Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Ann Thorac Surg ; 104(6): 1909-1914, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29100648

ABSTRACT

BACKGROUND: Paraplegia from spinal cord ischemia-reperfusion (SCIR) remains an elusive and devastating complication of complex aortic operations. Erythropoietin (EPO) attenuates this injury in models of SCIR. Upregulation of the EPO beta common receptor (ßcR) is associated with reduced damage in models of neural injury. The purpose of this study was to examine whether EPO-mediated neuroprotection was dependent on ßcR expression. We hypothesized that spinal cord neurons subjected to oxygen-glucose deprivation would mimic SCIR injury in aortic surgery and EPO treatment attenuates this injury in a ßcR-dependent fashion. METHODS: Lentiviral vectors with ßcR knockdown sequences were tested on neuron cell cultures. The virus with greatest ßcR knockdown was selected. Spinal cord neurons from perinatal wild-type mice were harvested and cultured to maturity. They were treated with knockdown or nonsense virus and transduced cells were selected. Three groups (ßcR knockdown virus, nonsense control virus, no virus control; n = 8 each) were subjected to 1 hour of oxygen-glucose deprivation. Viability was assessed. ßcR expression was quantified by immunoblot. RESULTS: EPO preserved neuronal viability after oxygen-glucose deprivation (0.82 ± 0.04 versus 0.61 ± 0.01; p < 0.01). Additionally, EPO-mediated neuron preservation was similar in the nonsense virus and control mice (0.82 ± 0.04 versus 0.80 ± 0.05; p = 0.77). EPO neuron preservation was lost in ßcR knockdown mice compared with nonsense control mice (0.46 ± 0.03 versus 0.80 ± 0.05; p < 0.01). CONCLUSIONS: EPO attenuates neuronal loss after oxygen-glucose deprivation in a ßcR-dependent fashion. This receptor holds immense clinical promise as a target for pharmacotherapies treating spinal cord ischemic injury.


Subject(s)
Erythropoietin/pharmacology , Neurons/drug effects , Neurons/metabolism , Neuroprotection/physiology , Receptors, Erythropoietin/metabolism , Spinal Cord/pathology , Cell Culture Techniques , Cell Survival , Humans , Spinal Cord/metabolism
2.
Semin Cardiothorac Vasc Anesth ; 20(4): 298-302, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27821787

ABSTRACT

Neurologic injuries following aortic arch operations can be devastating, with stroke occurring in up to 12% of elective operations and significant cerebral dysfunction occurring in up to 25% of cases. The primary challenge unique to aortic arch operations involves interruption of direct perfusion of the brachiocephalic vessels during arch reconstruction. For this reason, neuroprotection is paramount. The 2 main modes of protection are (1) reducing metabolic demand through hypothermia and (2) limiting, or even eliminating, the ischemic period. Preoperative selection of the cerebral perfusion plan for each operation is imperative to maintain maximal diffuse cerebral protection and prevent focal neurologic events.


Subject(s)
Aorta, Thoracic/surgery , Catheterization, Peripheral/methods , Cerebrovascular Circulation , Axillary Artery , Brachiocephalic Trunk , Humans , Hypothermia, Induced , Neuroprotection
4.
Adv Pediatr ; 62(1): 227-55, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26205116

ABSTRACT

A standardized classification system allows improvements in diagnostic accuracy. Multidisciplinary vascular anomaly centers combine medical, surgical, radiologic, and pathologic expertise. This collaborative approach tailors treatment and management of vascular anomalies for affected individuals.


Subject(s)
Disease Management , Vascular Malformations , Child , Global Health , Humans , Incidence , Vascular Malformations/diagnosis , Vascular Malformations/epidemiology , Vascular Malformations/therapy
5.
J Am Coll Surg ; 221(1): 197-205.e1, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26095572

