scholarly journals Severe Hypoglycemia Contributing to Cognitive Dysfunction in Diabetic Mice Is Associated With Pericyte and Blood–Brain Barrier Dysfunction

2021 ◽  
Vol 13 ◽  
Author(s):  
Lu Lin ◽  
Yubin Wu ◽  
Zhou Chen ◽  
Lishan Huang ◽  
Lijing Wang ◽  
...  

Background: Severe hypoglycemia can cause cognitive impairment in diabetic patients, but the underlying molecular mechanism remains unclear.Objective: To assess the effect of severe hypoglycemia on cognitive function in diabetic mice to clarify the relationship between the mechanism and dysfunction of pericytes and the blood–brain barrier (BBB).Method: We established type 1 diabetes mellitus in 80 male C57BL/6J mice by intraperitoneal injection of streptozotocin (150 mg/kg). Further intraperitoneal injection of short-acting insulin induced severe hypoglycemia. The mice were divided into normal, diabetes, and diabetic + severe hypoglycemia groups, and their blood glucose and general weight index were examined. Pericyte and BBB morphology and function were detected by histological and western blot analyses, BBB permeability was detected by Evans blue staining, and cognitive function was detected with the Morris water maze.Results: Severe hypoglycemia aggravated the histological damage, BBB damage, brain edema, and pericyte loss in the diabetic mice. It also reduced the expression of the BBB tight junction proteins occludin and claudin-5, the expression of the pericyte-specific markers PDGFR-β (platelet-derived growth factor receptor-β) and α-SMA, and increased the expression of the inflammatory factor MMP9. At the same time, diabetic mice with severe hypoglycemia had significantly reduced cognitive function.Conclusion: Severe hypoglycemia leads to cognitive dysfunction in diabetic mice, and its possible mechanism is related to pericyte dysfunction and BBB destruction.

2021 ◽  
Author(s):  
Lu Lin ◽  
Lishan Huang ◽  
Lijing Wang ◽  
Yubin Wu ◽  
Zhou Chen ◽  
...  

Abstract Background: Severe hypoglycemia can cause cognitive impairment in diabetic patients, but the underlying molecular mechanism remains unclear.Objective: To assess the effect of severe hypoglycemia on cognitive function in diabetic mice to clarify the relationship between the mechanism and dysfunction of pericytes and the blood–brain barrier (BBB).Method: We established type 1 diabetes mellitus in 80 male C57BL/6J mice by intraperitoneal injection of streptozotocin (180 mg/kg). Further abdominal injection of short-acting insulin induced severe hypoglycemia. The mice were divided into normal, diabetes, and diabetic + severe hypoglycemia groups, and their blood glucose and general weight index were examined. Pericyte and BBB morphology and function were detected by histological and western blot analyses, BBB permeability was detected by Evans blue staining, and cognitive function was detected with the Morris water maze.Results: Severe hypoglycemia aggravated the histological damage, BBB damage, brain edema, and pericyte loss in the diabetic mice. It also reduced the expression of the BBB tight junction proteins occludin and claudin-5, the expression of the pericyte-specific markers PDGFR-β (platelet-derived growth factor receptor-β) and α-SMA, and increased the expression of the inflammatory factor MMP9. At the same time, diabetic mice with severe hypoglycemia had significantly reduced cognitive function.Conclusion: Severe hypoglycemia leads to cognitive dysfunction in diabetic mice, and its possible mechanism is related to pericyte dysfunction and BBB destruction.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 5012-5012
Author(s):  
Catherine Gordon ◽  
Alan Nolan ◽  
Wanda Dorsett-Martin ◽  
Gail C. Megason ◽  
Laree Hiser

