Original Article Bumetanide protects focal cerebral ischemia-reperfusion injury in rat

Similar documents
GABA Induced Depolarization: A Tale of Opposing Forces

Chloride s Exciting Role in Neonatal Seizures Suggests Novel Therapeutic Approach

Novel Therapies for Neonatal Seizures December 3, 2010

GABA and Dynamic Chloride Regulation in Health and Disease

SUPPLEMENTARY INFORMATION

Cation chloride cotransporters play an essential role in the

1. GABA, Epilepsy 6:85-92, GABA, Clinical Neuroscience 30 (12): , GABA,, Clinical Neuroscience 30 (7): , 2012.

Effect of low temperatures on BAX and BCL2 proteins in rats with spinal cord ischemia reperfusion injury

When cells are already maximally potentiated LTP is occluded.

Hypertonic saline alleviates cerebral edema by inhibiting microglia-derived TNF-α and IL-1β-induced Na-K-Cl Cotransporter up-regulation

mm Distance (mm)

Supplementary Figure 1. GABA depolarizes the majority of immature neurons in the

Functional Development of Neuronal Networks in Culture -An in vitro Assay System of Developing Brain for Endocrine Disruptors

Supplementary Figure 1

Int J Clin Exp Med 2014;7(11): /ISSN: /IJCEM Qiuxia Wan 1*, Peng Pan 1*, Changqing Xu 2, Wenzhi Li 1

Na + -K + -Cl - Cotransporter in Rat Focal Cerebral Ischemia

Imaging ischemic strokes: Correlating radiological findings with the pathophysiological evolution of an infarct

Effect of anesthetic post-treatment on blood-brain barrier integrity and cerebral edema after transient cerebral ischemia in rats

Original Article Ulinastatin attenuates cerebral ischemia-reperfusion injury in rats

MOLECULAR AND CELLULAR NEUROSCIENCE

Is It Safe to Use a Diuretic to Treat Seizures Early in Development?

Chapter 17 DCX-Expressing Neurons Decrease in the Retrosplenial Cortex after Global Brain Ischemia

GABAergic Input onto CA3 Hippocampal Interneurons Remains Shunting throughout Development

Supporting Online Material for

Blood Supply. Allen Chung, class of 2013

Supplementary Figure 1: Kv7 currents in neonatal CA1 neurons measured with the classic M- current voltage-clamp protocol.

TOXIC AND NUTRITIONAL DISORDER MODULE

Synaptic Integration

Int J Clin Exp Pathol 2015;8(9): /ISSN: /IJCEP

Correlated network activity in the developing hippocampus: role in synaptogenesis

Pathophysiology and treatment of focal cerebral ischemia

Research Development: Bedside to Bench and Back

Original Article Protective effects of LM22A-4 on injured spinal cord nerves

SUPPLEMENTARY INFORMATION

ETIOLOGY AND PATHOGENESIS OF HYPOXIC-ISCHEMIC ENCEPHALOPATHY

CHAPTER 48: NERVOUS SYSTEMS

Complete Recovery of Perfusion Abnormalities in a Cardiac Arrest Patient Treated with Hypothermia: Results of Cerebral Perfusion MR Imaging

Homeostasis Practice Quiz 20 Questions SBI 4UI

Seizure: the clinical manifestation of an abnormal and excessive excitation and synchronization of a population of cortical

Deposited on: 25 July 2011

Implication of aquaporins in ischemic stroke. New target?