ABSTRACT

BACKGROUND: Energy-based devices are used in virtually every operation. Our purposes were to describe causes of energy-based device complications leading to injury or death, and to determine if common mechanisms leading to injury or death can be identified. STUDY DESIGN: The FDA's Manufacturer and User Facility Device Experience (MAUDE) database was searched for surgical energy-based device injuries and deaths reported over 20 years (January 1994 to December 2013). Device-related complications were recorded and analyzed. RESULTS: We analyzed 178 deaths and 3,553 injuries. Common patterns of complications were: thermal burns, 63% (n = 2,353); hemorrhage, 17% (n = 642); mechanical failure of device, 12% (n = 442); and fire, 8% (n = 294). Events were identified intraoperatively in 82% (3,056), inpatient postoperatively in 9% (n = 351), and after discharge in 9% (n = 324). Of the deaths, 12% (n = 22) occurred after discharge home. Common mechanisms for thermal burn injuries were: direct application, 30% (n = 694); dispersive electrode burn, 29% (n = 657); and insulation failure, 14% (n = 324). Thermal injury was the most common reason for death (39%, n = 70). The mechanism for these thermal injuries was most frequently direct application (84%, n = 59, p < 0.001 vs all other mechanisms). Fires were most common with monopolar "Bovie" instruments (88%, n = 258, p < 0.001 vs all other devices) when they were used in head and neck operations (66%, n = 193, p < 0.001 vs all other locations). CONCLUSIONS: Complications due to energy-based devices occur from 4 main causes: thermal burn, hemorrhage, mechanical failure, and fire. Thermal direct application injuries are the most common reason for both injury and death.


Subject(s)
Burns/etiology , Electrical Equipment and Supplies/adverse effects , Equipment Failure , Intraoperative Complications/etiology , Postoperative Hemorrhage/etiology , Burns/mortality , Databases, Factual , Fires/statistics & numerical data , Humans , Intraoperative Complications/mortality , Postoperative Hemorrhage/mortality , United States , United States Food and Drug Administration
6.
Ann Thorac Surg ; 100(1): 41-6; discussion 46, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25865765

ABSTRACT

BACKGROUND: Paraplegia remains a devastating complication of aortic surgery, occurring in up to 20% of complex thoracoabdominal repairs. Erythropoietin (EPO) attenuates this injury in models of spinal cord ischemia. Upregulation of the beta-common receptor (ßcR) subunit of the EPO receptor is associated with reduced damage in murine models of neural injury. This receptor activates anti-apoptotic pathways including signaling transducer and activator of transcription 3 (STAT3). We hypothesized that spinal cord ischemia-reperfusion injury upregulates the ßcR subunit with a subsequent increase in activated STAT3. METHODS: Adult male C57/BL6 mice received an intraperitoneal injection of 0.5 mL of EPO (10 U/kg) or 0.9% saline after induction of anesthesia. Spinal cord ischemia was induced through sternotomy and 4-minute thoracic aortic cross-clamp. Sham mice underwent sternotomy without cross-clamp placement. Four groups were studied: ischemic and sham groups, each with and without EPO treatment. After 4 hours of reperfusion, spinal cords were harvested and homogenized. The ßcR subunit expression and STAT3 activation were evaluated by immunoblot. RESULTS: Ischemia reperfusion increased ßcR subunit expression in spinal cords of ischemia + saline and ischemia + EPO mice compared with shams (3.4 ± 1.39 vs 1.31 ± 0.3, p = 0.01 and 3.80 ± 0.58 vs 1.56 ± 0.32, p = 0.01). Additionally, both ischemic groups demonstrated increased STAT3 activation compared with shams (1.35 ± 0.14 vs 1.09 ± 0.07, p = 0.01 and 1.66 ± 0.35 vs 1.08 ± 0.17, p = 0.02). CONCLUSIONS: Ischemia-reperfusion injury induces EPO receptor ßcR subunit expression and early downstream anti-apoptotic signaling through STAT3 activation. Further investigation into the role of the ßcR subunit is warranted to determine tissue protective functions of EPO. Elucidation of mechanisms involved in spinal cord protection is essential for reducing delayed paraplegia.


Subject(s)
Receptors, Erythropoietin/biosynthesis , Reperfusion Injury/metabolism , Spinal Cord Ischemia/metabolism , Spinal Cord/blood supply , Up-Regulation , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/physiology
7.
J Surg Res ; 195(1): 21-8, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25655994

