Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Sci Rep ; 11(1): 13696, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34211011

ABSTRACT

Resuscitative endovascular balloon occlusion of the aorta (REBOA) is a lifesaving maneuver for the management of lethal torso hemorrhage. However, its prolonged use leads to distal organ ischemia-reperfusion injury (IRI) and systemic inflammatory response syndrome (SIRS). The objective of this study is to investigate the blood-based biomarkers of IRI and SIRS and the efficacy of direct intestinal cooling in the prevention of IRI and SIRS. A rat lethal hemorrhage model was produced by bleeding 50% of the total blood volume. A balloon catheter was inserted into the aorta for the implementation of REBOA. A novel TransRectal Intra-Colon (TRIC) device was placed in the descending colon and activated from 10 min after the bleeding to maintain the intra-colon temperature at 37 °C (TRIC37°C group) or 12 °C (TRIC12°C group) for 270 min. The upper body temperature was maintained at as close to 37 °C as possible in both groups. Blood samples were collected before hemorrhage and after REBOA. The organ injury biomarkers and inflammatory cytokines were evaluated by ELISA method. Blood based organ injury biomarkers (endotoxin, creatinine, AST, FABP1/L-FABP, cardiac troponin I, and FABP2/I-FABP) were all drastically increased in TRIC37°C group after REBOA. TRIC12°C significantly downregulated these increased organ injury biomarkers. Plasma levels of pro-inflammatory cytokines TNF-α, IL-1b, and IL-17F were also drastically increased in TRIC37°C group after REBOA. TRIC12°C significantly downregulated the pro-inflammatory cytokines. In contrast, TRIC12°C significantly upregulated the levels of anti-inflammatory cytokines IL-4 and IL-10 after REBOA. Amazingly, the mortality rate was 100% in TRIC37°C group whereas 0% in TRIC12°C group after REBOA. Directly cooling the intestine offered exceptional protection of the abdominal organs from IRI and SIRS, switched from a harmful pro-inflammatory to a reparative anti-inflammatory response, and mitigated mortality in the rat model of REBOA management of lethal hemorrhage.


Subject(s)
Balloon Occlusion/methods , Hemorrhage/therapy , Animals , Balloon Occlusion/adverse effects , Balloon Occlusion/instrumentation , Cold Temperature , Hemorrhage/pathology , Inflammation/etiology , Inflammation/pathology , Intestines/injuries , Male , Rats, Sprague-Dawley , Resuscitation/adverse effects , Resuscitation/instrumentation , Resuscitation/methods
2.
Perfusion ; 36(3): 285-292, 2021 04.
Article in English | MEDLINE | ID: mdl-32723149

ABSTRACT

INTRODUCTION: Extracorporeal membrane oxygenation circuit performance can be compromised by oxygenator thrombosis. Stagnant blood flow in the oxygenator can increase the risk of thrombus formation. To minimize thrombogenic potential, computational fluid dynamics is frequently applied for identification of stagnant flow conditions. We investigate the use of computed tomography angiography to identify flow patterns associated with thrombus formation. METHODS: A computed tomography angiography was performed on a Quadrox D oxygenator, and video densitometric parameters associated with flow stagnation were measured from the acquired videos. Computational fluid dynamics analysis of the same oxygenator was performed to establish computational fluid dynamics-based flow characteristics. Forty-one Quadrox D oxygenators were sectioned following completion of clinical use. Section images were analyzed with software to determine oxygenator clot burden. Linear regression was used to correlate clot burden to computed tomography angiography and computational fluid dynamics-based flow characteristics. RESULTS: Clot burden from the explanted oxygenators demonstrated a well-defined pattern, with the largest clot burden at the corner opposite the blood inlet and outlet. The regression model predicted clot burden by region of interest as a function of time to first opacification on computed tomography angiography (R2 = 0.55). The explanted oxygenator clot burden map agreed well with the computed tomography angiography predicted clot burden map. The computational fluid dynamics parameter of residence time, when summed in the Z-direction, was partially predictive of clot burden (R2 = 0.35). CONCLUSION: In the studied oxygenator, clot burden follows a pattern consistent with clinical observations. Computed tomography angiography-based flow analysis provides a useful adjunct to computational fluid dynamics-based flow analysis in understanding oxygenator thrombus formation.


Subject(s)
Extracorporeal Membrane Oxygenation , Thrombosis , Computed Tomography Angiography , Humans , Hydrodynamics , Oxygenators , Oxygenators, Membrane , Thrombosis/diagnostic imaging
3.
Artif Organs ; 44(7): 717-726, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31970795