Expression of acid base transporters in the kidney collecting duct in Slc2a7 -/-

Immunostaining was performed on tumor biopsy samples arranged in a tissue-microarray format or on

211MDS Pain theories

Repeated intra-articular injections of acidic saline produce long-lasting joint pain and. widespread hyperalgesia

Comparison of Young and Old Cardiac Telocytes Using Atomic Force Microscopy

Microglia is an active player in how glibenclamide improves stroke outcome

Cl uptake promoting depolarizing GABA actions in immature rat neocortical neurones is mediated by NKCC1

Preconditioning the Brain for Stroke Prevention

Review Article Spatial and Temporal Dynamics in the Ionic Driving Force for GABA A Receptors

Structural basis for the role of inhibition in facilitating adult brain plasticity

Mingjin Jiang 1, Jing Li 1, Qiuxian Peng 1,2, Yi Liu 1, Wei Liu 1, Chaohua Luo 1, Ju Peng 1, Junkui Li 1, Ken Kin Lam Yung 2* and Zhixian Mo 1*

Yanbing He 1, 2, Shiyuan Xu 1, Junjie Huang 2, Qingjuan Gong 2. NEURAL REGENERATION RESEARCH May 2014,Volume 9,Issue 10

How Nicotinic Signaling Shapes Neural Networks

Evaluation of directed and random motility in microslides Assessment of leukocyte adhesion in flow chambers

Experimental Assessment of Infarct Lesion Growth in Mice using Time-Resolved T2* MR Image Sequences

PARA210 SUMMARY Hyperglycaemia (DKA & HHS) Brain & Nervous System Anatomy & Physiology Degenerative Neurological Disorders

Supplementary Material

Supplementary Table I Blood pressure and heart rate measurements pre- and post-stroke

All questions below pertain to mandatory material: all slides, and mandatory homework (if any).

The Chloride Transporter Na -K -Cl Cotransporter Isoform-1 Contributes to Intracellular Chloride Increases after In Vitro Ischemia

Original Article Activation of STAT3 is involved in neuronal apoptosis in focal cerebral ischemia/reperfusion rats via Bcl 2/Fas pathway

Cell-attached recordings of responses evoked by photorelease of GABA in the immature cortical neurons

Submitted to the University of Adelaide for the degree of. Doctor of Science. Robert Vink, BSc (Hons), PhD

Neural stem cells and the neurobiology of ageing. Chen Siyun 1, Dawe G.S. 2

Chapter 5 subtitles GABAergic synaptic transmission

Changes in Extracellular Ionic Composition q

Neuroprotective effects of scutellarin on rat neuronal damage induced by cerebral ischemia/reperfusion

Neurological Assessment Scores in Rabbit Embolic Stroke Models

N-methyl-D-aspartate Receptor Mediates X-irradiation-induced Drebrin Decrease in Hippocampus

Part 11: Mechanisms of Learning

Guided Reading Activities

Differences in Cortical versus Subcortical GABAergic Signaling: A Candidate Mechanism of Electroclinical Uncoupling of Neonatal Seizures

Biomedical Research 2018; 29 (21): ISSN X

The cation-chloride cotransporter NKCC1 promotes sharp waves in the neonatal rat hippocampus

Charlie Taylor, PhD CpTaylor Consulting Chelsea, MI, USA

biological psychology, p. 40 The study of the nervous system, especially the brain. neuroscience, p. 40

B-cell. Astrocyte SCI SCI. T-cell

Current status of temperature management in the neuro-icu

Neuroprotective properties of epoetin alfa

Rapamycin Suppresses Astrocytic and Microglial Activation and Reduced Development of Neuropathic Pain after Spinal Cord Injury in Mice.

Nicolas Bianchi M.D. May 15th, 2012

SWISS SOCIETY OF NEONATOLOGY. Neonatal cerebral infarction

Implantable Microelectronic Devices

Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting. protein3) regulate autophagy and mitophagy in renal tubular cells in. acute kidney injury

Downregulation of angiotensin type 1 receptor and nuclear factor-κb. by sirtuin 1 contributes to renoprotection in unilateral ureteral

Original Article Effect of activation of the Ca 2+ -permeable acid-sensing ion channel 1a on focal cerebral ischemia in diabetic rats

