Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc (Bayl Univ Med Cent) ; 33(2): 199-204, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32313461

ABSTRACT

Blood-brain barrier breakdown and associated vascular hyperpermeability leads to vasogenic edema in traumatic brain injury (TBI). Tight junctions maintain blood-brain barrier integrity; their disruption in TBI holds significant promise for diagnosis and treatment. A controlled cortical impactor was used for TBI in mouse studies. Blood was collected 1 h after injury and sent for antibody microarray analysis. Twenty human subjects with radiographic evidence of TBI were enrolled and blood collected within 48 h of admission. Control subjects were individuals with nontrauma diagnoses. The subjects were matched by age and gender. Enzyme-linked immunosorbent assays were performed on each TBI and control sample for tight junction-associated proteins (TJPs), inflammatory markers, and S100ß. Plasma was used to conduct in vitro monolayer permeability studies with human brain endothelial cells. S100ß and the TJP occludin were significantly elevated in TBI plasma in both the murine and human studies. Monolayer permeability studies showed increased hyperpermeability in TBI groups. Plasma from TBI subjects increases microvascular hyperpermeability in vitro. TJPs in the blood may be a potential biomarker for TBI.

2.
Methods Mol Biol ; 1717: 37-52, 2018.
Article in English | MEDLINE | ID: mdl-29468582

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death and disability worldwide. It is a silently growing epidemic with multifaceted pathogenesis, and current standards of treatments aim to target only the symptoms of the primary injury, while there is a tremendous need to explore interventions that can halt the progression of the secondary injuries. The use of a reliable animal model to study and understand the various aspects the pathobiology of TBI is extremely important in therapeutic drug development against TBI-associated complications. The controlled cortical impact (CCI) model of TBI described here, uses a mechanical impactor to inflict a mechanical injury into the mouse brain. This method is a reliable and reproducible approach to inflict mild, moderate or severe injuries to the animal for studying TBI-associated blood-brain barrier (BBB) dysfunctions, neuronal injuries, brain edema, neurobehavioral changes, etc. The present method describes how the CCI model could be utilized for determining the BBB dysfunction and hyperpermeability associated with TBI. Blood-brain barrier disruption is a hallmark feature of the secondary injury that occur following TBI, frequently associated with leakage of fluid and proteins into the extravascular space leading to vasogenic edema and elevation of intracranial pressure. The method described here focuses on the development of a CCI-based mouse model of TBI followed by the evaluation of BBB integrity and permeability by intravital microscopy as well as Evans Blue extravasation assay.


Subject(s)
Blood-Brain Barrier , Brain Injuries, Traumatic , Intracranial Hypertension , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/physiopathology , Disease Models, Animal , Humans , Intracranial Hypertension/metabolism , Intracranial Hypertension/pathology , Intracranial Hypertension/physiopathology , Mice
3.
J Biol Chem ; 291(53): 26958-26969, 2016 12 30.
Article in English | MEDLINE | ID: mdl-27875293

ABSTRACT

Blood-brain barrier (BBB) breakdown and the associated microvascular hyperpermeability followed by brain edema are hallmark features of several brain pathologies, including traumatic brain injuries (TBI). Recent studies indicate that pro-inflammatory cytokine interleukin-1ß (IL-1ß) that is up-regulated following traumatic injuries also promotes BBB dysfunction and hyperpermeability, but the underlying mechanisms are not clearly known. The objective of this study was to determine the role of calpains in mediating BBB dysfunction and hyperpermeability and to test the effect of calpain inhibition on the BBB following traumatic insults to the brain. In these studies, rat brain microvascular endothelial cell monolayers exposed to calpain inhibitors (calpain inhibitor III and calpastatin) or transfected with calpain-1 siRNA demonstrated attenuation of IL-1ß-induced monolayer hyperpermeability. Calpain inhibition led to protection against IL-1ß-induced loss of zonula occludens-1 (ZO-1) at the tight junctions and alterations in F-actin cytoskeletal assembly. IL-1ß treatment had no effect on ZO-1 gene (tjp1) or protein expression. Calpain inhibition via calpain inhibitor III and calpastatin decreased IL-1ß-induced calpain activity significantly (p < 0.05). IL-1ß had no detectable effect on intracellular calcium mobilization or endothelial cell viability. Furthermore, calpain inhibition preserved BBB integrity/permeability in a mouse controlled cortical impact model of TBI when studied using Evans blue assay and intravital microscopy. These studies demonstrate that calpain-1 acts as a mediator of IL-1ß-induced loss of BBB integrity and permeability by altering tight junction integrity, promoting the displacement of ZO-1, and disorganization of cytoskeletal assembly. IL-1ß-mediated alterations in permeability are neither due to the changes in ZO-1 expression nor cell viability. Calpain inhibition has beneficial effects against TBI-induced BBB hyperpermeability.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Injuries, Traumatic/drug therapy , Calpain/antagonists & inhibitors , Cell Membrane Permeability/drug effects , Endothelium, Vascular/drug effects , Glycoproteins/pharmacology , Animals , Brain Injuries, Traumatic/etiology , Brain Injuries, Traumatic/metabolism , Calpain/genetics , Calpain/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Interleukin-1beta/toxicity , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Rats
4.
PLoS One ; 11(5): e0154427, 2016.
Article in English | MEDLINE | ID: mdl-27152411