Abstract Abstract 5012 Background: Current combination chemotherapy regimens are highly effective for treating acute lymphoblastic leukemia (ALL). Unfortunately, vincristine, a component used in current regimens, generally causes significant neuropathy. The neuropathy is usually reversible, but it can have a negative effect on the treatment of ALL by delaying therapy or requiring reduced drug doses. The focus of this research is on exploring pharmaceutical solutions for minimizing the neurotoxicity of vincristine. We selected noscapine as a good candidate for use with vincristine. Noscapine, an FDA-approved antitussive, has no known significant side effects and good oral bioavailability, making it relatively easy to administer to children. Noscapine also demonstrates anticancer properties that are currently being studied, with encouraging preliminary results, in a variety of solid and hematological malignancies. Noscapine has been shown to protect against vincristine-induced demyelination of axons in vitro. In addition, vincristine and noscapine are synergistic in their antiproliferative effects in two leukemic cell lines. While noscapine has been used as a single anticancer agent, no preclinical or clinical studies examining it in combination with vincristine have been reported. Objectives: Methods: Animal studies: C57BL/6 female mice were used to test the safety and efficacy of noscapine as a protective agent against vincristine-induced neuropathy. Behavioral assays (tail-flick and rotarod) were used to assess neuropathy. Control groups receiving only saline, vincristine, or noscapine were compared to groups of mice receiving a combination of vincristine and noscapine. Independent experiments were performed that varied in drug dose and delivery. Vincristine was given as an intraperitoneal injection twice a week with doses varying from 0.1 to 1.5 mg/kg. Noscapine was administered along with vincristine by injection or ad libitum in the drinking water. Data regarding neuropathy and other observable side effects, including weight loss, were collected. Brain and peripheral organs were collected from euthanized mice, quick-frozen in liquid nitrogen and stored at −80 degrees C. Mass spectroscopy experiments: Tissues were homogenized, extracted with methanol and separated using Isolute C2EC SPC columns. Columns were conditioned with methanol, loaded and washed with 5% methanol and eluted with 100% methanol. Separation with liquid chromatography was followed by electrospray ionization and MS/MS analysis using a triple quadrupole-linear ion trap mass spectrometer. Results: The combination of vincristine and noscapine resulted in significant toxic effects including ataxia, seizures, and death in our mouse model. Side effects were not observed in mice given only noscapine. Mice given only vincristine had expected neuropathy as measured by the tail-flick and rotarod assays, but no other significant adverse effects. Noscapine had some protective effect against vincristine-induced neuropathy, but the effect was not statistically significant. Given the adverse neurologic reactions seen, it was hypothesized that noscapine permitted vincristine to cross the blood-brain barrier. Previous studies have shown that noscapine freely crosses the blood-brain barrier, and vincristine is fatal in humans when given intrathecally. Our hypothesis was not supported by analysis of whole brains collected one hour after intraperitoneal injection of vincristine and noscapine (or controls). Mass spectroscopy results showed a decreased uptake of noscapine into brain and peripheral organs in the mice that received noscapine and vincristine in combination. Accumulation of vincristine was unchanged. Conclusion: Noscapine has a significant drug-drug interaction with vincristine in mice. The exact mechanism responsible for this interaction remains undetermined, but several hypotheses including effects on transport mechanisms are currently being investigated. Mass spectroscopy data suggests that vincristine alters the pharmacokinetic or pharmacodynamic properties of noscapine. This may limit the use of noscapine as a neuroprotective agent. Disclosures: No relevant conflicts of interest to declare.


2008 ◽  
Vol 108 (6) ◽  
pp. 1071-1080 ◽  
Author(s):  
Qiang Wang ◽  
Xingchun Gou ◽  
Lize Xiong ◽  
Weilin Jin ◽  
Shaoyang Chen ◽  
...  

Background The Nogo-66 antagonistic peptide (NEP1-40) is a potential candidate for therapeutic intervention of neuronal injury. However, delivery of the proteins across the blood-brain barrier is severely limited by its size and biochemical properties. The current study was designed to evaluate the transducible effects of the trans-activator of transcription (TAT) transduction system for NEP1-40 to cross the blood-brain barrier and to clarify whether intraperitoneal administration of TAT-NEP1-40 could protect cerebral neurons from ischemic injury. Methods Adult male Sprague-Dawley rats were submitted to a 120-min focal ischemia and received an intraperitoneal injection of No-TAT-NEP1-40, TAT-NEP1-40, TAT-beta-galactosidase, or vehicle. The existence of the proteins in the brain was analyzed with immunofluorescence and Western blot techniques at 6 h after injection. Brain ischemic injury was evaluated by neurologic deficit scores, infarction volumes, terminal deoxynucleotidyl transferase-mediated dUDP-biotin nick end labeling staining, and assay of caspase-3 activity. Results Western blot analysis and immunofluorescence staining confirmed the presence of TAT-NEP1-40 protein in the brains 6 h after injection. Intraperitoneal injection of TAT-NEP1-40 could attenuate the numbers of terminal deoxynucleotidyl transferase-mediated dUDP-biotin nick end labeling-positive cells and activated caspase-3 positive cells, and increase the viability of the cells in the ischemic bounder zone, compared with that treated with No-TAT-NEP1-40, TAT-beta-Gal, or vehicle. Furthermore, treatment with TAT-NEP1-40 significantly improved the neurologic outcomes and reduced the size of the infarction in rats. Conclusions The results demonstrate that the TAT-NEP1-40 could be efficiently delivered into the rat brains and improve ischemia-induced neurologic outcomes through attenuating cell apoptosis in ischemic brains.


PLoS ONE ◽  
2018 ◽  
Vol 13 (2) ◽  
pp. e0191909 ◽  
Author(s):  
Jennifer M. Rutkowsky ◽  
Linda L. Lee ◽  
Michelle Puchowicz ◽  
Mari S. Golub ◽  
Douglas E. Befroy ◽  
...  

2017 ◽  
Vol 653 ◽  
pp. 152-158 ◽  
Author(s):  
Ravi K. Sajja ◽  
Shikha Prasad ◽  
Suni Tang ◽  
Mohammad A. Kaisar ◽  
Luca Cucullo

2017 ◽  
Vol 8 ◽  
Author(s):  
Siming Yang ◽  
Changping Gu ◽  
Emiri T. Mandeville ◽  
Yuanlin Dong ◽  
Elga Esposito ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document