Name: Period: Chapter 2 Reading Guide The Biology of Mind

Nature Neuroscience: doi: /nn Supplementary Figure 1

Ube3a is required for experience-dependent maturation of the neocortex

What Cell Make Up the Brain and Spinal Cord

SCIRF Award #2016 I-03 PI: Azizul Haque, PhD Grant Title: Neuron-specific Enolase and SCI

Supplementary Appendix

Central nervous system (CNS): brain and spinal cord Collections of cell body and dendrites (grey matter) are called nuclei/nucleus Nucleus can also

In the name of GOD. Animal models of cardiovascular diseases: myocardial infarction & hypertension

Targeting glutamatemediated. in the brain using NYX-104. Gary Housley, Ph.D. Industry Partner: Noxopharm Pty Ltd

Bumetanide, an NKCC1 Antagonist, Does Not Prevent Formation of Epileptogenic Focus but Blocks Epileptic Focus Seizures in Immature Rat Hippocampus

Mechanisms of GABAergic Homeostatic Plasticity

File name: Supplementary Information Description: Supplementary Figures, Supplementary Table and Supplementary References

Relaxation responses of aortic rings from salt-loaded high calcium fed rats to potassium chloride, calcium chloride and magnesium sulphate

Transcription:

Int J Clin Exp Pathol 2014;7(4):1487-1494 www.ijcep.com /ISSN:1936-2625/IJCEP1401050 Original Article Bumetanide protects focal cerebral ischemia-reperfusion injury in rat Genbao Wang 1,2*, Huansen Huang 1, Yanbing He 1*, Lin Ruan 1*, Junjie Huang 1 1 Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China; 2 Department of Anesthesiology, Affiliated Zhong San Chinese Hospital of Guang Zhou Chinese University, Zhong San 528400, China. * Equal contributors. Received January 16, 2014; Accepted February 27, 2014; Epub March 15, 2014; Published April 1, 2014 Abstract: Objective: Bumetanide has been reported to attenuate ischemia-evoked cerebral edema. However, whether bumetanide can protect cerebral ischemia-reperfusion injury (IRI) in vivo is unclear. In the present study, we aim to determine whether intravenously injection bumetanide can attenuate cerebral IRI and if its protection effect might be related to the modification of cerebral NKCC1 and KCC2 protein expression. Methods: Focal cerebral ischemia was induced by occluding the right middle cerebral artery (MCAO) for 2-h, followed by 3-h, 24-h or 48-h of reperfusion respectively. Brain edema, neurological deficits, and infarction volume were determined by (wet weights - dry weights)/dry weights 100, 5-point neurological function score evaluation system, and TTC staining, respectively. The expression levels of NKCC1 and KCC2 were determined by immunohistochemical staining. Results: Reperfusion increased brain edema, neurological deficits, and infarction volume. Bumetanide decreased brain edema, attenuated the neurological defects and reduced post-ischemic cerebral infarction. Cerebral ischemia-reperfusion injury increased NKCC1 expression level and decreased KCC2 expression level. Interestingly, bumetanide down-regulated the NKCC1 protein expression level without changing the KCC2 protein expression level in rat brain cortex. Conclusion: These results suggest that bumetanide protects focal cerebral ischemia-reperfusion injury in rat, which might through the inhibition of NKCC1. Keywords: Bumetanide, Na + -K + -Cl - -cotransporter 1 (NKCC1), Na + -K + -cotransporter 2 (KCC2), cerebral ischemiareperfusion injury, neuroprotection Introduction Imbalance of ion homeostasis is a crucial mechanism leading to ischemia-induced cell damage [1]. Cation chloride co-transporters play an essential role in the regulation of neuronal chloride homeostasis [1-3]. The Na + -K + -Cl - - co-transporter 1 (NKCC1) and Na + -K + -co-transporter 2 (KCC2) belong to the cation-cl - cotransporter family, which mediates the coupled movement of Na + and/or K + with Cl - across the plasma membrane of cells under physiologic conditions [1-3]. NKCC1 has a broad tissue distribution, whereas KCC2 is found mainly in the nervous system [1-3]. In detail, KCC2 is predominately expressed in mature neurons in the cortex, cerebellum, and the dorsal horn of the spinal cord, while NKCC1 is expressed at a high level in immature neurons [4, 5]. NKCC1 is important for the maintenance of intracellular Cl - in neurons and contributes to GABA-mediated depolarization in immature neurons [6, 7] while KCC2 is the main chloride extruder in the adult nervous system [8]. In ischemic conditions, particularly after oxygen glucose deprivation in hippocampal slices [9] and in in vivo models of global ischemia [10], KCC2 is generally down-regulated while NKCC1 is up-regulated. In vitro studies have shown that bumetanide, a member of sulfamoyl benzoic acid loop diuretic family which exerts its diuretic action by blockade of the Na + -K + -2Cl cotransporter, significantly reduces the oxygen-glucose-deprivation and glutamate-mediated neuronal excitotoxicity and apoptosis by inhibiting NKCC1. Moreover, bumetanide has also been reported to attenuate ischemia-evoked cerebral edema [11]. However, whether bumetanide can protect cerebral ischemia-reperfusion injury (IRI) in vivo is still unclear. In the present study, we aim to determine whether intravenously injection bumetanide