ABSTRACT

Microvascular hyperpermeability that occurs at the level of the blood-brain barrier (BBB) often leads to vasogenic brain edema and elevated intracranial pressure following traumatic brain injury (TBI). At a cellular level, tight junction proteins (TJPs) between neighboring endothelial cells maintain the integrity of the BBB via TJ associated proteins particularly, zonula occludens-1 (ZO-1) that binds to the transmembrane TJPs and actin cytoskeleton intracellularly. The pro-inflammatory cytokine, interleukin-1ß (IL-1ß) as well as the proteolytic enzymes, matrix metalloproteinase-9 (MMP-9) are key mediators of trauma-associated brain edema. Recent studies indicate that melatonin a pineal hormone directly binds to MMP-9 and also might act as its endogenous inhibitor. We hypothesized that melatonin treatment will provide protection against TBI-induced BBB hyperpermeability via MMP-9 inhibition. Rat brain microvascular endothelial cells grown as monolayers were used as an in vitro model of the BBB and a mouse model of TBI using a controlled cortical impactor was used for all in vivo studies. IL-1ß (10 ng/mL; 2 hours)-induced endothelial monolayer hyperpermeability was significantly attenuated by melatonin (10 µg/mL; 1 hour), GM6001 (broad spectrum MMP inhibitor; 10 µM; 1 hour), MMP-9 inhibitor-1 (MMP-9 specific inhibitor; 5 nM; 1 hour) or MMP-9 siRNA transfection (48 hours) in vitro. Melatonin and MMP-9 inhibitor-1 pretreatment attenuated IL-1ß-induced MMP-9 activity, loss of ZO-1 junctional integrity and f-actin stress fiber formation. IL-1ß treatment neither affected ZO-1 protein or mRNA expression or cell viability. Acute melatonin treatment attenuated BBB hyperpermeability in a mouse controlled cortical impact model of TBI in vivo. In conclusion, one of the protective effects of melatonin against BBB hyperpermeability occurs due to enhanced BBB integrity via MMP-9 inhibition. In addition, acute melatonin treatment provides protection against BBB hyperpermeability in a mouse model of TBI indicating its potential as a therapeutic agent for brain edema when established in humans.


Subject(s)
Blood-Brain Barrier , Matrix Metalloproteinase 9/drug effects , Melatonin/physiology , Protease Inhibitors/pharmacology , Animals , Blood-Brain Barrier/drug effects , Cells, Cultured , Gene Knockdown Techniques , Humans , Interleukin-1beta/therapeutic use , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Rats
5.
Am J Surg ; 211(1): 197-205, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26601648