ABSTRACT

BACKGROUND: Paraplegia secondary to spinal cord ischemia-reperfusion injury remains a devastating complication of thoracoabdominal aortic intervention. The complex interactions between injured neurons and activated leukocytes have limited the understanding of neuron-specific injury. We hypothesize that spinal cord neuron cell cultures subjected to oxygen-glucose deprivation (OGD) would simulate ischemia-reperfusion injury, which could be attenuated by specific alpha-2a agonism in an Akt-dependent fashion. MATERIALS AND METHODS: Spinal cords from perinatal mice were harvested, and neurons cultured in vitro for 7-10 d. Cells were pretreated with 1 µM dexmedetomidine (Dex) and subjected to OGD in an anoxic chamber. Viability was determined by MTT assay. Deoxyuridine-triphosphate nick-end labeling staining and lactate dehydrogenase (LDH) assay were used for apoptosis and necrosis identification, respectively. Western blot was used for protein analysis. RESULTS: Vehicle control cells were only 59% viable after 1 h of OGD. Pretreatment with Dex significantly preserves neuronal viability with 88% viable (P < 0.05). Dex significantly decreased apoptotic cells compared with that of vehicle control cells by 50% (P < 0.05). Necrosis was not significantly different between treatment groups. Mechanistically, Dex treatment significantly increased phosphorylated Akt (P < 0.05), but protective effects of Dex were eliminated by an alpha-2a antagonist or Akt inhibitor (P < 0.05). CONCLUSIONS: Using a novel spinal cord neuron cell culture, OGD mimics neuronal metabolic derangement responsible for paraplegia after aortic surgery. Dex preserves neuronal viability and decreases apoptosis in an Akt-dependent fashion. Dex demonstrates clinical promise for reducing the risk of paraplegia after high-risk aortic surgery.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/therapeutic use , Cardiovascular Surgical Procedures/adverse effects , Dexmedetomidine/therapeutic use , Reperfusion Injury/prevention & control , Spinal Cord Injuries/prevention & control , Animals , Apoptosis , Cell Survival , Cells, Cultured , Cytokines/metabolism , Drug Evaluation, Preclinical , Glucose/deficiency , Hypoxia , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord Injuries/etiology
8.
J Thorac Cardiovasc Surg ; 149(2): 578-84; discussion 584-6, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25454921

ABSTRACT

OBJECTIVES: Delayed paraplegia secondary to ischemia-reperfusion injury is a devastating complication of thoracoabdominal aortic surgery. Alpha-2 agonists have been shown to attenuate ischemia-reperfusion injury, but the mechanism for protection has yet to be elucidated. A growing body of evidence suggests that astrocytes play a critical role in neuroprotection by release of neurotrophins. We hypothesize that alpha-2 agonism with dexmedetomidine increases glial cell-line-derived neurotrophic factor in spinal cord astrocytes to provide spinal cord protection. METHODS: Spinal cords were isolated en bloc from C57BL/6 mice, and primary spinal cord astrocytes and neurons were selected for and grown separately in culture. Astrocytes were treated with dexmedetomidine, and glial cell-line-derived neurotrophic factor was tested for by enzyme-linked immunosorbent assay. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was used to assess neuronal viability. RESULTS: Spinal cord primary astrocytes treated with dexmedetomidine at 1 µmol/L and 10 µmol/L had significantly increased glial cell-line-derived neurotrophic factor production compared with control (P < .05). Neurons subjected to oxygen glucose deprivation had significant preservation (P < .05) of viability with use of dexmedetomidine-treated astrocyte media. Glial cell-line-derived neurotrophic factor neutralizing antibody eliminated the protective effects of the dexmedetomidine-treated astrocyte media (P < .05). CONCLUSIONS: Astrocytes have been shown to preserve neuronal viability via release of neurotrophic factors. Dexmedetomidine increases glial cell-derived neurotrophic factor from spinal cord astrocytes via the alpha-2 receptor. Treatment with alpha-2 agonist dexmedetomidine may be a clinical tool for use in spinal cord protection in aortic surgery.


Subject(s)
Astrocytes/drug effects , Reperfusion Injury/complications , Spinal Cord Ischemia/etiology , Spinal Cord Ischemia/prevention & control , Spinal Cord/drug effects , Animals , Cell Culture Techniques , Dexmedetomidine/pharmacology , Enzyme-Linked Immunosorbent Assay , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mice , Mice, Inbred C57BL , Spinal Cord/cytology
9.
J Thorac Cardiovasc Surg ; 149(1): 369-75, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25439778

ABSTRACT

OBJECTIVES: C-terminal tensin-like (Cten) protein, a component of focal adhesions, contributes to cell motility and invasion in multiple human cancers. Epidermal growth factor can activate signal transducer and activator of transcription 3, and both contribute to invasion through focal adhesion interactions. We hypothesize that Cten may mediate invasion of lung cancer cells provided by epidermal growth factor via signal transducer and activator of transcription 3. METHODS: Four human non-small cell lung cancer cell lines were treated with epidermal growth factor to evaluate activation of the signal transducer and activator of transcription 3 pathway and induction of Cten expression. Chemical inhibition of signal transducer and activator of transcription 3 was used to evaluate the effect on epidermal growth factor-induced Cten expression. Protein expression was quantified by Western blot. H125 and A549 cells were transduced with short-hairpin RNA via lentiviral vector to knockdown expression of Cten. An in vitro transwell invasion assay was used to assess the effects of Cten knockdown on cell invasion (n = 3 for all experiments). RESULTS: Stimulation of lung cancer cells with epidermal growth factor activated the signal transducer and activator of transcription 3 pathway and induced expression of Cten in all cell lines. Signal transducer and activator of transcription 3 inhibition significantly reduced epidermal growth factor-induced expression of Cten in H125 (P < .0001), H358 (P = .006), and H441 (P = .014) cells in a dose-dependent manner. Knockdown of Cten expression resulted in significant decreases in cellular invasion in both H125 (P = .0036) and A549 (P = .0006) cells. CONCLUSIONS: These are the first findings in lung cancer to demonstrate that Cten expression mediates invasion of human lung cancer cells and is upregulated by epidermal growth factor via signal transducer and activator of transcription 3 pathway. Cten should be considered a potential therapeutic target for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Lung Neoplasms/metabolism , Microfilament Proteins/metabolism , STAT3 Transcription Factor/metabolism , Aminosalicylic Acids/pharmacology , Benzenesulfonates/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Microfilament Proteins/genetics , Neoplasm Invasiveness , RNA Interference , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction , Tensins , Time Factors , Transfection
10.
J Thorac Cardiovasc Surg ; 149(3): 920-4, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25500291

ABSTRACT

OBJECTIVE: Paraplegia remains a devastating complication of complex aortic surgery. Erythropoietin (EPO) has been shown to prevent paraplegia after ischemia reperfusion, but the protective mechanism remains poorly described in the spinal cord. We hypothesized that EPO induces the CREB (cAMP [adenosine 3'5' cyclic monophosphate] response element-binding protein) pathway and neurotrophin production in the murine spinal cord, attenuating functional and cellular injury. METHODS: Adult male mice were subjected to 4 minutes of spinal cord ischemia via an aortic and left subclavian cross-clamp. Experimental groups included EPO treatment 4 hours before incision (n = 7), ischemic control (n = 7), and shams (n = 4). Hind-limb function was assessed using the Basso motor score for 48 hours after reperfusion. Spinal cords were harvested and analyzed for neuronal viability using histology and staining with a fluorescein derivative. Expression of phosphorylated (p)AKT (a serine/threonine-specific kinase), pCREB, B-cell lymphoma 2, and brain-derived neurotrophic factor were determined using immunoblotting. RESULTS: By 36 hours of reperfusion, EPO significantly preserved hind-limb function after ischemia-reperfusion injury (P < .01). Histology demonstrated preserved cytoarchitecture in the EPO treatment group. Cords treated with EPO expressed significant increases in pAKT (P = .021) and pCREB (P = .038). Treatment with EPO induced expression of both of the neurotrophins, B-cell lymphoma 2, and brain-derived neurotrophic factor, beginning at 12 hours. CONCLUSIONS: Erythropoietin-mediated induction of the CREB pathway and production of neurotrophins is associated with improved neurologic function and increased neuronal viability following spinal cord ischemia reperfusion. Further elucidation of EPO-derived neuroprotection will allow for expansion of adjunct mechanisms for spinal cord protection in high-risk thoracoabdominal aortic intervention.


Subject(s)
CREB-Binding Protein/metabolism , Erythropoietin/pharmacology , Paraplegia/prevention & control , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Spinal Cord Ischemia/drug therapy , Spinal Cord/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Paraplegia/enzymology , Paraplegia/physiopathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/physiopathology , Spinal Cord/enzymology , Spinal Cord/physiopathology , Spinal Cord Ischemia/enzymology , Spinal Cord Ischemia/physiopathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...