Figure 1. Bumetanide reduces brain edema after focal cerebral ischemia. Edema in bumetanide group (B+I/R) and saline group (N+I/R) was significantly higher than that in the sham group. Bumetanide significantly reduces brain edema after focal cerebral ischemia. Data are expressed as means±s.d. Bumetanide group and saline group rats, n=6; sham group rats, n=3; p<0.05 bumetanide and saline versus sham group. # p<0.05 versus bumetanide or saline group. can attenuate cerebral IRI and if its protection effect might be related to the modification of cerebral NKCC1 and KCC2 protein expression. Materials and methods This study was conducted in accordance to the guidelines for the care and use of animals in research, and under the protocols approved by the Guangzhou University of Connecticut Animal Care and Use Committee. Animal Adult male rats, weighing 250~320 g (8~10 weeks), were purchased from Guangdong Province Animals Center (Guangzhou, China). Rats were anesthetized with intraperitoneal injection of 10% chloral hydrate (3.5 ml/kg body weight). Rectal temperatures were monitored and maintained at 37 C±0.5 C with a heating blanket and a heating lamp. Rats were randomly divided into 3 groups (n=45 per group) including sham operation group (S), saline plus ischemia reperfusion group (N+I/R group), and bumetanide plus ischemia reperfusion group (B+I/R group). Rats in the B+I/R group and N+I/R group were respectively received injection of bumetanide (30 mg/kg) [12] or the same volume of saline through tail vein 10 min before cerebral ischemia inducing. All rats except those in the sham group underwent 2-h right middle cerebral artery occlusion (MCAO) as previously reported [13] followed by 3-h, 24-h or 48-h reperfusion respectively. Edema measurement Rats were decapitated under deep anesthesia with 10% chloral hydrate at 3-h, 24-h and 48-h of reperfusion. The ipsilateral and contralateral hemispheres were dissected and the wet weight of the tissue was determined. The tissues were dried at 120 C for 24 hours. The percent cerebral water was determined as (wet weights - dry weights)/ dry weights 100. Neurological evaluation Rats were examined for neurological deficits 3-h, 24-h or 48-h after MCAO by two investigators who were blinded to this study using a 5-point neurological function score [13]: 0, no deficit; 1, failure to extend right forepaw fully; 2, circling to the right; 3, failing to the right; 4, no spontaneous walking with a depressed level of consciousness. Only those rats showed no or incomplete forelimb placing with rotational asymmetry at 3-h, 24-h or 48-h after MCAO were included in the subsequent analysis [14]. TTC staining After 3-h, 24-h or 48-h of reperfusion, rat brains were removed and frozen at -80 C for 5 min. Two millimeter coronal slices were made with a rodent brain matrix. The sections were stained for 20 min at 37 C with 2% 2,3,5-triphenyltetrazolium chloride monohydrate (TTC, Sigma, USA) and then were fixed with 4% formalin. Infarction volume was calculated as previous reported [15]. Briefly, the sections were scanned, and the infarction area in each section was calculated by subtracting the noninfarct area of the ipsilateral side from the area of 1488 Int J Clin Exp Pathol 2014;7(4):1487-1494