ABSTRACT

BACKGROUND: Burns induce microvascular hyperpermeability. We hypothesize that this occurs partly through an imbalance between matrix metalloproteinases (MMPs) and endogenous MMP inhibitors such as tissue inhibitors of metalloproteinases (TIMPs), and that such derangements can be attenuated with the use of TIMP-2. METHOD: Rats underwent either sham or burn: serum and tissue were collected. Western blot was used to examine MMP-9 and TIMP-2 levels and MMP activity was assayed from lung tissue. Rat lung microvascular endothelial cells were used to assess monolayer permeability and evaluate the adherens junction proteins ß-catenin, vascular endothelial cadherin and filamentous actin after exposure to burn serum ± TIMP-2. RESULTS: Lung tissue from burn animals showed increased MMP activity, decreased levels of TIMP-2, and no difference in levels of active MMP-9 in burn vs control groups. Burn serum increased monolayer permeability, damaged adherens junction proteins, and incited actin stress fiber formation; TIMP-2 attenuated these derangements. CONCLUSIONS: Burns may lower TIMP-2 levels and increase MMP activity and that TIMP-2 application in vitro may attenuate burn-induced hyperpermeability and decreases damage to endothelial structural proteins. These links warrant further investigation.


Subject(s)
Burns/enzymology , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Matrix Metalloproteinase 9/metabolism , Microvessels/drug effects , Protective Agents/pharmacology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Animals , Biomarkers/metabolism , Blotting, Western , Burns/drug therapy , Burns/physiopathology , Capillary Permeability/physiology , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/physiology , Lung/drug effects , Lung/enzymology , Lung/physiopathology , Male , Microvessels/enzymology , Microvessels/physiopathology , Protective Agents/metabolism , Protective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-2/therapeutic use
6.
Shock ; 45(6): 626-33, 2016 06.
Article in English | MEDLINE | ID: mdl-26717103

ABSTRACT

INTRODUCTION: Lipopolysaccharide (LPS) is known to induce vascular derangements. The pathophysiology involved therein is unknown, but matrix metalloproteinases (MMPs) may be an important mediator. We hypothesized that in vitro LPS provokes vascular permeability, damages endothelial structural proteins, and increases MMP activity; that in vivo LPS increases permeability and fluid requirements; and that the MMP inhibitor doxycycline mitigates such changes. METHODS: Rat lung microvascular endothelial cells were divided into four groups: control, LPS, LPS plus doxycycline, and doxycycline. Permeability, structural proteins ß-catenin and Filamentous-actin, and MMP-9 activity were examined. Sprauge Dawley rats were divided into sham, IV LPS, and IV LPS plus IV doxycycline groups. Mesenteric postcapillary venules were observed. Blood pressure was measured as animals were resuscitated and fluid requirements were compared. Statistical analysis was conducted using Student's t-test and ANOVA. RESULTS: In vitro LPS increased permeability, damaged adherens junctions, induced actin stress fiber formation, and increased MMP-9 enzyme activity. In vivo, IV LPS administration induced vascular permeability. During resuscitation, significantly more fluid was necessary to maintain normotension in the IV LPS group. Doxycycline mitigated all derangements observed. CONCLUSIONS: We conclude that LPS increases permeability, damages structural proteins, and increases MMP-9 activity in endothelial cells. Additionally, endotoxemia induces hyperpermeability and increases the amount of IV fluid required to maintain normotension in vivo. Doxycycline mitigates such changes both in vitro and in vivo. Our findings illuminate the possible role of matrix metalloproteinases in the pathophysiology of lipopolysaccharide-induced microvascular hyperpermeability and pave the way for better understanding and treatment of this process.


Subject(s)
Anti-Bacterial Agents/pharmacology , Doxycycline/pharmacology , Endothelium, Vascular/metabolism , Lipopolysaccharides/metabolism , Matrix Metalloproteinases/drug effects , Actins/drug effects , Animals , Capillary Permeability/drug effects , Catenins/drug effects , Endothelial Cells/metabolism , In Vitro Techniques , Male , Matrix Metalloproteinase 9/drug effects , Rats , Rats, Sprague-Dawley
7.
J Vis Exp ; (99): e52699, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25992584

