101
|
Modulation of acetylation: creating a pro-survival and anti-inflammatory phenotype in lethal hemorrhagic and septic shock. J Biomed Biotechnol 2011; 2011:523481. [PMID: 21403879 PMCID: PMC3051174 DOI: 10.1155/2011/523481] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 11/16/2010] [Indexed: 12/12/2022] Open
Abstract
Histone deacetylases (HDACs) play a key role in homeostasis of protein acetylation in histone and nonhistone proteins and in regulating fundamental cellular activities. In this paper we review and discuss intriguing recent developments in the use of histone deacetylase inhibitors (HDACIs) to combat some critical conditions in an animal model of hemorrhagic and septic shock. HDACIs have neuroprotective, cardioprotective, renal-protective, and anti-inflammatory properties; survival improvements have been significantly shown in these models. We discuss the targets and mechanisms underlying these effects of HDACIs and comment on the potential new clinical applications for these agents in the future. This paper highlights the emerging roles of HDACIs as acetylation modulators in models of hemorrhagic and septic shock and explains some contradictions encountered in previous studies.
Collapse
|
102
|
Causey MW, Hoffer ZS, Miller SL, Huston LJ, Satterly SA, Martin M, Stallings JD. Microarray and functional cluster analysis implicates transforming growth factor beta1 in endothelial cell dysfunction in a swine hemorrhagic shock model. J Surg Res 2011; 170:120-32. [PMID: 21392802 DOI: 10.1016/j.jss.2011.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 12/21/2010] [Accepted: 01/06/2011] [Indexed: 11/26/2022]
Abstract
BACKGROUND Trauma leading to massive hemorrhage results in widespread tissue hypoxia, anaerobic metabolism, and production of inflammatory cytokines and oxidative molecules injurious to the vascular endothelium. Although trauma-related endothelial cell pathophysiology has been extensively studied, very little is known regarding gene transcriptional changes that occur during the event, particularly in endothelia. Thus, we employed fluorescent microarray analysis of gene transcription to elucidate critical pathways and gene products involved in endothelial dysfunction. MATERIALS AND METHODS A trauma-hemorrhage/shock (T-H/S) model mimicking the physiologic changes seen in human trauma was performed on 10 Yorkshire swine, consisting of 35% blood volume hemorrhage followed by 6 h of full resuscitation. Aortic endothelium was analyzed by microarray and functional clusters were identified through the use of Database for Annotation, Visualization, and Integrated Discovery (DAVID) software. RESULTS Injured swine developed profound acidosis, coagulopathy, massive resuscitative fluid requirements, and microscopic changes of ischemia/reperfusion injury. While 1007 transcripts were down-regulated, 529 transcripts were up-regulated. DAVID functional clustering analysis revealed 21 significantly altered biological processes that were grouped into 12 distinct functional categories. The transforming growth factor beta (TGFβ) family of genes was the most interrelated. In addition, vascular endothelial growth factor (VEGF) signaling members and leukocyte chemoattractants were also altered. CONCLUSIONS Our model identified two major signaling pathways, TGFβ and VEGF, which undergo early transcriptional changes in injured endothelial cells. Our results suggest that TGFβ and VEGF may play a crucial role in the development of endothelial cell injury leading to increased vascular permeability during shock-trauma.
Collapse
|
103
|
Effects of methylene blue and volatile anesthetics on survival in a murine hemorrhage resuscitation model. ACTA ACUST UNITED AC 2011; 69:1433-40; discussion 1440-1. [PMID: 21150523 DOI: 10.1097/ta.0b013e3181f8aa11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Hemorrhage is a frequent cause of morbidity and mortality, possibly complicated by volatile anesthetics administered during surgical emergencies. Because methylene blue (MB) was suggested to reduce bleeding, we reasoned that it may improve resuscitation. We used a rat model of controlled and uncontrolled hemorrhage with fluid resuscitation, aiming at high versus low mean arterial pressure (MAP) to assess the role of early MB injection on survival and the effects of different anesthetics on outcome. METHODS Wistar male rats (n = 160) were subjected to 15-minute controlled and 60-minute uncontrolled hemorrhage and received lactated Ringer's solution replacement. Four sets (four groups per set, N = 10 per group) were anesthetized with halothane, isoflurane, sevoflurane, or ketamine (KET; control). Resuscitation-targeted MAP was 80 mm Hg in two groups per set and 40 mm Hg in two groups per set: one group received MB 25 mg/kg intravenously and the other one did not receive. RESULTS All parameters were worse in the higher target groups compared with the lower MAP target groups. MB improved variable outcomes in the treated compared with the nontreated groups, independent of the MAP or anesthesia agent: the amount of replacement volume, lung tissue xanthine oxidase activity, and rats' survival rates. Outcomes with and without MB were worse in the halothane set, followed, in ascending order, by sevoflurane, isoflurane, and KET. CONCLUSIONS MB improved parameters and survival rates after controlled and uncontrolled hemorrhage and fluid resuscitation, even in high MAP-resuscitated rats. KET seemed to be the best anesthetic choice among the four classic agents tested. The effects of balanced anesthesia and total intravenous anesthesia in similar conditions require additional studies.
Collapse
|
104
|
Kim HY, Kim IO, Kim WS, Kang GH. Bowel sonography in sepsis with pathological correlation: an experimental study. Pediatr Radiol 2011; 41:237-43. [PMID: 20734039 DOI: 10.1007/s00247-010-1806-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2009] [Revised: 07/18/2010] [Accepted: 07/26/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Sepsis predisposes full-term infants to necrotizing enterocolitis (NEC). As such, experimental induction of NEC was applied to a sepsis model to evaluate the potential role of US in the early diagnosis of NEC in full-term infants. OBJECTIVE To evaluate the resistive index (RI) of the superior mesenteric artery (SMA) on Doppler sonography in experimentally induced sepsis and correlate it with the pathological findings. MATERIALS AND METHODS Fifteen 1-week-old New Zealand white rabbits (control group n = 3, sepsis group n = 12) were used in this study. We injected 1 mg/kg of E. coli O55-B5 lipopolysaccharide (LPS) into 12 rabbits to induce sepsis. Then we conducted grayscale evaluation of the caliber of the abdominal aorta as well as bowel wall thickness and echogenicity. In addition, we measured peak systolic and end-diastolic velocities and SMA RI on Doppler sonography. Pathological findings were analyzed and correlated with RI readings. Peak systolic and end-diastolic velocities and SMA RI values were analyzed statistically at each hour using the Wilcoxon rank sum test; the control and sepsis groups were compared using the Mann-Whitney test. RESULTS The bowel wall thickness in the sepsis group was significantly increased after LPS injection. The caliber of the abdominal aorta in the sepsis group was significantly decreased after LPS injection. There were echogenic foci (<10 in axial plane) in the bowel wall after LPS injection. Peak systolic velocity in the sepsis group was not significantly changed, but end-diastolic velocity was decreased. SMA RIs in the sepsis group were significantly increased post-LPS injection from baseline. In the control group there were no significant changes in bowel wall thickness, abdominal aorta caliber, bowel wall echogenicity or peak systolic and end-diastolic velocities and RIs. Pathologically, eight of the 12 rabbits in the sepsis group showed grade 1 intestinal injury, three showed grade 2 injury and one showed grade 3 injury. SMA RIs were higher in grades 2 and 3 than in grade 1 when measured at 2 h and 4 h. CONCLUSION Sepsis caused necrotic injury in the animal models, and these findings were accompanied by significant changes on Doppler US. These findings could facilitate early detection of intestinal injury in septic infants with NEC.
Collapse
Affiliation(s)
- Hwa-Young Kim
- Department of Radiology, Seoul National University College of Medicine and Institute of Radiation Medicine, 101 Daehangno, Jongno-Gu, Seoul, 110-744, South Korea
| | | | | | | |
Collapse
|
105
|
Pfeifer R, Kobbe P, Darwiche SS, Billiar TR, Pape HC. Role of hemorrhage in the induction of systemic inflammation and remote organ damage: analysis of combined pseudo-fracture and hemorrhagic shock. J Orthop Res 2011; 29:270-4. [PMID: 20690183 DOI: 10.1002/jor.21214] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study was performed to analyze the role of hemorrhage-induced hypotension in the induction of systemic inflammation and remote organ dysfunction. Male C57/BL6 mice (6- to 10-week old and 20-30 g) were used. Animals were either subjected to pseudo-fracture [PF; standardized soft-tissue injury and injection of crushed bone, PF group: n = 9], or PF combined with hemorrhagic shock (HS + PF group: n = 6). Endpoint was 6 h. Systemic inflammation was assessed by IL-6 and IL-10 levels. Myeloperoxidase (MPO) and NF-κB activity in the lung and liver tissue were obtained to assess remote organ damage. The increases of systemic cytokines are similar for animals subjected to PF and PF + HS (IL-6: 189 pg/ml ± 32.5 vs. 160 pg/ml ± 5.3; IL-10: 60.3 pg/ml ± 15.8 vs. 88 pg/ml ± 32.4). Furthermore, the features (ALT; NF-κB) of liver injury are equally elevated in mice subjected to PF (76.9 U/L ± 4.5) and HS + PF (80 U/L ± 5.5). Lung injury, addressed by MPO activity was more severe in group HS + PF (2.95 ng/ml ± 0.32) than in group PF (1.21 ng/ml ± 0.2). Both PF and additional HS cause a systemic inflammatory response. In addition, hemorrhage seems to be associated with remote affects on the lung.
Collapse
Affiliation(s)
- Roman Pfeifer
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, 3471 Fifth Avenue, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | |
Collapse
|
106
|
Abstract
Patients undergoing emergency surgery typically require resuscitation, either because they are hemorrhaging or because they are experiencing significant internal fluid shifts. Intravascular hypovolemia is common at the time of anesthesia induction and can lead to hemodynamic collapse if not promptly treated. Central pressure monitoring is associated with technical complications and does not improve outcomes in this population. Newer modalities are in use, but they lack validation. Fluid resuscitation is different in bleeding and septic patients. In the former group, it is advisable to maintain a deliberately low blood pressure to facilitate clot formation and stabilization. If massive transfusion is anticipated, blood products should be administered from the outset to prevent the coagulopathy of trauma. Early use of plasma in a ratio approaching 1:1 with red blood cells (RBCs) has been associated with improved outcomes. In septic patients, early fluid loading is recommended. The concept of "goal-directed resuscitation" is based on continuing resuscitation until venous oxygen saturation is normalized. In either bleeding or septic patients, however, the most important goal remains surgical control of the source of pathology, and nothing should be allowed to delay transfer to the operating room. We review the current literature and recommendations for the resuscitation of patients coming for emergency surgery procedures.