Figure 2. Bumetanide decreases the volume of cerebral infarction 3-h post-reperfusion. A. Representative results of TTC staining. B. Bumetanide decreases the volume of cerebral infarction 3-h post-reperfusion but not 24-h, and 48-h post-reperfusion. Data are expressed as means±s.d. n=6. p<0.05 saline versus bumtanide group rats. the contralateral side with Image-J analysis software. Infarction areas on each section were summed and multiplied by section thickness to give the total infarction volume. Immunohistochemical staining Rat brains were removed, and postfixed in 4% paraformaldehyde overnight at 4 C, and cryoprotected with 30% sucrose in PBS. Brains were cut into coronal sections (30 um) on a freezing microtome (Leica SM 2000 R; Leica, Nussloch, Germany). Coronal sections (0.2 mm anterior to bregma) were selected and processed for immuno-histochemistry. The sections from each rat were used and the freefloating method was used for avidin-biotin-peroxidase labeling. Coronal sections were rinsed with PBS and treated with 0.3% H 2 O 2 (v/v) in PBS for 10 min at room temperature to quench endogenous peroxidase. After washing with PBS, sections were incubated with a blocking solution (10% normal goat serum, 0.3% Triton X-100 in PBS) for 30 min at 37 C. The slices were probed respectively with primary anti- NKCC1 polyclonal antibody (1:200; Santa Cruz, U.S.A.) and anti-kcc2 polyclonal antibody 1489 Int J Clin Exp Pathol 2014;7(4):1487-1494

Figure 3. Bumetanide reduces neurological deficit scores 24-h and 48-h postreperfusion. Data are expressed as means±s.d. n=6. p<0.01 versus sham group rats; # p<0.05 bumetanide versus saline group rats, p<0.05 3-h versus 24-h and 48-h reperfusion in bumetanide group rats. (1:200; Santa Cruz, U.S.A.) for 1 hour at 37 C, followed by overnight incubation at 4 C. After rinsing, sections were incubated with goat antirabbit IgG (1:200; Sigma, U.S.A.) for 1 hour at 37 C. Color reaction was developed with 0.05% diaminobenzidime tetrachloride and 0.03% H 2 O 2 in PBS (ph 7.4). Slides were visualized using a Leica SP5 confocal microscope at 40x magnification using standard procedure. All imaging was performed with group identity blinded where at least 10 random images were obtained from each slide or group. Statistical analysis All values are presented as means±sd. Data shown were analyzed for significance using analysis of either the nonparametric Mann- Whitney test or ANOVA by SPSS13.0 software. If a significant difference was observed, Bonferroni s post-hoc test was conducted to identify groups with significant differences. P-values that were less than 0.05 were considered to be statistically significant. Results Bumetanide reduces brain edema after focal cerebral ischemia To determine whether bumetanide reduces edema formation resulting from MCAO, bumetanide or vehicle was administered intravenously 10 minutes before initiation of MCAO (or sham MCAO). Bumetanide reduced the increase of water content in ischemic brains by 33.3%, 49.7% and 45.8% respectively at 3-h, 24-h, and 48-h post-reperfusion (P<0.05) (Figure 1). Bumetanide decreases the volume of cerebral infarction 3-h post-reperfusion Figure 2 shows the representative results of TTC staining in rats subjected to N+I/R compared with rats subjected to B+I/R. The average total infarction volume in saline group was 231.5±40.6 mm 3 while the average total infarction vol- ume was reduced to 193.6±30.7 mm 3 after 3-h reperfusion in the bumetanide group (p<0.05). However, the average total infarction volume was not significantly changed in the bumetanide group at 24-h, and 48-h post-reperfusion (p>0.05). Bumetanide reduces neurological deficit scores 24-h and 48-h post-reperfusion In the sham group, no rats have symptoms of neurological deficits while the neurological deficits were obvious in the saline and bumetanide group. Moreover, the neurological deficits was significantly reduced in the bumetanide group rats 24-h and 48-h post-reperfusion compared to that in the saline group (P<0.05) (Figure 3). Bumetanide down-regulates the NKCC1 protein expression level in rat brain cortex It has been reported that the expression level of NKCC1 was increased in cortex after 24-h reperfusion following 2-h ischemia [16]. As shown in Figure 4, an enhanced immunostaining of the NKCC1 in neurons scattered throughout ischemic cortex 3-h, 24-h, 48-h post-reperfusion. Compared with saline groups, pretreatment with bumeitanide significantly reduced the protein expression level of NKCC1. Bumetanide does not change the KCC2 protein expression level in rat brain cortex As shown in Figure 5, KCC2 protein was expressed in the plasma membrane and dendrites of neurons. The expression level of KCC2 1490 Int J Clin Exp Pathol 2014;7(4):1487-1494

Figure 4. Bumetanide down-regulates the NKCC1 protein expression level in rat brain cortex. A-C. Respectively were sham group rats at 3h, 24h, 48h. D-F were saline group, G-I were bumetanide groups. J. Bumetanide down-regulates the NKCC1 protein expression level in rat brain cortex. Data are expressed as means±s.d. n=6. p<0.05 versus sham group rats; p<0.05 bumetanide versus saline group rats. is significantly down-regulated in the ischemic hemisphere 3-h, 24-h, and 48-h post-reperfusion. However, bumetanide does not change the KCC2 protein expression level in rat brain cortex (Figure 5). Discussion The major findings of the present study are as follows. Firstly, bumetanide reduces brain edema after focal cerebral ischemia. Secondly, 1491 Int J Clin Exp Pathol 2014;7(4):1487-1494

Figure 5. Bumetanide does not change the KCC2 protein expression level in rat brain cortex. A-C were the S group 3h, 24h, 48h post-reperfusion. D-F were N+I/R group 3h, 24h, 48h post-reperfusion and G-I were B+I/R group 3h, 24h, 48h post-reperfusion. J. Bumetanide does not change the KCC2 protein expression level in rat brain cortex. Data are expressed as means±s.d. n=6. p<0.05 versus sham group rats. bumetanide decreases the volume of cerebral infarction 3-h post-reperfusion. Thirdly, bumetanide reduces neurological deficit scores 24-h and 48-h post-reperfusion. Finally, bumetanide down-regulates the NKCC1 protein expression level without changing the KCC2 protein expression level in rat brain cortex. NKCC1 transports Na +, K +, and Cl - into cells under both physiological and pathophysiologi- 1492 Int J Clin Exp Pathol 2014;7(4):1487-1494

cal conditions with a stoichiometry of 1Na + : 1K + :2Cl - and it can be inhibited by either bumetanide or furosemide. Bumetanide can compete with Cl - for the second Cl - binding site and thus inhibits NKCC1 function [17-19]. After focal ischemia, extracellular K + increases, glutamate is released, intracellular Ca 2+ increases, excessive Na + and Cl - accumulates, resulting in the enhancement of NKCC1 activity [20]. Pathologic activation of NKCC1 seems to be an important mechanism of cerebral edema following ischemia. However, in certain pathological conditions, such as ischemia, hypoxia and trauma, NKCC1 expression was increased which led to cell swelling. NKCC1 has been suggested to play a role in K + uptake and swelling in astrocytes [21]. In this study, we found that bumetanide reduced brain edema, decreases the volume of cerebral infarction, and reduces neurological deficit scores after focal cerebral ischemia that might be related to its inhibition effects in NKCC1. KCC2 abundantly expresses in the adult mammalian nervous system, and it plays an important role in maintaining the low Cl environment of mature neurons. In addition, KCC2 participates in the regulation of ion balance, nerve growth and maturation, synaptic development and neuronal plasticity [22]. A decrease of KCC2 expression was previously described under ischemic conditions [9]. In the present study, we also demonstrated that KCC2 expression level was down-regulated after ischemiareperfusion in rat cerebral cortex, which is consistent with the previous report [23]. However, we did not observe significant changes in KCC2 expression following a single intravenous bumetanide (30 mg/kg) injection, which might be due to the fact that low concentrations of bumetanide (2~10 μm) specifically inhibits NKCC1 but does not affect KCC2 [24, 25]. Taken together, the present study suggests that bumetanide might confer its neuroprotection effect mainly by inhibiting NKCC1. In summary, cerebral ischemia-reperfusion injury increases NKCC1 expression level and decreases KCC2 expression level. Bumetanide attenuates the neurological defects and reduces post-ischemic cerebral infarction, which might through the inhibition of NKCC1. Acknowledgements This work was supported by the grants from Guang Zhou Medical University (HS. Huang). Disclosure of conflict of interest None to declare. Address correspondence to: Dr. Huansen Huang, Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, 250 Chang Gangdong Road, Guangzhou 510260, China. Tel: 0086-20-34152535; Fax: 0086-20-34152003; E-mail: huanghs5480_gz@163.com References [1] Haas M, Forbush B 3rd. The Na-K-Cl cotransporter of secretory epithelia. Ann Rev Physiol 2000; 62: 515-534. [2] Russell JM. Sodium-potassium-chloride cotransport. Physiol Rev 2000; 80: 211-276. [3] Blaesse P, Airaksinen MS, Rivera C, Kaila K. Cation-Chloride Cotransporters and Neuronal Function. Neuron 2009; 61: 820-838. [4] Hebert SC, Mount DB, Gamba G. Molecular physiology of cation-coupled Cl- cotransport: the SLC12 family. Pflugers Areh 2004; 447: 580-593. [5] Khirug S, Yamada J, Afzalov R, Voipio J, Khiroug L, Kaila K. GABAergic depolarization of the axon initial segment in cortical principal neurons is caused by the Na-K-2Cl cotransporter NKCC1. J Neurosci 2008; 28: 4635-4639. [6] Yamada J, Okabe A, Toyoda H, Kilb W, Luhmann HJ, Fukuda A. Cl - uptake promoting depolarizing GABA actions in immature rat neocortical neurones is mediated by NKCC1. J Physiol 2004; 557: 829-841. [7] Kahle KT, Simard JM, Staley KJ, Nahed BV, Jones PS, Sun D. Molecular mechanisms of ischemic cerebral edema: role of electroneutral ion transport. Physiology (Bethesda) 2009; 24: 257-265. [8] Rivera C, Voipio J, Payne JA, Ruusuvuori E, Lahtinen H, Lamsa K, Pirvola U, Saarma M, Kaila K. The K C co-transporter kcc2 rendersgaba hyperpolarizing during neuronal maturation. Nature 1999; 397: 251-255. [9] Galeffi F, Sah R, Pond BB, George A, Schwartz- Bloom RD. Changes in intracellular chloride after oxygen-glucose deprivation of the adult hippocampal slice: Effect of diazepam. J Neurosci 2004; 24: 4478-4488. [10] Papp E, Rivera C, Kaila K, Freund TF. Relationship between neuronal vulnerability and potassium-chloride cotransporter 2 immunoreactivity in hippocampus following transient forebrain ischemia. Neurosci 2008; 154: 677-689. [11] Lu KT, Wu CY, Cheng NC, Wo YY, Yang JT, Yen HH, Yang YL. Inhibition of the Na+-K+-2Cl--cotransporter in choroid plexus attenuatestraumatic brain injury-induced brain edema and 1493 Int J Clin Exp Pathol 2014;7(4):1487-1494

neuronal damage. Eur J Pharmacol 2006; 548: 99-105. [12] Liu Y, Shangguan Y, Barks JD, Silverstein FS. Bumetanide augments the neuroprotective efficacy of phenobarbital plus hypothermia in a neonatal hypoxia-ischemia model. Pediatr Res 2012; 71: 559-565. [13] Longa EZ, Weinstein PR, Carlson S, Cummins R. Reversible middle cerebral artery occlusion without craniectomy in rats. Stroke 1989; 20: 84-91. [14] Thored P, Wood J, Arvidsson A, Cammenga J, Kokaia Z, Lindvall O. Long-term neuroblast migration along blood vessels in an area with transient angiogenesis and increased vascularization after stroke. Stroke 2007; 11: 3032-3039. [15] Swanson RA, Morton MT, Tsao-Wu G, Savalos RA, Davidson C, Sharp FR. A semiautomated method for measuring brain infarct volume. J Cereb Blood Flow Metab 1990; 10: 290-293. [16] Yan Y, Dempsey RJ, Sun D. Na+-K+-Cl- contransporter in rat focal cerebral ischemia. J Cereb Blood Flow Metab 2001; 21: 711-721. [17] Yan Y, Dempsey RJ, Flemmer A, Forbush B, Sun D. Inhibition of Na+-K+-Cl- cotransporter during focal cerebral ischemia decreases edema and neuronal damage. Brain Res 2003; 961: 22-31. [18] Chen H, Luo J, Kintner DB, Shull GE, Sun D. Na+-dependent chloride transporter (NKCC1)- null mice exhibit less gray and white matter damage after focal cerebral ischemia. J Cereb Blood Flow Metab 2005; 25: 54-66. [19] Forbush B 3rd, Palfrey HC. [3H]bumetanide binding to membranes isolated from dog kidney outer medulla. Relationship to the Na,K, Cl co-transport system. J Biol Chem 1983; 258: 11787-11792. [20] O Donnell ME, Tran L, Lam TI, Liu XB, Anderson SE. Bumetanide inhibition of the blood-brain barrier Na-K-Cl cotransporter reduces edema formation in the rat middle cerebral artery occlusion model of stroke. J Cereb Blood Flow Metab 2004; 24: 1046-1056. [21] Su G, Haworth RA, Dempsey RJ, Sun D. Regulation of Na(+)-K(+)-Cl(-) cotransporter in primary astrocytes by dibutyryl camp and high [K(+)] (o). Am J Physiol 2000; 279: C1710-21. [22] Payne JA, Rivera C, Voipio J, Kaila K. Cationchloride co-transporters in neuronal communication, development and trauma. Trends Neurosci 2003; 26: 199-206. [23] Jaenisch N, Witte OW, Frahm C. Downregulation of potassium chloride cotransporter KCC2 after transient focal cerebral ischemia. Stroke 2010; 41: e151-e159. [24] Gillen CM, Brill S, Payne JA, Forbush B 3rd. Molecular cloning and funetional expression of the K-Cl cotransporter from rabbit, rat, and human. A new member of the cation-chloride cotransporter family. J Biol Chem 1996; 271: 16237-1624. [25] Tyzio R, Cossart R, Khalilov I, Minlebaev M, Hubner CA, Represa A, Ben-Ari Y, Khazipov R. Maternal oxytocin triggers a transient inhibitory switch in gaba signaling in the fetal brain during delivery. Science 2006; 314: 1788-1792. 1494 Int J Clin Exp Pathol 2014;7(4):1487-1494