ABSTRACT

The roles of the large membrane surface of the oxygenator and the high mechanical shear stress (HMSS) of the pump in the extracorporeal membrane oxygenation (ECMO) circuit were examined under a pediatric support setting. A clinical centrifugal pump and a pediatric oxygenator were used to construct the ECMO circuit. An identical circuit without the oxygenator was constructed for comparison. Fresh human blood was circulated in the two circuits for 4 hours under the identical pump speed and flow. Blood samples were collected hourly for blood damage assessment, including platelet activation, generation of platelet-derived microparticles (PDMP), losses of key platelet hemostasis receptors (glycoprotein (GP) Ibα (GPIbα) and GPVI), and high molecular weight multimers (HMWM) of von Willebrand factor (VWF) and plasma free hemoglobin (PFH). Platelet adhesion on fibrinogen, VWF, and collagen was further examined. The levels of platelet activation and generation of PDMP and PFH exhibited an increasing trend with circulation time while the expression levels of GPIbα and GPVI receptors on the platelet surface decreased. Correspondingly, the platelets in the blood samples exhibited increased adhesion capacity to fibrinogen and decreased adhesion capacities on VWF and collagen with circulation time. Loss of HMWM of VWF occurred in both circuits. No statistically significant differences were found in all the measured parameters for blood damage and platelet adhesion function between the two circuits. The results indicate that HMSS from the pump played a dominant role in blood damage associated with ECMO and the impact of the large surface of the oxygenator on blood damage was insignificant.


Subject(s)
Blood Platelets/metabolism , Extracorporeal Membrane Oxygenation/adverse effects , Hemorrhage/etiology , Oxygenators, Membrane/adverse effects , Thrombosis/etiology , Blood Platelets/cytology , Cell-Derived Microparticles/metabolism , Child , Extracorporeal Membrane Oxygenation/instrumentation , Healthy Volunteers , Hemorrhage/blood , Hemorrhage/prevention & control , Humans , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Stress, Mechanical , Thrombosis/blood , Thrombosis/prevention & control
4.
ASAIO J ; 66(5): 524-531, 2020 05.
Article in English | MEDLINE | ID: mdl-31192844

ABSTRACT

In order to explore the role of a disintegrin and metalloproteinase (ADAM) proteolysis and direct mechanical damage in non-physiologic shear stress (NPSS)-caused platelet receptor shedding, the healthy donor blood treated with/without ADAM inhibitor was exposed to NPSS (150 Pa). The expression of the platelet surface receptors glycoprotein (GP) Ibα and glycoprotein (GP) VI (GPVI) in NPSS-damaged blood was quantified with flow cytometry. The impact of ADAM inhibition on adhesion of NPSS-damaged platelets on von Willibrand factor (VWF) and collagen was explored with fluorescence microscopy. The impact of ADAM inhibition on ristocetin- and collagen-caused aggregation of NPSS-damaged platelets was examined by aggregometry. The results showed that ADAM inhibition could lessen the NPSS-induced loss of platelet surface receptor GPIbα (12%) and GPVI (9%), moderately preserve adhesion of platelets on VWF (7.4%) and collagen (8.4%), and partially restore the aggregation of NPSS-sheared platelets induced by ristocetin (18.6 AU*min) and collagen (48.2 AU*min). These results indicated that ADAM proteolysis played a role in NPSS-induced receptor shedding. However, the ADAM inhibition couldn't completely suppress the NPSS-caused loss of the platelet surface receptors (GPIbα and GPVI), only partially prevented the NPSS-induced reduction of platelet adhesion to VWF and collagen, and the agonist (ristocetin and collagen)-caused platelet aggregation. These results suggested that the direct mechanical damage is partially responsible for NPSS-induced receptor shedding in addition to the ADAM proteolysis. In conclusion, NPSS relevant to blood contacting medical devices can induce ADAM proteolysis and direct mechanical damage on the platelet receptor GPIbα and GPVI, leading to comprised hemostasis.


Subject(s)
Blood Platelets/metabolism , Disintegrins/metabolism , Metalloproteases/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Stress, Mechanical , Flow Cytometry , Humans , Proteolysis
5.
Artif Organs ; 44(5): E201-E213, 2020 May.
Article in English | MEDLINE | ID: mdl-31849074

ABSTRACT

The aim of this study was to examine the impact of the nonphysiological shear stress (NPSS) on platelet hemostatic function relevant to thrombosis and bleeding in mechanically assisted circulation. Fresh human blood was circulated for four hours in in vitro circulatory flow loops with a CentriMag blood pump operated under a flow rate of 4.5 L/min against three pressure heads (70 mm Hg, 150 mm Hg, and 350 mm Hg) at 2100, 2800, and 4000 rpm, respectively. Hourly blood samples from the CentriMag pump-assisted circulation loops were collected and analyzed for glycoprotein (GP) IIb/IIIa activation and receptor shedding of GPVI and GPIbα on the platelet surface with flow cytometry. Adhesion of platelets to fibrinogen, collagen, and von Willebrand factor (VWF) of the collected blood samples was quantified with fluorescent microscopy. In parallel, mechanical shear stress fields within the CentriMag pump operated under the three conditions were assessed by computational fluid dynamics (CFD) analysis. The experimental results showed that levels of platelet GPIIb/IIIa activation and platelet receptor shedding (GPVI and GPIbα) in the blood increased with increasing the circulation time. The levels of platelet activation and loss of platelet receptors GPVI and GPIbα were consistently higher with higher pressure heads at each increasing hour in the CentriMag pump-assisted circulation. The platelet adhesion on fibrinogen increased with increasing the circulation time for all three CentriMag operating conditions and was correlated well with the level of platelet activation. In contrast, the platelet adhesion on collagen and VWF decreased with increasing the circulation time under all the three conditions and was correlated well with the loss of the receptors GPVI and GPIbα on the platelet surface, respectively. The CFD results showed that levels of shear stresses inside the CentriMag pump under all three operating conditions exceeded the maximum level of shear stress in the normal physiological circulation and were strongly dependent on the pump operating condition. The level of platelet activation and loss of key platelet adhesion receptors (GPVI and GPIbα) were correlated with the level of NPSS generated by the CentriMag pump, respectively. In summary, the level of NPSS associated with pump operating condition is a critical determinant of platelet dysfunction in mechanically assisted circulation.


Subject(s)
Blood Platelet Disorders/etiology , Heart-Assist Devices/adverse effects , Hemostatic Disorders/etiology , Platelet Activation , Thrombosis , Adult , Female , Humans , Hydrodynamics , Male , Stress, Mechanical , Young Adult
6.
Artif Organs ; 44(5): 478-487, 2020 May.
Article in English | MEDLINE | ID: mdl-31854002

ABSTRACT

Extracorporeal membrane oxygenation (ECMO) has become a mainstay of therapy for patients suffering from severe respiratory failure. Ambulatory ECMO systems aim to provide long-term out-of-hospital respiratory support. As a patient's activity level changes, the required level of ECMO support varies with oxygen consumption and metabolic fluctuations. To compensate for such changes, an autoregulatory ECMO system (AR-ECMO) has been developed and its performance was evaluated as a proof of concept in an acute ovine model. The AR-ECMO system consists of a regular ECMO circuit and an electromechanical control system. A custom fuzzy logic control algorithm was implemented to adjust the blood flow and sweep gas flow of the ECMO circuit to meet the varying respiratory demand by utilizing two noninvasive sensors for venous oxyhemoglobin saturation and the oxygenator exhaust gas CO2 concentration. Disturbance responses of the AR-ECMO to induced acute respiratory distress were assessed for six hours in four juvenile sheep cannulated with a veno-pulmonary artery ECMO configuration, including acute ventilator shutoff, ventilator step change (off-on-off), and forced desaturation. All sheep survived for the study duration. The AR-ECMO system was able to respond and maintain stable hemodynamics and physiological blood gas contents (SpO2  = 96.3 % ± 4.29, pH 7.44 ± 0.09, pCO2  = 38.9 ± 9.9 mm Hg, and pO2 =237.9 ± 123.6 mm Hg) during simulated respiratory distress. Acceptable correlation between oxygenator exhaust gas CO2 and oxygenator outlet pCO2 were observed (R2  = 0.84). In summary, the AR-ECMO system successfully maintained physiologic control of peripheral oxygenation and carbon dioxide over the study period, utilizing only measurements taken directly from the ECMO circuit. The range of system response necessitates an adaptable system in the setting of variable metabolic demands. The ability of this system to respond to significant disturbances in ventilator support is encouraging. Future work to evaluate our AR-ECMO system in long-term, awake animal studies is necessary for further refinement.


Subject(s)
Extracorporeal Membrane Oxygenation/instrumentation , Animals , Fuzzy Logic , Male , Sheep
7.
Artif Organs ; 43(9): 897-908, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30972780

ABSTRACT

The PI3K/Akt signaling pathway has been implicated in playing an important role in platelet activation during hemostasis and thrombosis involving platelet-matrix interaction and platelet aggregation. Its role in non-physiological shear stress (NPSS)-induced platelet activation relevant to high-shear blood contacting medical devices (BCMDs) is unclear. In the context of blood cells flowing in BCMDs, platelets are subjected to NPSS (>100 Pa) with very short exposure time (<1 s). In this study, we investigated whether NPSS with short exposure time induces platelet activation through the PI3K/Akt signaling pathway. Healthy donor blood treated with or without PI3K inhibitor was subjected to NPSS (150 Pa) with short exposure time (0.5 s). Platelet activation indicated by the surface P-selectin expression and activated glycoprotein (GP) IIb/IIIa was quantified using flow cytometry. The phosphorylation of Akt, activation of the PI3K signaling, was characterized by western blotting. Changes in adhesion behavior of NPSS-sheared platelets on fibrinogen, collagen, and von Willebrand factor (vWF) were quantified with fluorescent microscopy by perfusing the NPSS-sheared and PI3K inhibitor-treated blood through fibrinogen, collagen, and vWF-coated microcapillary tubes. The results showed that the PI3K/Akt signaling was involved with both NPSS-induced platelet activation and platelet-matrix interaction. NPSS-sheared platelets exhibited exacerbated platelet adhesion on fibrinogen, but had diminished platelet adhesion on collagen and vWF. The inhibition of PI3K signaling reduced P-selectin expression and GPIIb/IIIa activation with suppressed Akt phosphorylation and abolished NPSS-enhanced platelet adhesion on fibrinogen in NPSS-sheared blood. The inhibition of PI3K signaling can attenuate the adhesion of unsheared platelets (baseline) on collagen and vWF, while had no impact on adhesion of NPSS-sheared platelets on collagen and vWF. This study confirmed the important role of PI3K/Akt signaling pathway in NPSS-induced platelet activation. The finding of this study suggests that blocking PI3K/Akt signaling pathway could be a potential method to treat thrombosis in patients implanted with BCMDs.


Subject(s)
Blood Platelets/cytology , Phosphatidylinositol 3-Kinases/metabolism , Platelet Activation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adult , Blood Platelets/metabolism , Female , Humans , Male , Phosphorylation , Stress, Mechanical , Young Adult
9.
Ann Thorac Surg ; 104(6): 2045-2053, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28760475

ABSTRACT

BACKGROUND: C-kit+ cardiac progenitor cells (CPCs) have been shown to be safe and effective in large-animal models and in an early-phase clinical trial for adult patients with ischemic heart disease. However, CPCs have not yet been evaluated in a preclinical model of right ventricular (RV) dysfunction, which is a salient feature of many forms of congenital heart disease. METHODS: Human c-kit+ CPCs were generated from right atrial appendage biopsy specimens obtained during routine congenital cardiac operations. Immunosuppressed Yorkshire swine (6 to 9 kg) underwent pulmonary artery banding to induce RV dysfunction. Thirty minutes after banding, pigs received intramyocardial injection into the RV free wall with c-kit+ CPCs (1 million cells, n = 5) or control (phosphate-buffered saline, n = 5). Pigs were euthanized at 30 days postbanding. RESULTS: Banding was calibrated to a consistent rise in the RV-to-systemic pressure ratio across both groups (postbanding: CPCs = 0.76 ± 0.06, control = 0.75 ± 0.03). At 30 days postbanding, the CPCs group demonstrated less RV dilatation and a significantly greater RV fractional area of change than the control group (p = 0.002). In addition, measures of RV myocardial strain, including global longitudinal strain and strain rate, were significantly greater in the CPCs group at 4 weeks relative to control (p = 0.004 and p = 0.01, respectively). The RV free wall in the CPCs group demonstrated increased arteriole formation (p < 0.0001) and less myocardial fibrosis compared with the control group (p = 0.02). CONCLUSIONS: Intramyocardial injection of c-kit+ CPCs results in enhanced RV performance relative to control at 30 days postbanding in neonatal pigs. This model is important for further evaluation of c-kit+ CPCs, including long-term efficacy.


Subject(s)
Pulmonary Artery/surgery , Stem Cell Transplantation , Stem Cells/cytology , Ventricular Dysfunction, Right/therapy , Ventricular Function, Right/physiology , Ventricular Remodeling , Animals , Animals, Newborn , Humans , Ligation , Swine , Ventricular Dysfunction, Right/etiology
10.
Mol Cell Biochem ; 433(1-2): 125-137, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28343311

ABSTRACT

Non-surgical bleeding (NSB) is the most common clinical complication in heart failure (HF) patients supported by continuous-flow left ventricular assist devices (CF-LVADs). In this study, oxidative stress and alteration of signal pathways leading to platelet apoptosis were investigated. Thirty-one HF patients supported by CF-LVADs were divided into bleeder (n = 12) and non-bleeder (n = 19) groups. Multiple blood samples were collected at pre-implant (baseline) and weekly up to 1-month post-implant. A single blood sample was collected from healthy subjects (reference). Production of reactive oxygen species (ROS) in platelets, total antioxidant capacity (TAC), oxidized low-density lipoproteins (oxLDL), expression of Bcl-2 and Bcl-xL, Bax and release of cytochrome c (Cyt.c), platelet mitochondrial membrane potential (Δψ m), activation of caspases, gelsolin cleavage and platelet apoptosis were examined. Significantly elevated ROS, oxLDL and depleted TAC were evident in the bleeder group compared to non-bleeder group (p < 0.05). Platelet pro-survival proteins (Bcl-2, Bcl-xL) were significantly reduced in the bleeder group in comparison to the non-bleeder group (p < 0.05). Translocation of Bax into platelet mitochondria membrane and subsequent release of Cyt.c were more prevalent in the bleeder group. Platelet mitochondrial damage, activation of caspases, gelsolin cleavage, and ultimate platelet apoptosis in the bleeder group were observed. Oxidative stress and activation of both intrinsic and extrinsic pathways of platelet apoptosis may be linked to NSB in CF-LVAD patients. Additionally, biomarkers of oxidative stress, examination of pro-survivals and pro-apoptotic proteins in platelets, mitochondrial damage, caspase activation, and platelet apoptosis may be used to help identify HF patients at high risk of NSB post-implant.


Subject(s)
Apoptosis , Blood Platelets/metabolism , Heart Failure , Heart-Assist Devices/adverse effects , Hemorrhage , Oxidative Stress , Adult , Aged , Blood Platelets/pathology , Female , Heart Failure/blood , Heart Failure/therapy , Hemorrhage/blood , Hemorrhage/etiology , Humans , Male , Middle Aged
11.
Ann Surg ; 266(6): 1091-1096, 2017 12.
Article in English | MEDLINE | ID: mdl-27735823

ABSTRACT

OBJECTIVE: We aimed to create a reproducible lung injury model utilizing injection of mitochondrial damage-associated molecular products. Our goal was to characterize the pathophysiologic response to damage-associated molecular pattern mediated organ injury. SUMMARY BACKGROUND DATA: There remain significant gaps in our understanding of acute respiratory distress syndrome, in part due to the lack of clinically applicable animal models of this disease. Animal models of noninfectious, tissue damage-induced lung injury are needed to understand the signals and responses associated with this injury. METHODS: Ten pigs (35-45 kg) received an intravenous dose of disrupted mitochondrial products and were followed for 6 hours under general anesthesia. These animals were compared to a control group (n = 5) and a model of lung injury induced by bacterial products (lipopolysaccharide n = 5). RESULTS: Heart rate and temperature were significantly elevated in the mitochondrial product (204 ±â€Š12 and 41 ±â€Š1) and lipopolysaccharide groups (178 ±â€Š18 and 42 ±â€Š0.5) compared with controls (100 ±â€Š13 and 38 ±â€Š0.5) (P <0.05). Lung oxygenation (PaO2/FiO2) was significantly lower 6 hours after injection in the mitochondrial products and lipopolysaccharide groups compared with controls (170 ±â€Š39, 196 ±â€Š27, and 564 ±â€Š75 mm Hg respectively, P = 0.001). Lung injury scoring of histological sections was significantly worse in mitochondrial and lipopolysaccharide groups compared with controls (mitochondrial-64 ±â€Š6, lipopolysaccharide-54 ±â€Š8, control-14 ±â€Š1.5, P= 0.002). CONCLUSIONS: Our data demonstrated that the presence of mitochondrial products in the circulation leads to systemic inflammatory response and lung injury. In its acute phase lung injury induced by tissue or bacterial products is clinically indistinguishable.


Subject(s)
Disease Models, Animal , Respiratory Distress Syndrome , Animals , Hemodynamics , Inflammation/pathology , Lipopolysaccharides , Lung/pathology , Lung/physiopathology , Mitochondria, Liver , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/physiopathology , Swine
12.
Perfusion ; 32(4): 301-305, 2017 May.
Article in English | MEDLINE | ID: mdl-27881701

ABSTRACT

AIM: We sought to quantify the location and volume of thrombus in used hollow-fiber membrane oxygenators and correlate the volume of thrombus with patient demographics, flow characteristics and anticoagulation parameters. METHODS: Hollow-fiber membrane oxygenators (Quadrox D, Maquet, Rastatt, Germany) were collected after clinical use in ECMO circuits and divided into sections. Each section was digitally imaged and analyzed using ImageJ software. The location and total volume (cm3) of thrombus was calculated for different sections. In an effort to predict thrombus formation, we correlated thrombus volume with possible aggravating and mitigating variables. RESULTS: We collected 41 oxygenators from 27 patients. Twenty-seven (66%) were configured in the veno-venous mode and 14 (34%) in the veno-arterial mode. The median duration of use was 131 hours (interquartile range 61-214 hours). Eighteen (44%) were removed when the patient recovered, six (15%) were removed after withdrawal of care and seventeen (41%) were exchanged. The median volume of thrombus was 11.4 cm3 (interquartile range 2.2-44.5 cm3). CONCLUSIONS: A multivariable linear regression model suggested that the combination of median flow, configuration of ECMO and visible thrombus partially predicted internal thrombus volume (adjusted R2=0.39).


Subject(s)
Extracorporeal Membrane Oxygenation/instrumentation , Extracorporeal Membrane Oxygenation/methods , Oxygenators, Membrane , Thrombosis/pathology , Adult , Female , Humans , Male , Middle Aged , Thrombosis/blood
13.
Ann Thorac Surg ; 102(6): 1919-1924, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27378553

ABSTRACT

BACKGROUND: The mechanism underlying left ventricular remodeling and reverse remodeling in the setting of mechanical support following acute myocardial infarction (MI) is unclear. We tested the hypothesis that left ventricular assist device (LVAD) unloading can decrease apoptotic signals after MI. METHODS: An MI model was created in 16 sheep by coronary artery ligation. Eight were unloaded with a LVAD during the first 2 weeks after MI and observed for 10 more weeks. Myocardial tissue was collected from the nonischemic adjacent zone and the remote zone. Proteins in the apoptotic matrix metalloproteinases (MMPs)-2/c-Jun N-terminal kinase (JNK) and prosurvival ß1D-integrin/focal adhesion kinase (FAK) pathway were quantified. RESULTS: Increased TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling) positive nuclei were observed in the MI group and to a lesser extent in the LVAD group (6.18 ± 0.26 versus 0.82 ± 0.18; p < 0.05). Pro-MMP-2, MMP-2, JNK, and phosphorylated (p)-JNK were all elevated in the adjacent zone of the MI-only group but not in the adjacent zone of the LVAD-supported group. There were higher levels of prosurvival p-FAK in the LVAD-supported group than in the MI group. CONCLUSIONS: MMP-2/JNK apoptotic and ß1D-integrin/FAK survival pathways are activated in the nonischemic adjacent zone after MI in adult sheep. LVAD unloading of approximately 50% cardiac output for 2 weeks attenuates remodeling in part by its negative effect on stretch-induced apoptosis and inhibition of MMP-2 activity.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/physiology , Heart Ventricles/physiopathology , Heart-Assist Devices , JNK Mitogen-Activated Protein Kinases/physiology , Matrix Metalloproteinase 2/physiology , Myocardial Infarction/physiopathology , Signal Transduction , Ventricular Remodeling/physiology , Animals , Apoptosis , Disease Models, Animal , In Situ Nick-End Labeling , Male , Myocardial Infarction/therapy , Myocardium/enzymology , Phosphorylation , Protein Processing, Post-Translational , Sheep , Stress, Mechanical
14.
Am J Physiol Heart Circ Physiol ; 310(11): H1816-26, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27106046

ABSTRACT

Limited therapies exist for patients with congenital heart disease (CHD) who develop right ventricular (RV) dysfunction. Bone marrow-derived mesenchymal stem cells (MSCs) have not been evaluated in a preclinical model of pressure overload, which simulates the pathophysiology relevant to many forms of CHD. A neonatal swine model of RV pressure overload was utilized to test the hypothesis that MSCs preserve RV function and attenuate ventricular remodeling. Immunosuppressed Yorkshire swine underwent pulmonary artery banding to induce RV dysfunction. After 30 min, human MSCs (1 million cells, n = 5) or placebo (n = 5) were injected intramyocardially into the RV free wall. Serial transthoracic echocardiography monitored RV functional indices including 2D myocardial strain analysis. Four weeks postinjection, the MSC-treated myocardium had a smaller increase in RV end-diastolic area, end-systolic area, and tricuspid vena contracta width (P < 0.01), increased RV fractional area of change, and improved myocardial strain mechanics relative to placebo (P < 0.01). The MSC-treated myocardium demonstrated enhanced neovessel formation (P < 0.0001), superior recruitment of endogenous c-kit+ cardiac stem cells to the RV (P < 0.0001) and increased proliferation of cardiomyocytes (P = 0.0009) and endothelial cells (P < 0.0001). Hypertrophic changes in the RV were more pronounced in the placebo group, as evidenced by greater wall thickness by echocardiography (P = 0.008), increased cardiomyocyte cross-sectional area (P = 0.001), and increased expression of hypertrophy-related genes, including brain natriuretic peptide, ß-myosin heavy chain and myosin light chain. Additionally, MSC-treated myocardium demonstrated increased expression of the antihypertrophy secreted factor, growth differentiation factor 15 (GDF15), and its downstream effector, SMAD 2/3, in cultured neonatal rat cardiomyocytes and in the porcine RV myocardium. This is the first report of the use of MSCs as a therapeutic strategy to preserve RV function and attenuate remodeling in the setting of pressure overload. Mechanistically, transplanted MSCs possibly stimulated GDF15 and its downstream SMAD proteins to antagonize the hypertrophy response of pressure overload. These encouraging results have implications in congenital cardiac pressure overload lesions.


Subject(s)
Hypertrophy, Right Ventricular/therapy , Mesenchymal Stem Cell Transplantation , Ventricular Dysfunction, Right/therapy , Ventricular Pressure/physiology , Animals , Disease Models, Animal , Humans , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Myosin Heavy Chains/metabolism , Myosin Light Chains/metabolism , Natriuretic Peptide, Brain/metabolism , Swine , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology , Ventricular Remodeling/physiology
15.
Artif Organs ; 40(11): 1046-1053, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27087252

ABSTRACT

Respiratory failure is one of the major causes of mortality and morbidity all over the world. Therapeutic options to treat respiratory failure remain limited. The objective of this study was to evaluate the gas transfer performance of a newly developed miniature portable integrated pediatric pump-lung device (PediPL) with small membrane surface for respiratory support in an acute ovine respiratory failure model. The respiratory failure was created in six adult sheep using intravenous anesthesia and reduced mechanical ventilation at 2 breaths/min. The PediPL device was surgically implanted and evaluated for respiratory support in a venovenous configuration between the right atrium and pulmonary artery. The hemodynamics and respiratory status of the animals during support with the device gas transfer performance of the PediPL were studied for 4 h. The animals exhibited respiratory failure 30 min after mechanical ventilation was reduced to 2 breaths/min, indicated by low oxygen partial pressure, low oxygen saturation, and elevated carbon dioxide in arterial blood. The failure was reversed by establishing respiratory support with the PediPL after 30 min. The rates of O2 transfer and CO2 removal of the PediPL were 86.8 and 139.1 mL/min, respectively. The results demonstrated that the PediPL (miniature integrated pump-oxygenator) has the potential to provide respiratory support as a novel treatment for both hypoxia and hypercarbia. The compact size of the PediPL could allow portability and potentially be used in many emergency settings to rescue patients suffering acute lung injury.


Subject(s)
Acute Lung Injury/surgery , Extracorporeal Membrane Oxygenation/instrumentation , Heart-Lung Machine , Respiratory Insufficiency/surgery , Animals , Carbon Dioxide/blood , Child , Disease Models, Animal , Heart Atria/surgery , Hemodynamics , Humans , Hypercapnia/therapy , Oxygen/blood , Pulmonary Artery/surgery , Respiration, Artificial/methods , Sheep
16.
Semin Thorac Cardiovasc Surg ; 28(4): 817-824, 2016.
Article in English | MEDLINE | ID: mdl-28417870

ABSTRACT

Clinical protocols for stem cell-based therapies are currently under development for patients with hypoplastic left heart syndrome. An ideal cell delivery method should have minimal safety risks and provide a wide distribution of cells to the nonischemic right ventricle (RV). However, the optimal strategy for stem cell delivery to the RV has yet to be explored in a preclinical model, necessary for a hypoplastic left heart syndrome trial. Human c-kit+ cardiac stem cells (CSCs) were delivered to healthy Yorkshire swine through the proximal right coronary artery with a stop and reflow technique. The effect of premedication with antiarrhythmic (AA) medications in this model was retrospectively reviewed, with the primary outcome of survival 2 hours after infusion. A group underwent CSC delivery to the RV without prophylactic AA medication (no AA, n = 7), whereas the second group was premedicated with a loading dose and intravenous infusion of amiodarone and lidocaine (AA, n = 13). Cardiac biopsies were obtained from each chamber to ascertain the biodistribution of CSCs. Survival was significantly greater in the prophylactic AA group compared with the group without AA (13/13 [100%] vs 1/7 [14.3%], P < 0.0001). Cardiac arrest during balloon inflation was the cause of death in each of the nonmedicated animals. In the premedicated group, 9 (69.2%) pigs experienced transient ST segment changes in the precordial leads during CSC delivery, which resolved spontaneously. Most c-kit+ CSCs were distributed to lateral segments of the RV free wall, consistent with the anatomical course of the right coronary artery (lateral RV, 19.2 ± 1.5 CSCs/field of view vs medial RV, 10.4 ± 1.3 CSCs/field of view, P < 0.0001). Few c-kit+ CSCs were identified in the right atrium, septum, or left ventricle. Prophylactic infusion of AA enhances survival in swine undergoing intracoronary delivery of human c-kit+ CSCs to the RV. Additionally, intracoronary delivery results in a limited biodistribution of c-kit+ CSCs within the RV. Human clinical protocols can be optimized by requiring infusion of AA medications before cell delivery.


Subject(s)
Amiodarone/administration & dosage , Anti-Arrhythmia Agents/administration & dosage , Arrhythmias, Cardiac/prevention & control , Heart Ventricles/surgery , Lidocaine/administration & dosage , Stem Cell Transplantation , Stem Cells , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/physiopathology , Biomarkers/metabolism , Cell Survival , Cells, Cultured , Female , Graft Survival , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Heterografts , Humans , Male , Models, Animal , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Retrospective Studies , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/methods , Stem Cells/metabolism , Stem Cells/pathology , Sus scrofa , Time Factors
17.
J Thorac Cardiovasc Surg ; 150(5): 1332-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26395041

ABSTRACT

OBJECTIVES: Left ventricular (LV) assist device (LVAD) support reduces pathological loading. However, load-induced adaptive responses may be suppressed. Pathological loading dysregulates cardiac G protein-coupled receptor (GPCR) signaling. Signaling through G proteins is deleterious, whereas beta (ß)-arrestin-mediated signaling is cardioprotective. We examined the effects of pathological LV loading/LV dysfunction and treatment via LVAD, on ß-arrestin-mediated signaling, and genetic networks downstream of load. METHODS: An ovine myocardial infarction (MI) model was used. Sheep underwent sham thoracotomy (n = 3), mid-left anterior descending coronary artery ligation to produce MI (n = 3), or MI with placement of a small-platform catheter-based LVAD (n = 3). LVAD support was continued for 2 weeks. Animals were maintained for a total of 12 weeks. Myocardial specimens were harvested and analyzed. RESULTS: MI induced ß-arrestin activation. Increased interactions between epidermal growth factor receptor and ß-arrestins were observed. LVAD support inhibited these responses to MI (P < .05). LVAD support inhibited the activation of cardioprotective signaling effectors Akt (P < .05), and, to a lesser extent, extracellular regulated kinase 1/2 (P not significant); however, MI resulted in regional activation of load-induced GPCR signaling via G proteins, as assessed by the induction of atrial natriuretic peptide mRNA expression in the MI-adjacent zone relative to the MI-remote zone (P < .05). MI-adjacent zone atrial natriuretic peptide expression was renormalized with LVAD support. CONCLUSIONS: LVAD support inhibited cardioprotective ß-arrestin-mediated signaling. However, net benefits of normalization of load-induced GPCR signaling were observed in the MI-adjacent zone. These findings may have implications for the optimal extent and duration of unloading, and for the development of adjunctive medical therapies.


Subject(s)
Cardiac Catheterization/instrumentation , Cardiac Catheters , Heart-Assist Devices , Myocardial Infarction/therapy , Myocardium/metabolism , Signal Transduction , Ventricular Dysfunction, Left/therapy , Ventricular Function, Left , Animals , Arrestins/metabolism , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Materials Testing , Myocardial Infarction/diagnosis , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/pathology , Phosphorylation , Prosthesis Design , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Recovery of Function , Sheep , Time Factors , Ventricular Dysfunction, Left/diagnosis , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , beta-Arrestins
18.
ASAIO J ; 61(2): 196-201, 2015.
Article in English | MEDLINE | ID: mdl-25396272

ABSTRACT

Despite the significant contribution of the Fontan procedure to the therapy of complex congenital heart diseases, many patients progress to failure of their Fontan circulation. The use of ventricular assist devices to provide circulatory support to these patients remains challenging. In the current study, a continuous axial-flow pump was used to support a univentricular Fontan circulation. A modified Fontan circulation (atrio-pulmonary connection) was constructed in six Yorkshire piglets (8-14 kg). A Dacron conduit (12 mm) with two branches was constructed to serve as a complete atrio-pulmonary connection without the use of cardiopulmonary bypass. The Impella pump was inserted into the conduit through an additional Polytetrafluoroethylene (PTFE) graft in five animals. Hemodynamic data were collected for 6 hours under the supported Fontan circulation. The control animal died after initiating the Fontan circulation independent of resuscitation. Four pump supported animals remained hemodynamically stable for 6 hours with pump speeds between 18,000 rpm and 22,000 rpm (P1-P3). Oxygen saturation was maintained between 95% and 100%. Normal organ perfusion was illustrated by blood gas analysis and biochemical assays. A continuous axial-flow pump can be used for temporal circulatory support to the failing Fontan circulation as "bridge" to heart transplantation or recovery.


Subject(s)
Assisted Circulation/instrumentation , Fontan Procedure/instrumentation , Animals , Assisted Circulation/methods , Fontan Procedure/adverse effects , Fontan Procedure/methods , Heart-Assist Devices , Hemodynamics , Lactic Acid/blood , Miniaturization , Models, Animal , Oxygen/blood , Sus scrofa
19.
J Heart Lung Transplant ; 33(8): 857-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24746636

ABSTRACT

BACKGROUND: Device availability of mechanical circulatory or respiratory support to the right heart has been limited. The purpose of this study was to investigate the effect of right heart unloading and respiratory support with a wearable integrated artificial pump-lung (APL). METHODS: The APL device was placed surgically between the right atrium and pulmonary artery in 7 sheep. Anti-coagulation was performed with heparin infusion. The device's ability to unload the right ventricle (RV) was investigated by echocardiograms and right heart catheterization at different bypass flow rates. Hemodynamics and echocardiographic data were evaluated. APL flow and gas transfer rates were also measured at different device speeds. RESULTS: Hemodynamics remained stable during APL support. There was no significant change in systemic blood pressure and cardiac index. Central venous pressure, RV pressure, RV end-diastolic dimension and RV ejection fraction were significantly decreased when APL device flow rate approached 2 liters/min. Linear regression showed significant correlative trends between the hemodynamic and cardiac indices and device speed. The oxygen transfer rate increased with device speed. The oxygen saturation from the APL outlet was fully saturated (>95%) during support. The impact of APL support on blood elements (plasma free hemoglobin and platelet activation) was minimal. CONCLUSIONS: APL device support significantly unloaded the RV with increasing device speed. The device also provided stable hemodynamics and respiratory support in terms of blood flow and oxygen transfer. The right heart unloading performance of this wearable device needs to be evaluated further in an animal model of right heart failure with long-term support.


Subject(s)
Equipment Design , Extracorporeal Membrane Oxygenation/instrumentation , Extracorporeal Membrane Oxygenation/methods , Heart-Assist Devices , Ventricular Dysfunction, Right/surgery , Animals , Disease Models, Animal , Heart Atria/physiopathology , Heart Atria/surgery , Hemodynamics/physiology , Linear Models , Pulmonary Artery/physiopathology , Pulmonary Artery/surgery , Sheep , Stroke Volume/physiology , Treatment Outcome , Ventricular Dysfunction, Right/physiopathology
20.
Acta Biomater ; 10(2): 701-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24185001

ABSTRACT

The body is unable to control massive blood loss without treatment. Available hemostatic agents are often expensive, ineffective or raise safety concerns. Synthetic hydrogel particles are an inexpensive and promising alternative. In this study we synthesized and characterized N-(3-aminopropyl)methacrylamide (APM) hydrogel particles and investigated their use as a hemostatic material. The APM hydrogel particles were synthesized via inverse suspension polymerization with a narrow size distribution and rapid swelling behavior. In vitro coagulation studies showed hydrogel particle blood aggregate formation as well as bulk blood coagulation inhibition. In vivo studies using multiple rat injury and ovine liver laceration models demonstrated the particles' ability to aid in rapid hemostasis. Subsequent hematoxylin and eosin and Carstairs' method staining of the ovine liver incision sites showed significant hemostatic plug formation. This study suggests that these cationic hydrogel particles form a physical barrier to blood loss by forming aggregates, while causing a general decrease in coagulation activity in the bulk. The formation of a rapid sealant through aggregation and the promotion of local hemostasis through electrostatic interactions are coupled with a decrease in overall coagulation activity. These interactions require the interplay of a variety of mechanisms stemming from a simple synthetic platform.


Subject(s)
Blood Coagulation/drug effects , Hemostatics/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Acrylamides/chemistry , Animals , Disease Models, Animal , Hemostatics/chemical synthesis , Hemostatics/chemistry , Hydrogel, Polyethylene Glycol Dimethacrylate/chemical synthesis , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Lacerations/pathology , Liver/pathology , Particle Size , Polyethylene Glycols/chemistry , Punctures , Rats , Rats, Sprague-Dawley , Sheep , Spectroscopy, Fourier Transform Infrared , Tail , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...