ABSTRACT

Ischemia-Reperfusion (IR) injury is known to contribute significantly to the morbidity and mortality associated with ischemic strokes. Ischemic cerebrovascular accidents account for 80% of all strokes. A common cause of IR injury is the rapid inflow of fluids following an acute/chronic occlusion of blood, nutrients, oxygen to the tissue triggering the formation of free radicals. Ischemic stroke is followed by blood-brain barrier (BBB) dysfunction and vasogenic brain edema. Structurally, tight junctions (TJs) between the endothelial cells play an important role in maintaining the integrity of the blood-brain barrier (BBB). IR injury is an early secondary injury leading to a non-specific, inflammatory response. Oxidative and metabolic stress following inflammation triggers secondary brain damage including BBB permeability and disruption of tight junction (TJ) integrity. Our protocol presents an in vitro example of oxygen-glucose deprivation and reoxygenation (OGD-R) on rat brain endothelial cell TJ integrity and stress fiber formation. Currently, several experimental in vivo models are used to study the effects of IR injury; however they have several limitations, such as the technical challenges in performing surgeries, gene dependent molecular influences and difficulty in studying mechanistic relationships. However, in vitro models may aid in overcoming many of those limitations. The presented protocol can be used to study the various molecular mechanisms and mechanistic relationships to provide potential therapeutic strategies. However, the results of in vitro studies may differ from standard in vivo studies and should be interpreted with caution.


Subject(s)
Blood-Brain Barrier/metabolism , Glucose/deficiency , Hypoxia-Ischemia, Brain/metabolism , Reperfusion Injury/metabolism , Animals , Blood-Brain Barrier/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Free Radicals/metabolism , Glucose/metabolism , Hypoxia-Ischemia, Brain/pathology , Oxygen/metabolism , Permeability , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Tight Junctions/metabolism
8.
Metab Brain Dis ; 30(5): 1093-104, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25624154

ABSTRACT

Traumatic brain injury is a serious cause of morbidity and mortality worldwide. After traumatic brain injury, the blood-brain barrier, the protective barrier between the brain and the intravascular compartment, becomes dysfunctional, leading to leakage of proteins, fluid, and transmigration of immune cells. As this leakage has profound clinical implications, including edema formation, elevated intracranial pressure and decreased perfusion pressure, much interest has been paid to better understanding the mechanisms responsible for these events. Various molecular pathways and numerous mediators have been found to be involved in the intricate process of regulating blood-brain barrier permeability following traumatic brain injury. This review provides an update to the existing knowledge about the various pathophysiological pathways and advancements in the field of blood-brain barrier dysfunction and hyperpermeability following traumatic brain injury, including the role of various tight junction proteins involved in blood-brain barrier integrity and regulation. We also address pitfalls of existing systems and propose strategies to improve the various debilitating functional deficits caused by this progressive epidemic.


Subject(s)
Blood-Brain Barrier/physiopathology , Brain Injuries/physiopathology , Brain/physiopathology , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain Edema/etiology , Brain Edema/metabolism , Brain Edema/physiopathology , Brain Injuries/complications , Brain Injuries/metabolism , Capillary Permeability/physiology , Humans , Intracranial Pressure/physiology
9.
Am J Surg ; 208(6): 954-60; discussion 960, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25312844

ABSTRACT

Traumatic brain injuries cause vascular hyperpermeability. Tumor necrosis factor-α (TNF-α), matrix metalloproteinase-9 (MMP-9), and caspase-3 may be important in these processes but the relationship between them has not been investigated. We hypothesized that TNF-α regulates caspase-3-mediated hyperpermeability and blood brain barrier damage and hyperpermeability directly or indirectly via activation of MMP-9. To test this, rat brain microvascular endothelial cells were treated with TNF-α with or without inhibition of MMP-9. Monolayer permeability was measured, zonula occludens-1 and F-actin configuration were examined, and MMP-9 and caspase-3 activities were quantified. TNF-α increased monolayer permeability, damaged zonula occludens-1, induced filamentous-actin stress fiber formation, and increased both MMP-9 and caspase-3 activities. Inhibition of MMP-9 attenuated these changes. These data highlight a novel link between TNF-α and MMP-9 and show that TNF-α regulated caspase-3-mediated hyperpermeability and vascular damage may be linked to MMP-9 in vitro. These findings augment the understanding of traumatic brain injury and pave the way for improved treatment.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/cytology , Matrix Metalloproteinase 9/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Brain Injuries/metabolism , Caspase 3/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Rats , Zonula Occludens-1 Protein/metabolism
10.
J Trauma Acute Care Surg ; 77(6): 899-905; discussion 905, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25051382

ABSTRACT

BACKGROUND: Burns induce systemic inflammatory reactions and vascular hyperpermeability. Breakdown of endothelial cell adherens junctions is integral in this process, and reactive oxygen species (ROS) and proteolytic enzymes such as matrix metalloproteinase-9 (MMP-9) play pivotal roles therein. Outside trauma, melatonin has shown to exhibit anti-MMP activity and to be a powerful antioxidant. Consequently, we hypothesized that burn-induced junctional damage and hyperpermeability could be attenuated with melatonin. METHODS: Sprague-Dawley rats were assigned to sham or burn groups. Fluorescein isothiocyanate-bovine albumin was administered intravenously. Venules were examined with intravital microscopy; fluorescence intensities were measured intravascularly and extravascularly. Serum was collected. Rat lung microvascular endothelial cells were grown as monolayers and divided into four groups: sham serum and burn serum with and without melatonin pretreatment. Fluorescein isothiocyanate-bovine albumin flux was measured. Immunofluorescence for adherens junction proteins and staining for actin were performed, and images were captured. Cells were grown on 96 well plates, and ROS species generation following application of burn and sham serum was analyzed with and without melatonin. Statistical analysis was conducted with the Student's t test. RESULTS: Intravital microscopy data revealed an increase in vascular hyperpermeability following burn (p < 0.05). Monolayer permeability was increased with burn serum (p < 0.05); this was attenuated with melatonin (p < 0.05). Immunofluorescence showed damage of rat lung microvascular endothelial cell adherens junctions with burn serum exposure, and melatonin restored integrity. Rhodamine phalloidin staining showed filamentous actin stress fiber formation after burn serum application, and melatonin decreased this. Burn serum significantly increased ROS species generation (p < 0.05), and melatonin negated this (p < 0.05). CONCLUSION: Burns damage endothelial adherens junctions and induce microvascular hyperpermeability; melatonin attenuates this process. This insight into the mechanisms of burn-induced fluid leak suggests the role of ROS and MMP-9 but more importantly hints at the possibility of new treatments to combat vascular hyperpermeability in burns.


Subject(s)
Burns/drug therapy , Capillary Permeability/drug effects , Endothelium, Vascular/injuries , Melatonin/therapeutic use , Microvessels/injuries , Adherens Junctions/drug effects , Adherens Junctions/physiology , Animals , Burns/physiopathology , Capillary Permeability/physiology , Endothelium, Vascular/chemistry , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Microscopy, Fluorescence , Microvessels/drug effects , Microvessels/physiopathology , Rats, Sprague-Dawley , Reactive Oxygen Species/analysis
11.
Microcirculation ; 21(2): 187-95, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24372803

ABSTRACT

OBJECTIVE: Microvascular hyperpermeability that occurs due to breakdown of the BBB is a major contributor of brain vasogenic edema, following IR injury. In microvascular endothelial cells, increased ROS formation leads to caspase-3 activation following IR injury. The specific mechanisms, by which ROS mediates microvascular hyperpermeability following IR, are not clearly known. We utilized an OGD-R in vitro model of IR injury to study this. METHODS: RBMEC were subjected to OGD-R in presence of a caspase-3 inhibitor Z-DEVD, caspase-3 siRNA or an ROS inhibitor L-AA. Cytochrome c levels were measured by ELISA and caspase-3 activity was measured fluorometrically. TJ integrity and cytoskeletal assembly were studied using ZO-1 immunofluorescence and rhodamine phalloidin staining for f-actin, respectively. RESULTS: OGD-R significantly increased monolayer permeability, ROS formation, cytochrome c levels, and caspase-3 activity (p < 0.05) and induced TJ disruption and actin stress fiber formation. Z-DEVD, L-AA and caspase-3 siRNA significantly attenuated OGD-R-induced hyperpermeability (p < 0.05) while only L-AA decreased cytochrome c levels. Z-DEVD and L-AA protected TJ integrity and actin cytoskeletal assembly. CONCLUSIONS: These results suggest that OGD-R-induced hyperpermeability is ROS and caspase-3 dependent and can be regulated by their inhibitors.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Caspase 3/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Reactive Oxygen Species/metabolism , Animals , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain Edema/metabolism , Brain Edema/pathology , Brain Edema/physiopathology , Cell Hypoxia , Cells, Cultured , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...