Collapse
|
107
|
Mi Q, Li NYK, Ziraldo C, Ghuma A, Mikheev M, Squires R, Okonkwo DO, Verdolini-Abbott K, Constantine G, An G, Vodovotz Y. Translational systems biology of inflammation: potential applications to personalized medicine. Per Med 2010; 7:549-559. [PMID: 21339856 PMCID: PMC3041597 DOI: 10.2217/pme.10.45] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A central goal of industrialized nations is to provide personalized, preemptive and predictive medicine, while maintaining healthcare costs at a minimum. To do so, we must confront and gain an understanding of inflammation, a complex, nonlinear process central to many diseases that affect both industrialized and developing nations. Herein, we describe the work aimed at creating a rational, engineering-oriented and evidence-based synthesis of inflammation geared towards rapid clinical application. This comprehensive approach, which we call 'Translational Systems Biology', to date has been utilized for in silico studies of sepsis, trauma/hemorrhage/traumatic brain injury, acute liver failure and wound healing. This framework has now allowed us to suggest how to modulate acute inflammation in a rational and individually optimized fashion using engineering principles applied to a biohybrid device. We suggest that we are on the cusp of fulfilling the promise of in silico modeling for personalized medicine for inflammatory disease.
Collapse
Affiliation(s)
- Qi Mi
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Sports Medicine & Nutrition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicole Yee-Key Li
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Communication Science & Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cordelia Ziraldo
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Computational Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ali Ghuma
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maxim Mikheev
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert Squires
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - David O Okonkwo
- Department of Neurological Surgery, University of Pittsburgh, PA, USA
| | - Katherine Verdolini-Abbott
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Communication Science & Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gregory Constantine
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Departments of Mathematics & Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gary An
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Yoram Vodovotz
- Center for Inflammation & Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
108
|
Jonker FHW, Mojibian H, Schlösser FJV, Botta DM, Indes JE, Moll FL, Muhs BE. The impact of hypovolaemic shock on the aortic diameter in a porcine model. Eur J Vasc Endovasc Surg 2010; 40:564-71. [PMID: 20727795 DOI: 10.1016/j.ejvs.2010.07.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 07/19/2010] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To investigate the impact of hypovolaemic shock on the aortic diameter in a porcine model, and to determine the implications for the endovascular management of hypovolaemic patients with traumatic thoracic aortic injury (TTAI). MATERIALS AND METHODS The circulating blood volume of seven Yorkshire pigs was gradually lowered in 10% increments. At 40% volume loss, an endograft was deployed in the descending thoracic aorta, followed by gradual fluid resuscitation. Potential changes in aortic diameter during the experiment were recorded using intravascular ultrasound (IVUS). RESULTS The aortic diameter decreased significantly at all evaluated levels during blood loss. The ascending aortic diameter decreased on average with 38% after 40% blood loss (range 24-62%, p = 0.018), the descending thoracic aorta with 32% (range 18-52%, p = 0.018) and the abdominal aorta with 28% (range 15-39%, p = 0.018). The aortic diameters regained their initial size during fluid resuscitation. CONCLUSION The aortic diameter significantly decreases during blood loss in this porcine model. If these changes take place in hypovolaemic TTAI patients as well, it may have implications for thoracic endovascular aortic repair (TEVAR). Increased oversizing of the endograft, or additional computed tomography (CT) or IVUS imaging after fluid resuscitation for more adequate aortic measurements, may be needed in TTAI patients with considerable blood loss.
Collapse
Affiliation(s)
- F H W Jonker
- Section of Vascular Surgery and Interventional Radiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | |
Collapse
|
109
|
Isayama K, Murao Y, Saito F, Hirakawa A, Nakatani T. Effects of hypertonic saline on CD4+CD25+Foxp3+ regulatory T cells after hemorrhagic shock in relation to iNOS and cytokines. J Surg Res 2010; 172:137-45. [PMID: 20888597 DOI: 10.1016/j.jss.2010.07.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 07/16/2010] [Accepted: 06/26/2010] [Indexed: 11/17/2022]
Abstract
BACKGROUND Hemorrhagic shock and resuscitation induce immunosuppression. CD4(+)CD25(+)Foxp3(+) regulatory T Cells (Foxp3(+) Tregs), iNOS and cytokines may affect these severe conditions such as acute respiratory distress syndrome and multiple organ failure after hemorrhagic shock and resuscitation. Foxp3(+) Tregs have been described to be specific and play a key role in the control of the immune system. Immune condition may be restored by hypertonic saline resuscitation that inhibits pro-inflammatory effects of cytokine. Our aim was to investigate how hypertonic saline resuscitation affected Foxp3(+) Tregs after hemorrhagic shock and resuscitation in relation to iNOS and cytokines. METHODS Male C57BL6/J and B6.129P2- NOS2(tm1Lau)/J (iNOS gene knockout) mice were used in creating hemorrhagic shock model. Mice were divided into two groups, each according to the type of resuscitation. (1) Wild HS: resuscitation with hypertonic saline (4 mL/Kg of 7.5% NaCl) and the shed blood (SB); (2) wild 2LR: resuscitation with lactated Ringer's solution and the SB; (3) iNOS knockout HS: similarly resuscitated as wild HS; (4) iNOS knockout 2LR: similarly resuscitated as wild 2LR. Samples of thymus and spleen were harvested at 2, 6, 24, 48, and 72 h after resuscitation. CD4(+) T cells and Foxp3(+) Tregs were analyzed at 24, 48, and 72 h. At 2, 6, 24, and 48 h, plasma cytokines were assayed and expression of iNOS (NOS2) was also measured by immunofluorescence. RESULTS NOS2 of HS and 2LR wild groups at 2 and 6 h in spleen increased compared with the control group. At 6h, NOS2 in HS wild group was significantly lower than in 2LR wild group. Plasma levels of interleukin (IL)-6, TNF- α, MCP-1, and IL-10 increased at 2 h. Both in wild type and iNOS knockout mice, hypertonic saline resuscitation decreased plasma IL-6, TNF-α, and MCP-1 levels at 2 h; CD4(+) T cells in spleen and thymus decreased at 24, 48, and 72 h, and Foxp3(+) Tregs in spleen at 48 h increased, however, hypertonic saline resuscitation did not affect the Foxp3(+). CONCLUSIONS These results show that in early phase, the inflammatory cytokines in plasma might affect iNOS expression and cytokines. Further, this study showed that hypertonic saline resuscitation and suppression of iNOS might improve immunosuppressive reaction after hemorrhagic shock.
Collapse
Affiliation(s)
- Kenji Isayama
- Department of Emergency and Critical Care Medicine, Kansai Medical University, Osaka, Japan
| | | | | | | | | |
Collapse
|
110
|
Abstract
Pathophysiology of multiple trauma is characterized by different trauma-associated repercussions like organ destruction, haemorrhage, immune cell activation by foreign antigen, for example. The length of time while such impairments take hold of the organism substantially impacts the extent of the post trauma secondary injury. Short interruptions in microcirculation can mostly be compensated, whereas elongated ischemic periods definitely cause structural cell damage up to death. The current review highlights the importance of the time duration of posttrauma second hits on the pathophysiology of systemic inflammation and multiple organ failure. The quick termination of such secondary impairments by immediate therapeutic intervention mainly impacts the patients' prognosis.
Collapse
|
111
|
Wan Z, Sun S, Ristagno G, Weil MH, Tang W. The cerebral microcirculation is protected during experimental hemorrhagic shock. Crit Care Med 2010; 38:928-32. [PMID: 20068466 DOI: 10.1097/ccm.0b013e3181cd100c] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Decreases in buccal microcirculation are indicative of the severity of hemorrhage, but incidental observations suggest that this may not apply to the cerebral microcirculation. We therefore hypothesized that the cerebral microcirculation may be preserved in hemorrhagic shock in which systemic and buccal microcirculatory flow are reduced. We propose to relate changes in the macrocirculation to the buccal and cerebral microcirculations during hemorrhage and after fluid resuscitation. DESIGN Prospective, randomized, controlled animal study. SETTING University-affiliated research laboratory. SUBJECTS Sprague-Dawley rats. INTERVENTIONS Fifteen male Sprague-Dawley rats were anesthetized and endotracheally intubated. Craniotomy exposed the parietal cortex for orthogonal polarization spectral imaging. Mean arterial pressure, cardiac output, arterial blood gases, and lactate were measured concurrently with determination of microcirculatory indices in buccal and cerebral areas. Animals were randomly assigned to bleeding either 35% or 25% of estimated total blood volume and compared with sham bled animals. Hypovolemia was maintained for 60 mins in test animals, after which saline in amounts to 2 times the blood loss, was administered over 30 mins. Cerebral and buccal microvascular indices were measured in vessels smaller than 20 mum, representing capillaries. MEASUREMENTS AND MAIN RESULTS Mean arterial pressure and cardiac output were reduced and arterial blood lactate was increased in relationship to the magnitude of blood loss. Saline infusion increased mean arterial pressure and cardiac output. Buccal microcirculation decreased after bleeding but was restored after saline infusion. However, the cerebral microcirculation was essentially unaffected by hemorrhage and saline infusion. CONCLUSION In contrast to the systemic decreases in pressure and flow characteristics of hemorrhagic shock, including decreases in microcirculations of buccal mucosa, cerebral microvascular flow was preserved during moderate and severe blood losses.
Collapse
Affiliation(s)
- Zhi Wan
- Weil Institute of Critical Care Medicine, Rancho Mirage, CA, USA
| | | | | | | | | |
Collapse
|
112
|
Vasopressin use is associated with death in acute trauma patients with shock. J Crit Care 2010; 25:173.e9-14. [DOI: 10.1016/j.jcrc.2009.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 04/06/2009] [Accepted: 05/05/2009] [Indexed: 11/21/2022]
|
113
|
Abstract
BACKGROUND Tight junction breakdown, with loss of the important sealing protein claudin-3, is an early event in the development of intestinal damage. Therefore, noninvasive analysis of intestinal tight junction status could be helpful in early detection of intestinal injury. AIM To investigate the usefulness of urinary claudin-3 as marker for intestinal tight junction loss. METHODS A rat hemorrhagic shock model and a human setting of known intestinal damage, that is, patients with relapsed inflammatory bowel disease (IBD), were used to investigate intestinal tight junction status by immunohistochemical staining and urinary claudin-3 levels by western blot. RESULTS In rats claudin-3 urine levels increased rapidly after histologically proven intestinal tight junction loss, with significantly elevated levels at 90 minutes after shock compared with sham-operated animals [mean+/-SEM: 611+/-101 intensity (INT), n=6 vs. 232+/-30 INT, n=6; P<0.05]. Moreover, in colonic biopsies of patients with IBD relapse claudin-3 staining was reduced compared with biopsies of patients with IBD without signs of disease. Concomitantly, significantly increased claudin-3 urine levels were found in these patients (502+/-67 INT, n=10) compared with patients with IBD in remission (219+/-17 INT, n=10, P<0.001) and healthy volunteers (225+/-38 INT, n=10, P<0.001). CONCLUSION Here we show for the first time in both an experimental and clinical setting a strong relation between intestinal tight junction loss and urinary claudin-3 levels. These findings suggest that measurement of urinary claudin-3 can be used as noninvasive marker for intestinal tight junction loss. This offers new opportunities for early diagnosis and follow-up of intestinal injury.
Collapse
|
114
|
Attenuation of the effects of rat hemorrhagic shock with a reperfusion injury-inhibiting agent specific to mice. Shock 2009; 32:295-301. [PMID: 19106810 DOI: 10.1097/shk.0b013e3181995e0c] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Death after hemorrhagic shock (HS) may be caused by a generalized reperfusion injury, particularly noticeable in the gut. A period of tissue ischemia followed by reinstitution of perfusion produces severe inflammation that can be blocked in mice by preventing the binding of a pathogenic natural immunoglobulin M (IgM) of defined specificity to antigens in reperfused tissue by using a soluble peptide analogue of the IgM tissue target. We hypothesize that this agent can improve end points of rat HS: death, intestinal injury, and lung injury. Male Sprague-Dawley rats were anesthetized; 50% of calculated blood volume was removed for 120 min, shed blood, then returned; and animals were sacrificed at 72 h. One group of rats received i.v. analogue ([N2] 300 microg) with the return of shed blood. Small intestine and lung were evaluated by histological examination and immunohistochemistry. Lung edema was assessed by Evans blue extravasation and histological examination. I.v. N2 decreased experimental mortality from 62% to 12% (P < 0.05). Associated with this was diminution of gut injury score from 57.8% +/- 5.5% to 19.5% +/- 2.5% (P < 0.05), lung injury from 21.4 +/- 1.5 to 14.8 +/- 1.3 polymorphonuclear leucocytes per high-power field x400 (P < 0.05), and Evans blue extravasation index from 0.61 +/- 0.14 to 0.18 +/- 0.06 (P < 0.05). As well, the deposition of IgM and C3 that is seen in intestinal villi from HS was not present in N2-treated rats. The N2 peptide agent that blocks reperfusion injury in mice prevents death from rat HS, as well as attenuates gut reperfusion injury and its remote target injuries. These data suggest that death from HS is caused by reperfusion injury, and that an agent derived from mice is effective in rats when given in real therapeutic time.
Collapse
|
115
|
Namas R, Ghuma A, Torres A, Polanco P, Gomez H, Barclay D, Gordon L, Zenker S, Kim HK, Hermus L, Zamora R, Rosengart MR, Clermont G, Peitzman A, Billiar TR, Ochoa J, Pinsky MR, Puyana JC, Vodovotz Y. An adequately robust early TNF-alpha response is a hallmark of survival following trauma/hemorrhage. PLoS One 2009; 4:e8406. [PMID: 20027315 PMCID: PMC2794373 DOI: 10.1371/journal.pone.0008406] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 10/22/2009] [Indexed: 12/31/2022] Open
Abstract
Background Trauma/hemorrhagic shock (T/HS) results in cytokine-mediated acute inflammation that is generally considered detrimental. Methodology/Principal Findings Paradoxically, plasma levels of the early inflammatory cytokine TNF-α (but not IL-6, IL-10, or NO2-/NO3-) were significantly elevated within 6 h post-admission in 19 human trauma survivors vs. 4 non-survivors. Moreover, plasma TNF-α was inversely correlated with Marshall Score, an index of organ dysfunction, both in the 23 patients taken together and in the survivor cohort. Accordingly, we hypothesized that if an early, robust pro-inflammatory response were to be a marker of an appropriate response to injury, then individuals exhibiting such a response would be predisposed to survive. We tested this hypothesis in swine subjected to various experimental paradigms of T/HS. Twenty-three anesthetized pigs were subjected to T/HS (12 HS-only and 11 HS + Thoracotomy; mean arterial pressure of 30 mmHg for 45–90 min) along with surgery-only controls. Plasma obtained at pre-surgery, baseline post-surgery, beginning of HS, and every 15 min thereafter until 75 min (in the HS only group) or 90 min (in the HS + Thoracotomy group) was assayed for TNF-α, IL-6, IL-10, and NO2-/NO3-. Mean post-surgery±HS TNF-α levels were significantly higher in the survivors vs. non-survivors, while non-survivors exhibited no measurable change in TNF-α levels over the same interval. Conclusions/Significance Contrary to the current dogma, survival in the setting of severe, acute T/HS appears to be associated with an immediate increase in serum TNF-α. It is currently unclear if this response was the cause of this protection, a marker of survival, or both. This abstract won a Young Investigator Travel Award at the SHOCK 2008 meeting in Cologne, Germany.
Collapse
Affiliation(s)
- Rajaie Namas
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ali Ghuma
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Andres Torres
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Patricio Polanco
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hernando Gomez
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Derek Barclay
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Lisa Gordon
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Sven Zenker
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hyung Kook Kim
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Linda Hermus
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Matthew R. Rosengart
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Gilles Clermont
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Andrew Peitzman
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Juan Ochoa
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Michael R. Pinsky
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Juan Carlos Puyana
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Center for Inflammation and Regenerative Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
116
|
Mathematical modeling of posthemorrhage inflammation in mice: studies using a novel, computer-controlled, closed-loop hemorrhage apparatus. Shock 2009; 32:172-8. [PMID: 19008782 DOI: 10.1097/shk.0b013e318193cc2b] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hemorrhagic shock (HS) elicits a global acute inflammatory response, organ dysfunction, and death. We have used mathematical modeling of inflammation and tissue damage/dysfunction to gain insight into this complex response in mice. We sought to increase the fidelity of our mathematical model and to establish a platform for testing predictions of this model. Accordingly, we constructed a computerized, closed-loop system for mouse HS. The intensity, duration, and time to achieve target MAP could all be controlled using a software. Fifty-four male C57/black mice either were untreated or underwent surgical cannulation. The cannulated mice were divided into 8 groups: (a) 1, 2, 3, or 4 h of surgical cannulation alone and b) 1, 2, 3, or 4 h of cannulation + HS (25 mmHg). MAP was sustained by the computer-controlled reinfusion and withdrawal of shed blood within +/-2 mmHg. Plasma was assayed for the cytokines TNF, IL-6, and IL-10 as well as the NO reaction products NO2-/NO3-. The cytokine and NO2-/NO3- data were compared with predictions from a mathematical model of post-hemorrhage inflammation, which was calibrated on different data. To varying degrees, the levels of TNF, IL-6, IL-10, and NO2/NO3 predicted by the mathematical model matched these data closely. In conclusion, we have established a hardware/software platform that allows for highly accurate, reproducible, and mathematically predictable HS in mice.
Collapse
|
117
|
Namas R, Ghuma A, Hermus L, Zamora R, Okonkwo DO, Billiar TR, Vodovotz Y. The acute inflammatory response in trauma / hemorrhage and traumatic brain injury: current state and emerging prospects. Libyan J Med 2009; 4:97-103. [PMID: 21483522 PMCID: PMC3066737 DOI: 10.4176/090325] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Traumatic injury/hemorrhagic shock (T/HS) elicits an acute inflammatory response that may result in death. Inflammation describes a coordinated series of molecular, cellular, tissue, organ, and systemic responses that drive the pathology of various diseases including T/HS and traumatic brain injury (TBI). Inflammation is a finely tuned, dynamic, highly-regulated process that is not inherently detrimental, but rather required for immune surveillance, optimal post-injury tissue repair, and regeneration. The inflammatory response is driven by cytokines and chemokines and is partially propagated by damaged tissue-derived products (Damage-associated Molecular Patterns; DAMP's). DAMPs perpetuate inflammation through the release of pro-inflammatory cytokines, but may also inhibit anti-inflammatory cytokines. Various animal models of T/HS in mice, rats, pigs, dogs, and non-human primates have been utilized in an attempt to move from bench to bedside. Novel approaches, including those from the field of systems biology, may yield therapeutic breakthroughs in T/HS and TBI in the near future.
Collapse
|
118
|
Klemcke HG, Ryan KL, Britton SL, Koch LG, Dubick MA, Convertino VA. Rat strains bred for low and high aerobic running capacity do not differ in their survival time to hemorrhage. Exp Biol Med (Maywood) 2009; 234:1503-10. [PMID: 19657068 DOI: 10.3181/0812-rm-355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Hemorrhagic shock reflects low tissue perfusion that is inadequate to maintain normal metabolic functions. Often associated with this condition are impairments in cellular oxygen delivery and utilization. Rat strains divergent in their running endurance have been artificially selected over 12 generations. As these rats bred for high (HCR) vs low (LCR) aerobic running capacity have greater tissue O(2) utilization capacity and improved cardiovascular function, we hypothesized that HCR would be more tolerant (i.e., have greater survivability) to the global ischemia of hemorrhagic shock than LCR. To address this hypothesis, survival time to a severe-as substantiated by dramatic changes in plasma lactate, HCO(3), and base deficit-controlled hemorrhage was measured. Male rats were catheterized and, approximately 24 h later, an estimated >35% of the calculated blood volume was removed during a 26-min period while the rats were conscious and unrestrained. Rats were observed for 6 h or until death. Contrary to our hypothesis, survival time in HCR (220 +/- 63 min; n = 6) did not differ statistically (P = 0.46) from that in LCR (279 +/- 53 min; n = 7). Similarly, there were no statistical differences (P >or= 0.08) between rat lines in blood pH, lactate, HCO(3), and base deficit pre- or post-hemorrhage. In addition, few significant differences between lines in response to hemorrhage were detected by measures of cellular antioxidant status in heart, liver, or lung. Since animals with genetically greater tissue oxygen utilization capacity failed to show longer survival times, our results suggest that other mechanisms must play a more dominant role in determining survivability to hemorrhage under conditions of this hemorrhage.
Collapse
Affiliation(s)
- Harold G Klemcke
- U.S. Army Institute of Surgical Research, 3400 Rawley E. Chambers Avenue, Fort Sam Houston, TX 78234, USA.
| | | | | | | | | | | |
Collapse
|
119
|
Dyson A, Stidwill R, Taylor V, Singer M. The impact of inspired oxygen concentration on tissue oxygenation during progressive haemorrhage. Intensive Care Med 2009; 35:1783-91. [DOI: 10.1007/s00134-009-1577-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 05/21/2009] [Indexed: 11/28/2022]
|
120
|
Schmid-Schönbein GW. 2008 Landis Award lecture. Inflammation and the autodigestion hypothesis. Microcirculation 2009; 16:289-306. [PMID: 19384726 DOI: 10.1080/10739680902801949] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although long recognized in microvascular research, an increasing body of evidence suggests that inflammatory markers are present in human diseases. Since the inflammatory cascade serves as a repair mechanism, the presence of inflammatory markers in patient groups has raised an important question about the mechanisms that initiate the inflammatory cascade (i.e., the mechanisms that cause tissue injury). Using a severe form of inflammation, shock, and multiorgan failure, for which there is no accepted injury mechanism, we summarize studies that suggest that the powerful pancreatic digestive enzymes play a central role in the destruction of the intestine and other tissues if their compartmentalization in the lumen of the intestine and in the pancreas is compromised. Further, we summarize evidence that uncontrolled degrading enzyme activity in plasma causes proteolytic cleavage of the extracellular domain of membrane receptors and loss of associated cell functions. For example, in a model of metabolic disease with type II diabetes, proteolytic cleavage of the insulin receptor causes the inability of insulin to signal glucose transport across membranes. The evidence suggests that uncontrolled proteolytic and lipolytic enzyme activity may trigger the mechanism for tissue injury. The significance of such mechanisms remain to be explored in human diseases.
Collapse
Affiliation(s)
- Geert W Schmid-Schönbein
- Department of Bioengineering, University of California-San Diego, La Jolla, California 92093-0412, USA.
| |
Collapse
|
121
|
Recipient treatment with L-arginine attenuates donor lung injury associated with hemorrhagic shock. Transplantation 2009; 87:1602-8. [PMID: 19502950 DOI: 10.1097/tp.0b013e3181a52ce1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Organ donors are frequently trauma victims, but the impact of donor hemorrhagic shock and resuscitation (HSR) on pulmonary graft function has not been assessed. L-arginine treatment during reperfusion increases the production of endothelial nitric oxide and thus ameliorates ischemia-reperfusion injury. Objective of the present porcine study was to investigate the effect of donor hemorrhage on pulmonary graft function and potential beneficial effects of L-arginine administration. METHODS In the control-group (n=6), lungs were harvested from donors without hypotensive periods. In the HSR-group (n=6) and HSR-Arg-group (n=6), donors were subjected to hemorrhagic shock (40% blood shed) and resuscitation before harvest. Left lungs were transplanted after hypothermic preservation of 18 hr, and graft function was observed for 6 hr after reperfusion. Recipients in the HSR-Arg-group received a bolus of L-arginine (50 mg/kg BW) intravenously 5 min before reperfusion followed by a continuous intravenous administration of L-arginine 200 mg/kg BW for 2 hr. Tissue specimens and bronchoalveolar lavage fluid were obtained at the end of the observation period. RESULTS Donor lung function did not differ between study groups. Compared with the control group, pulmonary graft gas exchange was significantly impaired in the HSR-group. Graft function in the HSR-Arg-group did not differ from control organs. Neutrophil fraction, protein content, and malondialdehyde levels in the bronchoalveolar lavage fluid in the HSR-group were higher compared with control and HSR-Arg-Group. CONCLUSION Although fulfilling ideal donor criteria, pulmonary graft function of lungs harvested from donors subjected to HSR is impaired, but improves significantly when l-arginine is administered during reperfusion.
Collapse
|
122
|
Toll-like receptor 4 regulates heme oxygenase-1 expression after hemorrhagic shock induced acute lung injury in mice: requirement of p38 mitogen-activated protein kinase activation. Shock 2009; 31:486-92. [PMID: 18827744 DOI: 10.1097/shk.0b013e318188f7e1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Acute lung injury (ALI) leading to respiratory distress is a common sequela of shock or trauma. The toll-like receptors (TLRs) stand at the interface of innate immune activation in the settings of both infection and sterile injury by responding to a variety of microbial and endogenous ligands alike. This work explored the effects of TLR-4 on hemorrhage-induced ALI and characterizes the signaling pathways and the mechanisms involved in noninfectious ALI. Mice underwent hemorrhagic shock and resuscitation (HSR). Arterial blood gases; expressions of TLR-4, heme oxygenase 1 (HO-1), and p38 mitogen-activated protein kinase (p38MAPK); myeloperoxidase activity; lung wet/dry ratios; and IL-10 levels in lung tissues were obtained at 6, 24, and 48 h after HSR. Hemorrhagic shock and resuscitation induced significant expressions of TLR-4, HO-1, and p38MAPK in C3H/HeN mice. IL-10 and myeloperoxidase were markedly increased at 24 h after HSR, and C3H/HeN mice had ALI with PaO2/fraction of inspired oxygen less than 300 mmHg. The induced amount of each cytokine level and the expressions of TLR-4, HO-1, and p38MAPK of C3H/HeN mice were significantly higher compared with C3H/HeJ mice. This study demonstrated that lung p38MAPK is activated after HSR, and p38MAPK inhibitor FR167653 suppresses HO-1 induction after ALI. We concluded that TLR-4 might induce HO-1 messenger RNA expression, which is probably involved in p38MAPK activation in the development of the lung dysfunction after HSR.
Collapse
|
123
|
Abstract
Hemorrhagic shock (HS) leads to intestinal barrier loss, causing systemic inflammation, which in turn can ultimately lead to multiorgan dysfunction syndrome. Barrier function is based on tight junctions (TJs) between intact epithelial cells. These TJs are anchored in the cell via the filamentous actin (F-actin) cytoskeleton. We hypothesize that HS causes hypoperfusion, leading to loss of F-actin, via activation of actin-depolymerizing factor/cofilin (AC), and consequently TJ loss. This study is aimed at unraveling the changes in cytoskeleton and TJ integrity after HS in organs commonly affected in multiorgan dysfunction syndrome (liver, kidney, and intestine) and to elucidate the events preceding cytoskeleton loss. Adult rats were subjected to a nonlethal HS and sacrificed, along with unshocked controls, at 15, 30, 60, and 90 min after induction of shock. Cytoskeleton, TJ integrity loss, and its consequences were studied by assessment of globular actin, F-actin, AC, zonula occludens protein 1, claudin 3, and bacterial translocation. In the liver and kidney, TJ and the F-actin cytoskeleton remained intact at all time points studied. However, in the intestine, significant loss of F-actin and increase of globular actin was seen from 15 min after shock. This change preceded statistically significant loss of the TJ proteins claudin 3 and zonula occludens protein 1, which were observed starting at 60 min after induction of shock (P < 0.05 vs. controls). Early after induction of shock (15 and 30 min) the nonactive AC (phosphorylated AC) in the intestine was significantly decreased (by 21% and 27%, P < 0.05 vs. control), whereas total AC remained constant, reflecting an increase in activated AC in the intestine from 15 min after shock. Bacterial translocation to mesenteric lymph nodes, liver, and spleen was present from 30 min after shock. This study shows for the first time that HS results in AC activation, selective intestinal actin cytoskeleton disruption, and TJ loss very early after the onset of shock. Loss of this intestinal barrier results in translocation of toxins and bacteria, which enhances inflammation and leads to infections.
Collapse
|
124
|
Chung AJ, Huh YS, Erickson D. A robust, electrochemically driven microwell drug delivery system for controlled vasopressin release. Biomed Microdevices 2009; 11:861-7. [PMID: 19353273 DOI: 10.1007/s10544-009-9303-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
125
|
Zhang Y, Liang ZY, Zhang SY, Huang FF, Wu W, Gao Y, Chen ZB. Albumin resuscitation protects against traumatic/hemorrhagic shock-induced lung apoptosis in rats. J Zhejiang Univ Sci B 2009; 9:871-8. [PMID: 18988306 DOI: 10.1631/jzus.b0820130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To determine the effects of albumin administration on lung injury and apoptosis in traumatic/hemorrhagic shock (T/HS) rats. METHODS Studies were performed on an in vivo model of spontaneously breathing rats with induced T/HS; the rats were subjected to femur fracture, ischemia for 30 min, and reperfusion for 20 min with Ringer's lactate solution (RS) or 5% (w/v) albumin (ALB), and the left lower lobes of the lungs were resected. RESULTS Albumin administered during reperfusion markedly attenuated injury of the lung and decreased the concentration of lactic acid and the number of in situ TdT-mediated dUTP nick-end labelling (TUNEL)-positive cells. Moreover, immunohistochemistry performed 24 h after reperfusion revealed increases in the level of nuclear factor kappaB (NF-kappaB), and phosphorylated p38 mitogen-activated protein kinase (MAPK) in the albumin-untreated group was down-regulated by albumin treatment when compared with the sham rats. CONCLUSION Resuscitation with albumin attenuates tissue injury and inhibits T/HS-induced apoptosis in the lung via the p38 MAPK signal transduction pathway that functions to stimulate the activation of NF-kappaB.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Emergency Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | | | | | | | | | | | | |
Collapse
|
126
|
Metukuri MR, Beer-Stolz D, Namas RA, Dhupar R, Torres A, Loughran PA, Jefferson BS, Tsung A, Billiar TR, Vodovotz Y, Zamora R. Expression and subcellular localization of BNIP3 in hypoxic hepatocytes and liver stress. Am J Physiol Gastrointest Liver Physiol 2009; 296:G499-509. [PMID: 19147804 PMCID: PMC2660177 DOI: 10.1152/ajpgi.90526.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We have previously demonstrated that the Bcl-2/adenovirus EIB 19-kDa interacting protein 3 (BNIP3), a cell death-related member of the Bcl-2 family, is upregulated in vitro and in vivo in both experimental and clinical settings of redox stress and that nitric oxide (NO) downregulates its expression. In this study we sought to examine the expression and localization of BNIP3 in murine hepatocytes and in a murine model of hemorrhagic shock (HS) and ischemia-reperfusion (I/R). Freshly isolated mouse hepatocytes were exposed to 1% hypoxia for 6 h followed by reoxygenation for 18 h, and protein was isolated for Western blot analysis. Hepatocytes grown on coverslips were fixed for localization studies. Similarly, livers from surgically cannulated C57Bl/6 mice and from mice cannulated and subjected to 1-4 h of HS were processed for protein isolation and Western blot analysis. In hepatocytes, BNIP3 was expressed constitutively but was upregulated under hypoxic conditions, and this upregulation was countered by treatment with a NO donor. Surprisingly, BNIP3 was localized in the nucleus of normoxic hepatocytes, in the cytoplasm following hypoxia, and again in the nucleus following reoxygenation. Upregulation of BNIP3 partially required p38 MAPK activation. BNIP3 contributed to hypoxic injury in hepatocytes, since this injury was diminished by knockdown of BNIP3 mRNA. Hepatic BNIP3 was also upregulated in two different models of liver stress in vivo, suggesting that a multitude of inflammatory stresses can lead to the modulation of BNIP3. In turn, the upregulation of BNIP3 appears to be one mechanism of hepatocyte cell death and liver damage in these settings.
Collapse
Affiliation(s)
- Mallikarjuna R. Metukuri
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Donna Beer-Stolz
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rajaie A. Namas
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rajeev Dhupar
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Andres Torres
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Patricia A. Loughran
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Bahiyyah S. Jefferson
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Allan Tsung
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Timothy R. Billiar
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yoram Vodovotz
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ruben Zamora
- Departments of Surgery and Medicine and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
127
|
Khodoun M, Strait R, Orekov T, Hogan S, Karasuyama H, Herbert DR, Köhl J, Finkelman FD. Peanuts can contribute to anaphylactic shock by activating complement. J Allergy Clin Immunol 2009; 123:342-51. [PMID: 19121857 DOI: 10.1016/j.jaci.2008.11.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 11/07/2008] [Accepted: 11/10/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Peanut allergy is the most common food-related cause of lethal anaphylaxis and, unlike other food allergies, typically persists into adulthood. Resistance to digestion and dendritic cell activation by the major peanut allergen Ara h 1 are reported to contribute to its allergenicity. OBJECTIVE We sought to evaluate whether peanut molecules might also promote anaphylaxis through an innate immune mechanism. METHODS Naive mice were treated with a beta-adrenergic receptor antagonist and long-acting IL-4 to increase sensitivity to vasoactive mediators and injected with peanut extract (PE). Shock was detected and quantified by means of rectal thermometry. Gene-deficient mice and specific antagonists were used to determine the roles of specific cell types, complement, Fc receptors, and vasoactive mediators in shock pathogenesis. RESULTS PE induces dose-dependent shock. PE activates complement in vivo in mice and in vitro in mice and human subjects. C3a and, to a lesser extent, stimulatory immunoglobulin receptors contribute to PE-induced shock. PE-induced shock depends more on macrophages and basophils than on mast cells. Platelet-activating factor and, to a lesser extent, histamine contribute to PE-induced shock. PE induces shock in the absence of the adaptive immune system. LPS contamination is not responsible for PE-induced shock. PE and IgE-mediated mast cell degranulation synergistically induce shock. Tree nuts have similar effects to PE, and skim milk and egg white do not. CONCLUSION Peanuts can contribute to shock by causing production of C3a, which stimulates macrophages, basophils, and mast cells to produce platelet-activating factor and histamine.
Collapse
Affiliation(s)
- Marat Khodoun
- Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio; Division of Immunology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Abstract
Abstract This is a review paper that provides an overview of current information on programmed cell death in haemorrhagic shock, including the identification of different molecular receptor signals. A PubMed search for all dates was undertaken using the search terms apoptosis, trauma and haemorrhagic shock. Original research, sentinel and review papers from peer-reviewed journals were included for identification of key concepts. Haemorrhagic shock remains a primary cause of death in civilian and military trauma. Apoptosis is accelerated following haemorrhagic shock. Many methods are used to detect and quantify apoptosis. Fluid resuscitation regimens vary in their effect on the extent of apoptosis. Investigators are examining the effects of haemorrhagic shock and fluid resuscitation on apoptotic signalling pathways. Molecular information is becoming available and being applied to the care of patients experiencing haemorrhagic shock, making it essential for nurses and other health care providers to consider the mechanisms and consequences of apoptosis.
Collapse
Affiliation(s)
- William J Mach
- School of Nursing, University of Kansas, Kansas City, Kansas, USA
| | - Amanda R Knight
- School of Nursing, University of Kansas, Kansas City, Kansas, USA
| | - James A Orr
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
| | - Janet D Pierce
- School of Nursing, University of Kansas, Kansas City, Kansas, USA
| |
Collapse
|
129
|
Takahashi T, Shimizu H, Morimatsu H, Maeshima K, Inoue K, Akagi R, Matsumi M, Katayama H, Morita K. Heme Oxygenase-1 is an Essential Cytoprotective Component in Oxidative Tissue Injury Induced by Hemorrhagic Shock. J Clin Biochem Nutr 2008; 44:28-40. [PMID: 19177185 PMCID: PMC2613496 DOI: 10.3164/jcbn.08-210-ho] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 08/27/2008] [Indexed: 01/11/2023] Open
Abstract
Hemorrhagic shock causes oxidative stress that leads to tissue injuries in various organs including the lung, liver, kidney and intestine. Excess amounts of free heme released from destabilized hemoproteins under oxidative conditions might constitute a major threat because it can catalyze the formation of reactive oxygen species. Cells counteract this by rapidly inducing the rate-limiting enzyme in heme breakdown, heme oxygenase-1 (HO-1), which is a low-molecular-weight stress protein. The enzymatic HO-1 reaction removes heme. As such, endogenous HO-1 induction by hemorrhagic shock protects tissues from further degeneration by oxidant stimuli. In addition, prior pharmacological induction of HO-1 ameliorates oxidative tissue injuries induced by hemorrhagic shock. In contrast, the deletion of HO-1 expression, or the chemical inhibition of increased HO activity ablated the beneficial effect of HO-1 induction, and exacerbates tissue damage. Thus, HO-1 constitutes an essential cytoprotective component in hemorrhagic shock-induced oxidative tissue injures. This article reviews recent advances in understanding of the essential role of HO-1 in experimental models of hemorrhagic shock-induced oxidative tissue injuries with emphasis on the role of its induction in tissue defense.
Collapse
Affiliation(s)
- Toru Takahashi
- Department of Anesthesiology and Resuscitology, Okayama University Medical School, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Chen J, Yang S, Hu S, Choudhry MA, Bland KI, Chaudry IH. Estrogen prevents intestinal inflammation after trauma-hemorrhage via downregulation of angiotensin II and angiotensin II subtype I receptor. Am J Physiol Gastrointest Liver Physiol 2008; 295:G1131-7. [PMID: 18832446 PMCID: PMC2584827 DOI: 10.1152/ajpgi.90443.2008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although angiotensin II (Ang II) plays a key role in development of organ ischemia-reperfusion injury, it remains unclear whether it is involved in development of intestinal injury following trauma-hemorrhage (T-H). Studies have shown that 17beta-estradiol (E2) administration following T-H improves small intestinal blood flow; however, it is unclear whether Ang II plays a role in this E2-mediated salutary effect. Male Sprague-Dawley rats underwent laparotomy and hemorrhagic shock (removal of 60% total blood volume, fluid resuscitation after 90 min). At onset of resuscitation, rats were treated with vehicle, E2, or E2 and estrogen receptor antagonist ICI 182,780 (ICI). A separate group of rats was treated with Ang II subtype I receptor (AT1R) antagonist losartan. At 24 h after T-H, plasma Ang II, IL-6, TNF-alpha, intercellular adhesion molecule (ICAM)-1, cytokine-induced neutrophil chemoattractant (CINC)-1 and CINC-3 levels, myeloperoxidase (MPO) activity, and AT1R expression were determined. T-H significantly increased plasma and intestinal Ang II, IL-6, TNF-alpha levels, intestinal ICAM-1, CINC-1, CINC-3 levels, MPO activity, and AT1R protein compared with shams. E2 treatment following T-H attenuated increased intestinal MPO activity, Ang II level, and AT1R protein expression. ICI administration abolished the salutary effects of E2. In contrast, losartan administration attenuated increased MPO activity without affecting Ang II and AT1R levels. Thus Ang II plays a role in producing small intestine inflammation following T-H, and the salutary effects of E2 on intestinal inflammation are mediated in part by Ang II and AT1R downregulation.
Collapse
Affiliation(s)
- Jianguo Chen
- Department of Surgery, Center for Surgical Research, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shaolong Yang
- Department of Surgery, Center for Surgical Research, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shunhua Hu
- Department of Surgery, Center for Surgical Research, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mashkoor A. Choudhry
- Department of Surgery, Center for Surgical Research, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kirby I. Bland
- Department of Surgery, Center for Surgical Research, University of Alabama at Birmingham, Birmingham, Alabama
| | - Irshad H. Chaudry
- Department of Surgery, Center for Surgical Research, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
131
|
de Haan JJ, Lubbers T, Hadfoune M, Luyer MD, Dejong CH, Buurman WA, Greve JWM. Postshock intervention with high-lipid enteral nutrition reduces inflammation and tissue damage. Ann Surg 2008; 248:842-848. [PMID: 18948813 DOI: 10.1097/sla.0b013e318188752c] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To investigate the effects of high-lipid enteral nutrition in a setting of developing inflammation and tissue damage. BACKGROUND An excessive inflammatory response following severe trauma is associated with poor clinical outcome. Currently, therapies directed at attenuation of an ongoing inflammatory cascade are lacking. Administration of high-lipid enteral nutrition before hemorrhagic shock has been shown to effectively inhibit early and late proinflammatory cytokines by activation of the autonomic nervous system via cholecystokinin (CCK)-receptors. METHODS A rat model of hemorrhagic shock was used in which animals were either fasted or treated with high-lipid or control low-lipid enteral nutrition. CCK-receptor antagonists were administered before feeding. Tissues and plasma were collected to assess inflammation and intestinal integrity. RESULTS Administration of high-lipid enteral nutrition after shock reduced plasma interferon-gamma (IFN-gamma) significantly in comparison with those in low-lipid-treated and fasted animals (P < 0.01 and P < 0.001, respectively). Also, interleukin (IL)-10 levels in plasma were decreased in comparison with those in fasted animals (P < 0.001). Enterocyte damage, expressed as circulating ileal lipid-binding protein (ILBP), was prevented by early high-lipid nutrition in comparison with that in low-lipid-treated and fasted animals (P = 0.05 and P < 0.001, respectively). Furthermore, high-lipid feeding preserved intestinal integrity in comparison with that observed in low-lipid-treated and fasted animals, as assessed by bacterial translocation (BT) to distant organs (P < 0.05 and P < 0.001, respectively) and ileal permeability to horseradish peroxidase (HRP) (P = 0.05 and P < 0.001, respectively). The protective effects of high-lipid intervention were nullified by CCK-receptor antagonists (IFN-gamma; IL-10; BT; and HRP; P < 0.05). CONCLUSION High-lipid enteral nutrition given postshock reduces inflammation and preserves tissue integrity via a CCK-receptor-dependent mechanism. These findings implicate that intervention with high-lipid enteral nutrition following events such as severe trauma is a potential therapy to attenuate the developing inflammatory response.
Collapse
Affiliation(s)
- Jacco-Juri de Haan
- Department of Surgery, Nutrition and Toxicology Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
132
|
Abstract
Enhancing survival to hemorrhage of both civilian and military patients is a major emphasis for trauma research. Previous observations in humans and outbred rats show differential survival to similar levels of hemorrhage. In an initial attempt to determine potential genetic components of such differential outcomes, survival time after a controlled hemorrhage was measured in 15 inbred strains of rats. Anesthetized rats were catheterized, and approximately 24 h later, 55% of the calculated blood volume was removed during a 26-min period from conscious unrestrained animals. Rats were observed for a maximum of 6 h. Survival time was 7.7-fold longer in the longest-lived strain (Brown Norway/Medical College of Wisconsin; 306 +/- 36 min; mean +/- SEM) than in the shortest-lived strain (DA; 40 +/- 5 min; P < or = 0.01). Mean survival times for the remaining inbred strains ranged from 273 +/- 44 to 49 +/- 4 min (Dahl-Salt Sensitive > Brown Norway > Munich Wistar Fromter> Dahl-Salt Resistant > Copenhagen > Noble > Spontaneous-hypertensive > Lewis > BDIX > Fawn Hooded Hypertensive > FISCHER 344 > Black agouti > PVG). The variance in the hazard of death attributable to different strains was estimated to be 1.22 log-hazard units, corresponding to a heritability of approximately 48%. Graded and divergent survival times to hemorrhage in inbred rat strains are remarkable and suggest multiple genetic components for this characteristic. However, this interpretation of differential responses to hemorrhage may be confounded by potential strain-associated differences related to the surgical preparation. Identification of inbred strains divergent in survival time to hemorrhage provides the opportunity for future use of these strains to identify genes associated with this complex response.
Collapse
|
133
|
Raju R, Chaudry IH. Sex steroids/receptor antagonist: their use as adjuncts after trauma-hemorrhage for improving immune/cardiovascular responses and for decreasing mortality from subsequent sepsis. Anesth Analg 2008; 107:159-66. [PMID: 18635483 DOI: 10.1213/ane.0b013e318163213d] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Studies in human as well as animal models demonstrate that females in the proestrus cycle (i.e., with high estrogen) tolerate trauma-hemorrhage and sepsis far better than males. The female sex steroid, estrogen, is the significant factor contributing to this observed gender difference in outcome. One reason for the lack of significant gender association in some clinical studies is the possibility of heterogeneity of the population in terms of their hormonal status at the time of injury. Several experimental investigations have revealed that androgens produce immune and cardiovascular depression after trauma-hemorrhage. However, the use of an androgen receptor antagonist after trauma-hemorrhage has salutary effects of immune and cardiovascular function. Likewise, estrogen produces beneficial effects on immune and cardiovascular function after trauma-hemorrhage and significantly decreases mortality rates from subsequent sepsis. The salutary effects of estrogen after trauma-hemorrhage have been shown to be due to both genomic and nongenomic effects. Thus, the use of an estrogen or androgen receptor antagonist as an adjunct after trauma-hemorrhage is a safe and novel approach for restoring immune and cardiovascular function after trauma-hemorrhage and for decreasing the mortality from subsequent sepsis.
Collapse
Affiliation(s)
- Raghavan Raju
- Center for Surgical Research and Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | |
Collapse
|
134
|
Angele MK, Schneider CP, Chaudry IH. Bench-to-bedside review: latest results in hemorrhagic shock. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 12:218. [PMID: 18638356 PMCID: PMC2575549 DOI: 10.1186/cc6919] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hemorrhagic shock is a leading cause of death in trauma patients worldwide. Bleeding control, maintenance of tissue oxygenation with fluid resuscitation, coagulation support, and maintenance of normothermia remain mainstays of therapy for patients with hemorrhagic shock. Although now widely practised as standard in the USA and Europe, shock resuscitation strategies involving blood replacement and fluid volume loading to regain tissue perfusion and oxygenation vary between trauma centers; the primary cause of this is the scarcity of published evidence and lack of randomized controlled clinical trials. Despite enormous efforts to improve outcomes after severe hemorrhage, novel strategies based on experimental data have not resulted in profound changes in treatment philosophy. Recent clinical and experimental studies indicated the important influences of sex and genetics on pathophysiological mechanisms after hemorrhage. Those findings might provide one explanation why several promising experimental approaches have failed in the clinical arena. In this respect, more clinically relevant animal models should be used to investigate pathophysiology and novel treatment approaches. This review points out new therapeutic strategies, namely immunomodulation, cardiovascular maintenance, small volume resuscitation, and so on, that have been introduced in clinics or are in the process of being transferred from bench to bedside. Control of hemorrhage in the earliest phases of care, recognition and monitoring of individual risk factors, and therapeutic modulation of the inflammatory immune response will probably constitute the next generation of therapy in hemorrhagic shock. Further randomized controlled multicenter clinical trials are needed that utilize standardized criteria for enrolling patients, but existing ethical requirements must be maintained.
Collapse
Affiliation(s)
- Martin K Angele
- Department of Surgery, Klinikum Grosshadern, Ludwig-Maximilians-University, Marchionistrasse 15, 81377 Munich, Germany
| | | | | |
Collapse
|
135
|
Wu R, Dong W, Qiang X, Ji Y, Cui T, Yang J, Zhou M, Blau S, Marini CP, Ravikumar TS, Wang P. Human vasoactive hormone adrenomedullin and its binding protein rescue experimental animals from shock. Peptides 2008; 29:1223-30. [PMID: 18403050 PMCID: PMC2488201 DOI: 10.1016/j.peptides.2008.02.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 02/26/2008] [Accepted: 02/28/2008] [Indexed: 10/22/2022]
Abstract
We recently discovered that vascular responsiveness to adrenomedullin (AM), a vasoactive hormone, decreases after hemorrhage, which is markedly improved by the addition of its binding protein AMBP-1. One obstacle hampering the development of AM/AMBP-1 as resuscitation agents in trauma victims is the potential immunogenicity of rat proteins in humans. Although less potent than rat AM, human AM has been shown to increase organ perfusion in rats. We therefore hypothesized that administration of human AM/AMBP-1 improves organ function and survival after severe blood loss in rats. To test this, male Sprague-Dawley rats were bled to and maintained at an MAP of 40 mmHg for 90 min. They were then resuscitated with an equal volume of shed blood in the form of Ringer's lactate (i.e., low-volume resuscitation) over 60 min. At 15 min after the beginning of resuscitation, human AM/AMBP-1 (12/40 or 48/160 microg/kg BW) were administered intravenously over 45 min. Various pathophysiological parameters were measured 4h after resuscitation. In additional groups of animals, a 12-day survival study was conducted. Our result showed that tissue injury as evidenced by increased levels of transaminases, lactate, and creatinine, was present at 4h after hemorrhage and resuscitation. Moreover, pro-inflammatory cytokines TNF-alpha and IL-6 were also significantly elevated. Administration of AM/AMBP-1 markedly attenuated tissue injury, reduced cytokine levels, and improved the survival rate from 29% (vehicle) to 62% (low-dose) or 70% (high-dose). However, neither human AM alone nor human AMBP-1 alone prevented the significant increase in ALT, AST, lactate and creatinine at 4h after the completion of hemorrhage and resuscitation. Moreover, the half-life of human AM and human AMBP-1 in rats was 35.8 min and 1.68 h, respectively. Thus, administration of human AM/AMBP-1 may be a useful approach for attenuating organ injury, and reducing mortality after hemorrhagic shock.
Collapse
Affiliation(s)
- Rongqian Wu
- Department of Surgery, North Shore University Hospital and Long Island Jewish Medical Center, Manhasset, NY 11030, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Bracht H, Krejci V, Hiltebrand L, Brandt S, Sigurdsson G, Ali SZ, Takala J, Jakob SM. Orthogonal polarization spectroscopy to detect mesenteric hypoperfusion. Intensive Care Med 2008; 34:1883-90. [DOI: 10.1007/s00134-008-1130-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Accepted: 01/26/2008] [Indexed: 11/28/2022]
|
137
|
Santone DJ, Shahani R, Rubin BB, Romaschin AD, Lindsay TF. Mast cell stabilization improves cardiac contractile function following hemorrhagic shock and resuscitation. Am J Physiol Heart Circ Physiol 2008; 294:H2456-64. [PMID: 18390822 DOI: 10.1152/ajpheart.00925.2007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hemorrhagic shock (HS) is associated with cardiac contractile dysfunction. Mast cell (MC) degranulation is hypothesized to mediate the cardiodepressant effect. Cardiac function was assessed after HS and resuscitation (HS/R) with the administration of the MC stabilizers to prevent MC degranulation. Anesthetized male Sprague-Dawley rats were randomized to sham-operated control or HS/R groups and underwent 60 min of HS followed by 2 h of resuscitated reperfusion. Animals in the HS/R groups were randomized to receive cromolyn (5 mg/kg), ketotifen (1 mg/kg), or saline 15 min before shock. Hearts were excised following HS or 2 h of reperfusion, and function was assessed on a Langendorff apparatus. A second group of randomized animals had serial blood samples taken to assess MC degranulation by quantifying levels of serum beta-hexosaminidase. Hearts were excised at 0 min (before HS) and following 60 min of HS (before resuscitation) for a histological evaluation of MC density and degranulation. In vivo MC stabilization using ketotifen and cromolyn improved cardiac peak systolic pressure (P < 0.05), contractility (P < 0.05), and relaxation (P < 0.05) compared with that of HS controls. Serum beta-hexosaminidase increased during HS/R and was inhibited by MC stabilization (P < 0.05). Degranulation was inhibited when assessed by histochemistry and immune fluorescence. The inhibition of MC degranulation can significantly improve cardiac function following HS/R.
Collapse
Affiliation(s)
- David J Santone
- Division of Vascular Surgery, University Health Network, and the Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
138
|
Wu D, Arias J, Bassuk J, Doods H, Seidler R, Adams JA, Abraham WM. Na+/H+ EXCHANGE INHIBITION DELAYS THE ONSET OF HYPOVOLEMIC CIRCULATORY SHOCK IN PIGS. Shock 2008; 29:519-25. [PMID: 17724433 DOI: 10.1097/shk.0b013e318150757a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Severe blood loss is a major cause of death occurring within hours of traumatic injury. Na+/H+ exchange (NHE-1) activity is an important determinant of the extent of ischemic myocardial injury. The goal of the present study was to test the hypothesis that NHE-1 inhibition delays the onset of hypovolemic circulatory shock, thereby preventing early death due to severe hemorrhage in pigs. Severe hypovolemia was studied in 16 (25.2 kg) anesthetized male pigs in steps of 10-, 20-, 30-, 40-, and 50-mL kg(-1) blood loss, each in 30-min intervals. Shed blood resuscitation was started 30 min after 50 mL kg(-1) blood loss. The experiment was terminated after 3 h of resuscitation. Eight pigs were used as seline control. Eight pigs received 3 mg kg(-1) benzamide, N-(aminoiminomethyl)-4-[4-(2-furanylcarbonyl)-1-piperazinyl]-3-(methylsulfonyl), methanesulfonate (NHE-1 inhibitor) 15 min before hemorrhage. Seven control pigs died at 40- to 50-mL kg(-1) blood loss. One control pig survived initial resuscitation but died soon after. In contrast, all animals treated with NHE-1 inhibitor survived the entire protocol. In control animals, cardiac output and MAP gradually decreased at each step of blood loss with marked increase in heart rate. Cardiovascular decompensation occurred at 40 mL kg(-1) blood loss. Na+/H+ exchange inhibition increased oxygen delivery, attenuated cardiovascular decompensation, delayed the onset of irreversible hypovolemic circulatory shock, and enabled resuscitation to survival. Echocardiography analysis showed that myocardial hypercontracture gradually developed with each step of blood loss in control animals, but this hypercontracture was attenuated in the animals receiving the NHE-1 inhibitor. We conclude that NHE-1 inhibition attenuates ischemic myocardial hypercontracture, cardiovascular decompensation, delays the onset of hypovolemic circulatory shock, and prevents early death in severe hemorrhage.
Collapse
Affiliation(s)
- Dongmei Wu
- Department of Research, Division of Neonatology, Mount Sinai Medical Canter, Miami Beach, Florida 33140, USA.
| | | | | | | | | | | | | |
Collapse
|
139
|
Alten JA, Moran A, Tsimelzon AI, Mastrangelo MAA, Hilsenbeck SG, Poli V, Tweardy DJ. Prevention of hypovolemic circulatory collapse by IL-6 activated Stat3. PLoS One 2008; 3:e1605. [PMID: 18270592 PMCID: PMC2225503 DOI: 10.1371/journal.pone.0001605] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 01/03/2008] [Indexed: 01/26/2023] Open
Abstract
Half of trauma deaths are attributable to hypovolemic circulatory collapse (HCC). We established a model of HCC in rats involving minor trauma plus severe hemorrhagic shock (HS). HCC in this model was accompanied by a 50% reduction in peak acceleration of aortic blood flow and cardiomyocyte apoptosis. HCC and apoptosis increased with increasing duration of hypotension. Apoptosis required resuscitation, which provided an opportunity to intervene therapeutically. Administration of IL-6 completely reversed HCC, prevented cardiac dysfunction and cardiomyocyte apoptosis, reduced mortality 5-fold and activated intracardiac signal transducer and activator of transcription (STAT) 3. Pre-treatment of rats with a selective inhibitor of Stat3, T40214, reduced the IL-6-mediated increase in cardiac Stat3 activity, blocked successful resuscitation by IL-6 and reversed IL-6-mediated protection from cardiac apoptosis. The hearts of mice deficient in the naturally occurring dominant negative isoform of Stat3, Stat3β, were completely resistant to HS-induced apoptosis. Microarray analysis of hearts focusing on apoptosis related genes revealed that expression of 29% of apoptosis related genes was altered in HS vs. sham rats. IL-6 treatment normalized the expression of these genes, while T40214 pretreatment prevented IL-6-mediated normalization. Thus, cardiac dysfunction, cardiomyocyte apoptosis and induction of apoptosis pathway genes are important components of HCC; IL-6 administration prevented HCC by blocking cardiomyocyte apoptosis and induction of apoptosis pathway genes via Stat3 and warrants further study as a resuscitation adjuvant for prevention of HCC and death in trauma patients.
Collapse
Affiliation(s)
- Jeffrey A. Alten
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ana Moran
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anna I. Tsimelzon
- Breast Care Center and Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Mary-Ann A. Mastrangelo
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Susan G. Hilsenbeck
- Breast Care Center and Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Valeria Poli
- Department of Genetics, Biology and Biochemistry, University of Turin, Turin, Italy
| | - David J. Tweardy
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- *E-mail:
| |
Collapse
|
140
|
Oxygen saturation determined from deep muscle, not thenar tissue, is an early indicator of central hypovolemia in humans. Crit Care Med 2008; 36:176-82. [PMID: 18090350 DOI: 10.1097/01.ccm.0000295586.83787.7e] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To compare the responses of noninvasively measured tissue oxygen saturation (StO2) and calculated muscle oxygen tension (PmO2) to standard hemodynamic variables for early detection of imminent hemodynamic instability during progressive central hypovolemia in humans. DESIGN Prospective study. SETTING Research laboratory. SUBJECTS Sixteen healthy human volunteers. INTERVENTIONS Progressive lower body negative pressure (LBNP) to onset of cardiovascular collapse. MEASUREMENTS AND MAIN RESULTS Noninvasive measurements of blood pressures, heart rate, and stroke volume were obtained during progressive LBNP with simultaneous assessments of StO2, PmO2, and muscle oxygen saturation (SmO2). Forearm SmO2 and PmO2 were determined with a novel near infrared spectroscopic measurement device (UMMS) and compared with thenar StO2 measured by a commercial device (HT). All values were normalized to the duration of LBNP exposure required for cardiovascular collapse in each subject (i.e., LBNP maximum). Stroke volume was significantly decreased at 25% of LBNP maximum, whereas blood pressure was a late indicator of imminent cardiovascular collapse. PmO2 (UMMS) was significantly decreased at 50% of maximum LBNP while SmO2 (UMMS) decreased at 75% of maximum LBNP. Thenar StO2 (HT) showed no statistical change throughout the entire LBNP protocol. CONCLUSIONS Spectroscopic assessment of forearm muscle PO2 and SmO2 provides noninvasive and continuous measures that are early indicators of impending cardiovascular collapse resulting from progressive reductions in central blood volume.
Collapse
|
141
|
Femoral nailing-related coagulopathy determined by first-hit magnitude: an animal study. Clin Orthop Relat Res 2008; 466:473-80. [PMID: 18196434 PMCID: PMC2505120 DOI: 10.1007/s11999-007-0066-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Accepted: 11/02/2007] [Indexed: 01/31/2023]
Abstract
We asked whether coagulopathy worsened during femoral intramedullary nailing in the presence of lung contusion and hemorrhagic shock and whether reamed or unreamed nailing influenced these results. In 30 Merino sheep, we induced hemorrhagic shock and/or standardized lung contusion followed by femoral nailing. Six groups of five each were assigned as follows: thoracotomy control groups treated with reamed or unreamed nailing, lung contusion groups treated with reamed or unreamed nailing, and shock and lung contusion groups treated with reamed or unreamed nailing. After lung contusion alone (first hit), the serum values of antithrombin III, factor V, and fibrinogen were considerably altered after reamed and unreamed femoral nailing (second hit) 4 hours postoperatively. In the lung contusion and shock groups, we found a substantial reduction for all serum coagulative parameters between baseline and fixation after reamed and unreamed nailing. The magnitude of the first hit is increased if hemorrhagic shock is added to a lung contusion determined by hemostatic reactions. The magnitude of the injury appears equally important as the type of subsequent surgery and should be considered in planning for fracture fixation in patients at high risk for complications.
Collapse
|
142
|
Abstract
BACKGROUND We previously demonstrated that the degradation of a suspension of Jurkat cells in Ringer's lactate (RL) was inhibited by the addition of a 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid/Tris buffer. Given the ability of histidine to buffer protons in the physiologic range (pKa = 6.0), we hypothesized that this amino acid would have the same effect. METHODS RL was made in our laboratory using sodium l-lactate. Jurkat cells were suspended in RL alone or RL with various concentrations of histidine or other test reagents at 37 degrees C for 4 hours or 24 hours in an atmosphere of 95% air and 5% CO2. Using flow cytometry, we measured cell shrinkage, phosphatidylserine translocation, propidium iodide uptake, and intracellular oxygen free radical production. RESULTS Cell shrinkage was induced by suspension in RL after 4 hours incubation. At 4 hours, cell shrinkage was inhibited by all concentrations of histidine tested, 7.8 mumol/L to 10 mmol/L. There was no statistical difference between cells suspended in medium and cells suspended in 1 mmol/L or 10 mmol/L histidine. After 24 hours incubation, 100% of the cells in RL had undergone cell shrinkage whereas in 10 mmol/L histidine only a mean of 20% of the cells had undergone cell shrinkage. The inhibitory effect of 1 mmol/L histidine at pH 7.4 was compared with that at pH 6.8. After 4 hours incubation, there was no difference. After 24 hours incubation, the inhibitory effect at pH 7.4 was significantly greater that that at pH 6.8. Histidine at 1 mmol/L to 10 mmol/L significantly reduced the percentage of cells that underwent phosphatidylserine translocation and propidium iodide uptake. The effect of the dipeptide buffer, glycylglycine, and the two other positively charged amino acids, arginine and lysine, after 4 hours incubation was compared with histidine at 1 mmol/L. At 1 mmol/L, histidine was superior to arginine and lysine and indistinguishable from glycylglycine. Intracellular free radical production was measured at 0.5 mmol/L, 1.0 mmol/L, and 10 mmol/L histidine concentrations. There was significant inhibition only at 10 mmol/L. CONCLUSIONS Characteristics of apoptotic cell death that occur in cells suspended in RL are inhibited by the addition of histidine, arginine, and lysine as well as the dipeptide glycylglycine, which, with a pKa of 8.25, also buffers in the physiologic range. Histidine is superior to lysine and arginine at 1 mmol/L. The salutary effect of histidine at 0.5 mmol/L and 1 mmol/L is caused by a mechanism other than the inhibition of oxygen free radicals. Moreover, the buffering of protons may play a role at 24 hours but made no difference at 4 hours.
Collapse
|
143
|
Thresholded area over the curve of spectrometric tissue oxygen saturation as an indicator of volume resuscitability in porcine hemorrhagic shock. ACTA ACUST UNITED AC 2008; 63:573-8; discussion 578-80. [PMID: 18073603 DOI: 10.1097/ta.0b013e31812f6095] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND A rapid, reliable, and noninvasive functional measure of responsiveness to resuscitation in posttraumatic hemorrhagic shock could prove useful in guiding therapy, especially under circumstances such as the battlefield and civilian mass casualties. Tissue oxygen saturation (Sto2) is a promising candidate for this application. We therefore explored the value of peripheral muscle Sto2 in predicting systemic responsiveness to colloid volume resuscitation in a porcine model of hemorrhagic shock. METHODS Fourteen isoflurane-anesthetized piglets were subjected to a standardized hemorrhage protocol that maintained mean arterial pressure (MAP) between 30 and 40 mm Hg. Asanguineous resuscitation with a volume of Hextend equal to the total volume bled was initiated when compensation was exhausted (MAP <30 mm Hg). We recorded continuous MAP and Sto2 values, and calculated the contiguous area over the Sto2 curve yet below a given threshold of Sto2 (TAOC) as a function of this threshold before the selected timepoint for timepoints up to 30 minutes before resuscitation. RESULTS Hemorrhage resulted in significant fluctuations of MAP and high interindividual variability of disease dynamics and outcome: 4 nonsurvivors and 10 survivors at 2 hours postresuscitation. Sto2 measurements reflected hemodynamic conditions in most animals, with a pronounced drop preceding final decompensation in 7 of 14 animals. TAOC discriminated three of four nonresuscitable (nonsurvivor) animals from the survivors, with group differences reaching significance even for the earliest examined timepoint (30 minutes before resuscitation), depending on the choice of TAOC threshold. CONCLUSIONS Sto2 may serve as a marker of decompensation, whereas TAOC, a physiologically motivated correlate of perfusion debt and cumulative hypoperfusion injury, may be a useful early indicator of responsiveness to volume resuscitation in hemorrhagic shock.
Collapse
|
144
|
Dries DJ. Traumatic Shock and Tissue Hypoperfusion: Nonsurgical Management. Crit Care Med 2008. [DOI: 10.1016/b978-032304841-5.50030-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
145
|
Soller BR, Yang Y, Soyemi OO, Ryan KL, Rickards CA, Walz JM, Heard SO, Convertino VA. Noninvasively determined muscle oxygen saturation is an early indicator of central hypovolemia in humans. J Appl Physiol (1985) 2007; 104:475-81. [PMID: 18006869 DOI: 10.1152/japplphysiol.00600.2007] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Ten healthy human volunteers were subjected to progressive lower body negative pressure (LBNP) to the onset of cardiovascular collapse to compare the response of noninvasively determined skin and fat corrected deep muscle oxygen saturation (SmO2) and pH to standard hemodynamic parameters for early detection of imminent hemodynamic instability. Muscle SmO2 and pH were determined with a novel near infrared spectroscopic (NIRS) technique. Heart rate (HR) was measured continuously via ECG, and arterial blood pressure (BP) and stroke volume (SV) were obtained noninvasively via Finometer and impedance cardiography on a beat-to-beat basis. SmO2 and SV were significantly decreased during the first LBNP level (-15 mmHg), whereas HR and BP were late indicators of impending cardiovascular collapse. SmO2 declined in parallel with SV and inversely with total peripheral resistance, suggesting, in this model, that SmO2 is an early indicator of a reduction in oxygen delivery through vasoconstriction. Muscle pH decreased later, suggesting an imbalance between delivery and demand. Spectroscopic determination of SmO2 is noninvasive and continuous, providing an early indication of impending cardiovascular collapse resulting from progressive reduction in central blood volume.
Collapse
Affiliation(s)
- Babs R Soller
- Dept. of Anesthesiology, UMass Medical School, 55 Lake Ave North, Worcester, MA 01655, USA.
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Mollen KP, McCloskey CA, Tanaka H, Prince JM, Levy RM, Zuckerbraun BS, Billiar TR. Hypoxia activates c-Jun N-terminal kinase via Rac1-dependent reactive oxygen species production in hepatocytes. Shock 2007; 28:270-7. [PMID: 17545941 DOI: 10.1097/shk.0b013e3180485acd] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The earliest events after the induction of hemorrhagic shock (HS) are complex and poorly understood. We have recently demonstrated that decreased tissue perfusion and hypoxia during HS lead to an increased phosphorylation of c-Jun N-terminal kinase (JNK) in vivo. The purpose of these investigations was to test the hypothesis that hypoxia activates JNK via Rac1-dependent reactive oxygen species (ROS) signaling. Mice subjected to HS and resuscitated with Ringer's ethyl pyruvate solution (REPS) or N-acetylcysteine (NAC), two scavengers of ROS, demonstrated decreased levels of phosphorylated JNK. Exposure of primary mouse hepatocytes in culture to 1% oxygen led to increased production of ROS and phosphorylation of JNK. The duration of hypoxia correlated with the level of generation of ROS and JNK activation. The phosphorylation of JNK was attenuated in the presence of ROS scavengers or the nicotinamide adenosine dinucleotide phosphate [NDA(P)H] oxidase inhibitor, diphenyleneiodonium (DPI). In addition, hypoxia increased activation of Rac1. Inhibition of Rac1 activation by adenoviral gene transfer of dominant-negative Rac1 (AdRac1) attenuated both ROS formation and JNK activation. Together, these data suggest that ROS generation during hypoxia in the liver directly leads to JNK activation in a Rac1-dependent process.
Collapse
Affiliation(s)
- Kevin P Mollen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
147
|
Chatterjee PK. Novel pharmacological approaches to the treatment of renal ischemia-reperfusion injury: a comprehensive review. Naunyn Schmiedebergs Arch Pharmacol 2007; 376:1-43. [PMID: 18038125 DOI: 10.1007/s00210-007-0183-5] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 08/01/2007] [Indexed: 02/07/2023]
Abstract
Renal ischemia-reperfusion (I-R) contributes to the development of ischemic acute renal failure (ARF). Multi-factorial processes are involved in the development and progression of renal I-R injury with the generation of reactive oxygen species, nitric oxide and peroxynitrite, and the decline of antioxidant protection playing major roles, leading to dysfunction, injury, and death of the cells of the kidney. Renal inflammation, involving cytokine/adhesion molecule cascades with recruitment, activation, and diapedesis of circulating leukocytes is also implicated. Clinically, renal I-R occurs in a variety of medical and surgical settings and is responsible for the development of acute tubular necrosis (a characteristic feature of ischemic ARF), e.g., in renal transplantation where I-R of the kidney directly influences graft and patient survival. The cellular mechanisms involved in the development of renal I-R injury have been targeted by several pharmacological interventions. However, although showing promise in experimental models of renal I-R injury and ischemic ARF, they have not proved successful in the clinical setting (e.g., atrial natriuretic peptide, low-dose dopamine). This review highlights recent pharmacological developments, which have shown particular promise against experimental renal I-R injury and ischemic ARF, including novel antioxidants and antioxidant enzyme mimetics, nitric oxide and nitric oxide synthase inhibitors, erythropoietin, peroxisome-proliferator-activated receptor agonists, inhibitors of poly(ADP-ribose) polymerase, carbon monoxide-releasing molecules, statins, and adenosine. Novel approaches such as recent research involving combination therapies and the potential of non-pharmacological strategies are also considered.
Collapse
Affiliation(s)
- Prabal K Chatterjee
- Division of Pharmacology and Therapeutics, School of Pharmacy and Biomolecular Sciences, University of Brighton, Cockcroft Building, Lewes Road, Moulsecoomb, Brighton BN2 4GJ, UK.
| |
Collapse
|
148
|
Cabrales P, Tsai AG, Intaglietta M. Hemorrhagic shock resuscitation with carbon monoxide saturated blood. Resuscitation 2007; 72:306-18. [PMID: 17092627 DOI: 10.1016/j.resuscitation.2006.06.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2006] [Revised: 06/07/2006] [Accepted: 06/07/2006] [Indexed: 11/28/2022]
Abstract
The response to exchange transfusion with red blood cells (RBCs) saturated with carbon monoxide (CO) in amelioration of microvascular function and providing tissue protection in hemorrhagic shock resuscitation was investigated in the hamster chamber window model. Shock was induced by the withdrawal of 50% of blood volume (BV). Blood volume was restored 1 h after hemorrhage with a single volume infusion (resuscitation) of 25% BV with fresh RBCs (saturated or unsaturated with CO) suspended in human serum albumin (HSA). Hemorrhage, shock and resuscitation were monitored continuously in terms of mean arterial pressure (MAP), microvascular blood flow, capillary perfusion and systemic gas parameters. Eight hours after resuscitation, Annexin V and propidium iodide (PI) were injected into the window chamber to study tissue viability, and labeled cells were observed by using intravital epifluorescence microscopy. TUNEL staining was performed on the tissue to confirm in vivo results. Systemic and microvascular restoration were not different with or without CO up to 90 min after resuscitation. CO concentration decreased over 90 min, increasing oxygen carrying capacity and gradually reoxygenating the tissue. CO saturated blood partially mitigated cell injury at 8 h after resuscitation. The precise cellular mechanisms involved require further elucidation. CO is a novel experimental strategy to improve tissue viability and requires the appropriated preclinical studies to confirm its efficacy.
Collapse
Affiliation(s)
- Pedro Cabrales
- La Jolla Bioengineering Institute, 505 Coast Boulevard South, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
149
|
Kuboki S, Okaya T, Schuster R, Blanchard J, Denenberg A, Wong HR, Lentsch AB. Hepatocyte NF-kappaB activation is hepatoprotective during ischemia-reperfusion injury and is augmented by ischemic hypothermia. Am J Physiol Gastrointest Liver Physiol 2007; 292:G201-7. [PMID: 16950761 DOI: 10.1152/ajpgi.00186.2006] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The present study examined the role of hepatocyte NF-kappaB activation during ischemia-reperfusion injury. Second, we evaluated the effects of ischemic hypothermia on NF-kappaB activation and liver injury. C57BL/6 mice underwent 90 min of partial hepatic ischemia and up to 8 h of reperfusion. Body temperature was regulated during the ischemic period between 35 and 37 degrees C, 33 and 35 degrees C, 29 and 33 degrees C or unregulated, where temperature fell to <29 degrees C. Liver injury, as measured by serum alanine aminotransferase as well as liver histopathology, was inversely proportional to regulated body temperature, with the unregulated group (<29 degrees C) being highly protected and the normothermic group (35-37 degrees C) displaying the greatest injury. Inflammation, as measured by production of TNF-alpha and liver recruitment of neutrophils, was greatest in the normothermic groups and lowest in the ischemic hypothermia groups. Interestingly, hepatocyte NF-kappaB activation was highest in the hypothermic group and least in the normothermic group. Paradoxically, degradation of IkappaB proteins, IkappaB-alpha and IkappaB-beta, was greatest in the normothermic group, suggesting an alternate NF-kappaB regulatory mechanism during ischemia-reperfusion injury. Subsequently, we found that NF-kappaB p65 protein was increasingly degraded in normothermic versus hypothermic groups, and this degradation was specific for hepatocytes and was associated with decreased expression of the peptidyl-prolyl isomerase Pin1. The data suggest that NF-kappaB activation in hepatocytes is a protective response during ischemia-reperfusion and can be augmented by ischemic hypothermia. Furthermore, it appears that Pin1 promotes NF-kappaB p65 protein stability such that decreased expression of Pin1 during ischemia-reperfusion results in p65 degradation, reduced nuclear translocation of NF-kappaB, and enhanced hepatocellular injury.
Collapse
Affiliation(s)
- Satoshi Kuboki
- The Laboratory of Trauma, Sepsis and Inflammation Research, Department of Surgery, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA
| | | | | | | | | | | | | |
Collapse
|
150
|
Abstract
Shock resulting from life-threatening blood loss (hemorrhage) remains a common complication of traumatic injury. Intensive experimental efforts are needed if we are to understand the pathological effect(s) of hemorrhagic shock, alone or in association with traumatic tissue injury, and to reverse this deleterious process in trauma patients. Here, we overview selected studies that are representative of the different hemorrhagic shock models, considering their advantages and disadvantages from a scientific and clinical perspective. Fixed-pressure versus fixed-volume versus uncontrolled hemorrhage models, with or without tissue injury, will be discussed, as well as small versus large animal models. Most of these models are nonlethal in nature, and allow the researcher to understand the changes that contribute to increased susceptibility to subsequent infection or the development of multiple organ failure. We also consider some of the confounders in these models, including anesthesia, the nature of resuscitation, and the use of anticoagulants. The selection of model must take into consideration not only the need for experimental control but must also adequately reflect the clinical pathobiology of shock if we are to develop better pharmacological interventions.
Collapse
Affiliation(s)
- J L Lomas-Niera
- Division of Surgical Research/Department of Surgery, Rhode Island Hospital and Brown University School of Medicine, Providence, RI 02903, USA
| | | | | | | |
Collapse
|