alterations of the endogenous antioxidative response system (ARE) regulated by the nuclear factor erythroid 2-related factor (Nrf2). Preclinical studies have also shown that nicotine (the principal e-liquid's ingredient used in e-cigarettes) can also cause OS, exacerbation of cerebral ischemia and secondary brain injury. Herein we provide evidence that likewise to TS, chronic e-Cigarette (e-Cig) vaping can be prodromal to the loss of blood-brain barrier (BBB) integrity and vascular inflammation as well as act as a promoting factor for the onset of stroke and worsening of post-ischemic brain injury. In addition, recent reports have shown that Metformin (MF) treatment before and after ischemic injury reduces stress and inhibits inflammatory responses. Recent published data by our group revealead that MF promotes the activation of counteractive mechanisms mediated by the activation of Nrf2 which drastically reduce TS toxicity at the brain and cerebrovascular levels and protect BBB integrity. In this study we provide additional in vivo evidence showing that MF can effectively reduce the oxidative and inflammatory risk for stroke and attenuate post-ischemic brain injury promoted by TS and e-Cig vaping. Our data also suggest that MF administration could be extended as prophylactic care during the time window required for the renormalization of the risk levels of stroke following smoking cessation thus further studies in that direction are warrated.


Subject(s)
Antioxidants/therapeutic use , Blood-Brain Barrier/drug effects , Hypoglycemic Agents/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Metformin/therapeutic use , Tobacco Smoking/adverse effects , Vaping/adverse effects , Animals , Antioxidants/pharmacology , Blood-Brain Barrier/metabolism , Cells, Cultured , Hypoglycemic Agents/pharmacology , Male , Metformin/pharmacology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Tobacco Smoking/drug therapy
13.
Redox Biol ; 12: 58-69, 2017 08.
Article in English | MEDLINE | ID: mdl-28212524

ABSTRACT

Cigarette smoking (CS) is associated with vascular endothelial dysfunction in a causative way primarily related to the TS content of reactive oxygen species (ROS), nicotine, and inflammation. TS promotes glucose intolerance and increases the risk of developing type-2 diabetes mellitus (2DM) with which it shares other pathogenic traits including the high risk of cerebrovascular and neurological disorders like stroke via ROS generation, inflammation, and blood-brain barrier (BBB) impairment. Herein we provide evidence of the role played by nuclear factor erythroid 2-related factor (Nrf2) in CS-induced cerebrobvascular/BBB impairments and how these cerebrovascular harmful effects can be circumvented by the use of metformin (MF; a widely prescribed, firstline anti-diabetic drug) treatment. Our data in fact revealed that MF activates counteractive mechanisms primarily associated with the Nrf2 pathway which drastically reduce CS toxicity at the cerebrovascular level. These include the suppression of tight junction (TJ) protein downregulation and loss of BBB integrity induced by CS, reduction of inflammation and oxidative stress, renormalization of the expression levels of the major BBB glucose transporter Glut-1 and that of the anticoagulant factor thrombomodulin. Further, we provide additional insights on the controversial interplay between Nrf2 and AMPK.


Subject(s)
Brain/drug effects , Metformin/administration & dosage , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/administration & dosage , Smoke/adverse effects , Animals , Blood-Brain Barrier/drug effects , Brain/cytology , Brain/metabolism , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Metformin/pharmacology , Mice , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Nicotiana/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL