1
|
Ford VJ, Applefeld WN, Wang J, Sun J, Solomon SB, Sidenko S, Feng J, Sheffield C, Klein HG, Yu ZX, Torabi-Parizi P, Danner RL, Sachdev V, Solomon MA, Chen MY, Natanson C. Cardiac Magnetic Resonance Studies in a Large Animal Model That Simulates the Cardiac Abnormalities of Human Septic Shock. J Am Heart Assoc 2024; 13:e034026. [PMID: 39101510 DOI: 10.1161/jaha.123.034026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/30/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Septic shock is associated with increases in end-diastolic volume (EDV) and decreases in ejection fraction that reverse within 10 days. Nonsurvivors do not develop EDV increases. The mechanism is unknown. METHODS AND RESULTS Purpose-bred beagles (n=33) were randomized to receive intrabronchial Staphylococcus aureus or saline. Over 96 hours, cardiac magnetic resonance imaging and echocardiograms were performed. Tissue was obtained at 66 hours. From 0 to 96 hours after bacterial challenge, septic animals versus controls had significantly increased left ventricular wall edema (6%) and wall thinning with loss of mass (15%). On histology, the major finding was nonocclusive microvascular injury with edema in myocytes, the interstitium, and endothelial cells. Edema was associated with significant worsening of biventricular ejection fractions, ventricular-arterial coupling, and circumferential strain. Early during sepsis, (0-24 hours), the EDV decreased; significantly more in nonsurvivors (ie, greater diastolic dysfunction). From 24 to 48 hours, septic animals' biventricular chamber sizes increased; in survivors significantly greater than baseline and nonsurvivors, whose EDVs were not different from baseline. Preload, afterload, or heart rate differences did not explain these differential changes. CONCLUSIONS The cardiac dysfunction of sepsis is associated with wall edema. In nonsurvivors, at 0 to 24 hours, sepsis induces a more severe diastolic dysfunction, further decreasing chamber size. The loss of left ventricular mass with wall thinning in septic survivors may, in part, explain the EDV increases from 24 to 48 hours because of a potentially reparative process removing damaged wall tissue. Septic cardiomyopathy is most consistent with a nonocclusive microvascular injury resulting in edema causing reversible systolic and diastolic dysfunction with more severe diastolic dysfunction being associated with a decreased EDV and death.
Collapse
MESH Headings
- Animals
- Dogs
- Disease Models, Animal
- Shock, Septic/physiopathology
- Shock, Septic/complications
- Stroke Volume
- Magnetic Resonance Imaging
- Edema, Cardiac/physiopathology
- Edema, Cardiac/pathology
- Edema, Cardiac/diagnostic imaging
- Ventricular Function, Left
- Time Factors
- Humans
- Staphylococcal Infections/complications
- Staphylococcal Infections/physiopathology
- Echocardiography
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/etiology
- Male
Collapse
Affiliation(s)
- Verity J Ford
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
| | - Willard N Applefeld
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
- Division of Cardiology Duke University Medical Center Durham NC USA
| | - Jeffrey Wang
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
- Emory University Atlanta GA USA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
| | - Steven B Solomon
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
| | - Stanislav Sidenko
- National Heart Lung and Blood Institute National Institutes of Health Bethesda MD USA
| | - Jing Feng
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
| | | | - Harvey G Klein
- Department of Transfusion Medicine, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
| | - Zu-Xi Yu
- National Heart Lung and Blood Institute National Institutes of Health Bethesda MD USA
| | - Parizad Torabi-Parizi
- National Heart Lung and Blood Institute National Institutes of Health Bethesda MD USA
| | - Robert L Danner
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
- National Heart Lung and Blood Institute National Institutes of Health Bethesda MD USA
| | - Vandana Sachdev
- National Heart Lung and Blood Institute National Institutes of Health Bethesda MD USA
| | - Michael A Solomon
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
- National Heart Lung and Blood Institute National Institutes of Health Bethesda MD USA
| | - Marcus Y Chen
- National Heart Lung and Blood Institute National Institutes of Health Bethesda MD USA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center National Institutes of Health, (NIH, CC) Bethesda MD USA
- National Heart Lung and Blood Institute National Institutes of Health Bethesda MD USA
| |
Collapse
|
2
|
Ford VJ, Applefeld WN, Wang J, Sun J, Solomon SB, Klein HG, Feng J, Lertora J, Parizi-Torabi P, Danner RL, Solomon MA, Chen MY, Natanson C. In a Canine Model of Septic Shock, Cardiomyopathy Occurs Independent of Catecholamine Surges and Cardiac Microvascular Ischemia. J Am Heart Assoc 2024; 13:e034027. [PMID: 39101496 DOI: 10.1161/jaha.123.034027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/20/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND High levels of catecholamines are cardiotoxic and associated with stress-induced cardiomyopathies. Using a septic shock model that reproduces the reversible cardiomyopathy seen over 10 days associated with human septic shock, we investigated the effects of catecholamines on microcirculatory perfusion and cardiac dysfunction. METHODS AND RESULTS Purpose-bred beagles received intrabronchial Staphylococcus aureus (n=30) or saline (n=6). The septic animals were than randomized to epinephrine (1 μg/kg per minute, n=15) or saline (n=15) infusions from 4 to 44 hours. Serial cardiac magnetic resonance imaging, catecholamine levels, and troponins were collected over 92 hours. Serial adenosine-stress perfusion cardiac magnetic resonance imaging was performed on septic animals randomized to receive saline (n=8 out of 15) or epinephrine (n=8 out of 15). High-dose sedation was given to suppress endogenous catecholamine release. Despite catecholamine levels largely remaining within the normal range throughout, by 48 hours, septic animals receiving saline versus nonseptic animals still developed significant worsening of left ventricular ejection fraction, circumferential strain, and ventricular-aortic coupling. In septic animals that received epinephrine versus saline infusions, plasma epinephrine levels increased 800-fold, but epinephrine produced no significant further worsening of left ventricular ejection fraction, circumferential strain, or ventricular-aortic coupling. Septic animals receiving saline had a significant increase in microcirculatory reserve without troponin elevations. Septic animals receiving epinephrine had decreased edema, blunted microcirculatory perfusion, and elevated troponin levels that persisted for hours after the epinephrine infusion stopped. CONCLUSIONS Cardiac dysfunction during sepsis is not primarily due to elevated endogenous or exogenous catecholamines nor due to decreased microvascular perfusion-induced ischemia. However, epinephrine itself has potentially harmful long-lasting ischemic effects during sepsis including impaired cardiac microvascular perfusion that persists after stopping the infusion.
Collapse
Affiliation(s)
- Verity J Ford
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
| | - Willard N Applefeld
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
- Division of Cardiology Duke University Medical Center Durham NC USA
| | - Jeffrey Wang
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
- Emory University Atlanta GA USA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
| | - Steven B Solomon
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
| | - Harvey G Klein
- Department of Transfusion Medicine, Clinical Center National Institutes of Health Bethesda MD USA
| | - Jing Feng
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
| | - Juan Lertora
- Pennington Biomedical Research Center Louisiana State University Baton Rouge LA USA
| | - Parizad Parizi-Torabi
- National Heart Lung and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Robert L Danner
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
| | - Michael A Solomon
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
- National Heart Lung and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Marcus Y Chen
- National Heart Lung and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center National Institutes of Health Bethesda MD USA
- National Heart Lung and Blood Institute, National Institutes of Health Bethesda MD USA
| |
Collapse
|
3
|
Ford VJ, Applefeld WN, Wang J, Sun J, Solomon SB, Sidenko S, Feng J, Sheffield C, Klein HG, Yu ZX, Torabi-Parizi P, Danner RL, Sachdev V, Solomon MA, Chen MY, Natanson C. Cardiac Magnetic Resonance Studies in a Large Animal Model that Simulates the Cardiac Abnormalities of Human Septic Shock. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578971. [PMID: 38903100 PMCID: PMC11188083 DOI: 10.1101/2024.02.05.578971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Background Septic shock, in humans and in our well-established animal model, is associated with increases in biventricular end diastolic volume (EDV) and decreases in ejection fraction (EF). These abnormalities occur over 2 days and reverse within 10 days. Septic non-survivors do not develop an increase in EDV. The mechanism for this cardiac dysfunction and EDV differences is unknown. Methods Purpose-bred beagles randomized to receive intrabronchial Staphylococcus aureus (n=27) or saline (n=6) were provided standard ICU care including sedation, mechanical ventilation, and fluid resuscitation to a pulmonary arterial occlusion pressure of over 10mmHg. No catecholamines were administered. Over 96h, cardiac magnetic resonance imaging, echocardiograms, and invasive hemodynamics were serially performed, and laboratory data was collected. Tissue was obtained at 66h from six septic animals. Results From 0-96h after bacterial challenge, septic animals vs. controls had significantly increased left ventricular wall edema (6%) and wall thinning with loss of mass (15%) which was more pronounced at 48h in non-survivors than survivors. On histology, edema was located predominantly in myocytes, the interstitium, and endothelial cells. Edema was associated with significantly worse biventricular function (lower EFs), ventricular-arterial coupling, and circumferential strain. In septic animals, from 0-24h, the EDV decreased from baseline and, despite cardiac filling pressures being similar, decreased significantly more in non-survivors. From 24-48h, all septic animals had increases in biventricular chamber sizes. Survivors biventricular EDVs were significantly greater than baseline and in non-survivors, where biventricular EDVs were not different from baseline. Preload, afterload, or HR differences did not explain these differential serial changes in chamber size. Conclusion Systolic and diastolic cardiac dysfunction during sepsis is associated with ventricular wall edema. Rather than differences in preload, afterload, or heart rate, structural alterations to the ventricular wall best account for the volume changes associated with outcome during sepsis. In non-survivors, from 0-24h, sepsis induces a more severe diastolic dysfunction, further decreasing chamber size. The loss of left ventricular mass with wall thinning in septic survivors may, in part explain, the EDV increases from 24-48h. However, these changes continued and even accelerated into the recovery phase consistent with a reparative process rather than ongoing injury.
Collapse
Affiliation(s)
- Verity J. Ford
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Willard N. Applefeld
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey Wang
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- Emory, 100 Woodruff Circle, Atlanta, GA 30322
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Steven B. Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Stanislav Sidenko
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Jing Feng
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | | | - Harvey G. Klein
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Zu-Xi Yu
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Parizad Torabi-Parizi
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Robert L. Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
| | - Vandana Sachdev
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Michael A. Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Marcus Y. Chen
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, (NIH, CC) Bethesda, Maryland 20892 USA
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892 USA
| |
Collapse
|
4
|
Vintrych P, Al-Obeidallah M, Horák J, Chvojka J, Valešová L, Nalos L, Jarkovská D, Matějovič M, Štengl M. Modeling sepsis, with a special focus on large animal models of porcine peritonitis and bacteremia. Front Physiol 2023; 13:1094199. [PMID: 36703923 PMCID: PMC9871395 DOI: 10.3389/fphys.2022.1094199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Infectious diseases, which often result in deadly sepsis or septic shock, represent a major global health problem. For understanding the pathophysiology of sepsis and developing new treatment strategies, reliable and clinically relevant animal models of the disease are necessary. In this review, two large animal (porcine) models of sepsis induced by either peritonitis or bacteremia are introduced and their strong and weak points are discussed in the context of clinical relevance and other animal models of sepsis, with a special focus on cardiovascular and immune systems, experimental design, and monitoring. Especially for testing new therapeutic strategies, the large animal (porcine) models represent a more clinically relevant alternative to small animal models, and the findings obtained in small animal (transgenic) models should be verified in these clinically relevant large animal models before translation to the clinical level.
Collapse
Affiliation(s)
- Pavel Vintrych
- Department of Cardiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Mahmoud Al-Obeidallah
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Jan Horák
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Jiří Chvojka
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Lenka Valešová
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Lukáš Nalos
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Dagmar Jarkovská
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Martin Matějovič
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Milan Štengl
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia,*Correspondence: Milan Štengl,
| |
Collapse
|
5
|
Liu S, Chong W. Roles of LncRNAs in Regulating Mitochondrial Dysfunction in Septic Cardiomyopathy. Front Immunol 2021; 12:802085. [PMID: 34899764 PMCID: PMC8652231 DOI: 10.3389/fimmu.2021.802085] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/10/2021] [Indexed: 01/20/2023] Open
Abstract
Sepsis is an abnormal systemic inflammatory response of the host immune system to infection and can lead to fatal multiorgan dysfunction syndrome. Epidemiological studies have shown that approximately 10-70% of sepsis cases can lead to septic cardiomyopathy. Since the pathogenesis of septic cardiomyopathy is not clear, it is difficult for medical doctors to treat the disease. Therefore, finding effective interventions to prevent and reduce myocardial damage in septic cardiomyopathy is clinically significant. Epigenetics is the study of stable genetic phenotype inheritance that does not involve changing gene sequences. Epigenetic inheritance is affected by both gene and environmental regulation. Epigenetic studies focus on the modification and influence of chromatin structure, mainly including chromatin remodelling, DNA methylation, histone modification and noncoding RNA (ncRNA)-related mechanisms. Recently, long ncRNA (lncRNA)-related mechanisms have been the focus of epigenetic studies. LncRNAs are expected to become important targets to prevent, diagnose and treat human diseases. As the energy metabolism centre of cells, mitochondria are important targets in septic cardiomyopathy. Intervention measures to prevent and treat mitochondrial damage are of great significance for improving the prognosis of septic cardiomyopathy. LncRNAs play important roles in life activities. Recently, studies have focused on the involvement of lncRNAs in regulating mitochondrial dysfunction. However, few studies have revealed the involvement of lncRNAs in regulating mitochondrial dysfunction in septic cardiomyopathy. In this article, we briefly review recent research in this area.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Emergency, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Chong
- Department of Emergency, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Kawaguchi S, Okada M. Cardiac Metabolism in Sepsis. Metabolites 2021; 11:metabo11120846. [PMID: 34940604 PMCID: PMC8707959 DOI: 10.3390/metabo11120846] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
The mechanism of sepsis-induced cardiac dysfunction is believed to be different from that of myocardial ischemia. In sepsis, chemical mediators, such as endotoxins, cytokines, and nitric oxide, cause metabolic abnormalities, mitochondrial dysfunction, and downregulation of β-adrenergic receptors. These factors inhibit the production of ATP, essential for myocardial energy metabolism, resulting in cardiac dysfunction. This review focuses on the metabolic changes in sepsis, particularly in the heart. In addition to managing inflammation, interventions focusing on metabolism may be a new therapeutic strategy for cardiac dysfunction due to sepsis.
Collapse
Affiliation(s)
- Satoshi Kawaguchi
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Bloomington, IN 46202, USA;
| | - Motoi Okada
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa 078-8510, Japan
- Correspondence: ; Tel.: +81-166-68-2852
| |
Collapse
|
7
|
Wasyluk W, Nowicka-Stążka P, Zwolak A. Heart Metabolism in Sepsis-Induced Cardiomyopathy-Unusual Metabolic Dysfunction of the Heart. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18147598. [PMID: 34300048 PMCID: PMC8303349 DOI: 10.3390/ijerph18147598] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/26/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022]
Abstract
Due to the need for continuous work, the heart uses up to 8% of the total energy expenditure. Due to the relatively low adenosine triphosphate (ATP) storage capacity, the heart's work is dependent on its production. This is possible due to the metabolic flexibility of the heart, which allows it to use numerous substrates as a source of energy. Under normal conditions, a healthy heart obtains approximately 95% of its ATP by oxidative phosphorylation in the mitochondria. The primary source of energy is fatty acid oxidation, the rest of the energy comes from the oxidation of pyruvate. A failed heart is characterised by a disturbance in these proportions, with the contribution of individual components as a source of energy depending on the aetiology and stage of heart failure. A unique form of cardiac dysfunction is sepsis-induced cardiomyopathy, characterised by a significant reduction in energy production and impairment of cardiac oxidation of both fatty acids and glucose. Metabolic disorders appear to contribute to the pathogenesis of cardiac dysfunction and therefore are a promising target for future therapies. However, as many aspects of the metabolism of the failing heart remain unexplained, this issue requires further research.
Collapse
Affiliation(s)
- Weronika Wasyluk
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
- Doctoral School, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| | - Patrycja Nowicka-Stążka
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
| | - Agnieszka Zwolak
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, 20-093 Lublin, Poland; (P.N.-S.); (A.Z.)
| |
Collapse
|
8
|
Lado-Abeal J. Non-thyroidal illness syndrome, the hidden player in the septic shock induced myocardial contractile depression. Med Hypotheses 2020; 142:109775. [PMID: 32344285 DOI: 10.1016/j.mehy.2020.109775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022]
Abstract
Septic shock causes high mortality in hospitalized patients, especially in those that develop myocardial dysfunction as an early complication. The myocardial dysfunction of septic shock is characterized by a decrease in ventricular relaxation (diastolic dysfunction) and reduced ventricular ejection fraction (systolic dysfunction). Most patients with septic shock have low serum thyroid hormone levels, a condition known as non-thyroidal illness syndrome. Thyroid hormones sustain myocardial contractility and energy metabolism. Septic shock non-thyroidal illness syndrome causes myocardial hypothyroidism, and hypothyroidism causes myocardial dysfunction that resembles the myocardial depression of septic shock. We hypothesize that the myocardial hypothyroidism that occurs during septic shock has a causal role in the pathogenesis of septic shock-induced myocardial dysfunction. Thyroid hormones regulate the calcium cycle, the phenotype of contractile proteins, adrenergic response, and fatty acid transport and oxidation in the cardiomyocytes. Therefore, the administration of levothyroxine and liothyronine to normalize thyroid hormones level within the myocardium will improve the myocardial function. The hypothesis will be tested in humans with septic shock by performing a prospective, randomized, placebo-controlled study to compare the effect of thyroid hormone administration with placebo on myocardial function. The proposed hypothesis challenges the idea that non-thyroidal illness syndrome is a beneficial response of the thyroid hormone axis to illness and that thyroid hormone replacement is detrimental. The administration of thyroid hormone in order to prevent and reverse myocardial hypothyroidism during septic shock is a new theoretical concept on thyroid hormone metabolism and action at the tissue level during non-thyroidal illness syndrome. If the hypothesis is correct, clinicians should consider cardiac hypothyroidism as a central player in myocardial dysfunction caused by sepsis. Thyroid hormone replacement should be incorporated into the armamentarium of septic shock treatment.
Collapse
Affiliation(s)
- Joaquin Lado-Abeal
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Truman Medical Centers and University of Missouri Kansas City, MO, USA.
| |
Collapse
|
9
|
Zhang H, Feng YW, Yao YM. Potential therapy strategy: targeting mitochondrial dysfunction in sepsis. Mil Med Res 2018; 5:41. [PMID: 30474573 PMCID: PMC6260865 DOI: 10.1186/s40779-018-0187-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 11/08/2018] [Indexed: 12/21/2022] Open
Abstract
Recently, the definition of sepsis was concluded to be a life-threatening organ dysfunction caused by a dysregulated host response to infection. Severe patients always present with uncorrectable hypotension or hyperlactacidemia, which is defined as septic shock. The new definition emphasizes dysregulation of the host response and multiple organ dysfunction, which is partially attributed to metabolic disorders induced by energy crisis and oxidative stress. Mitochondria are a cellular organelle that are well known as the center of energy production, and mitochondrial damage or dysfunction is commonly induced in septic settings and is a predominant factor leading to a worse prognosis. In the present review, we determine the major mitochondrial disorders from morphology to functions in sepsis. In the following, several clinical or pre-clinical assays for monitoring mitochondrial function are demonstrated according to accumulated evidence, which is the first step of specific therapy targeting to modulate mitochondrial function. Accordingly, various reagents used for regulating mitochondrial enzyme activities and promoting biogenesis have been documented, among which mitochondria-targeted cation, TPP-conjugated antioxidants are the most valuable for future trials and clinical treatment to improve mitochondrial function as they may take advantage of the prognosis associated with septic complications.
Collapse
Affiliation(s)
- Hui Zhang
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing, 100048, China
| | - Yong-Wen Feng
- Department of Critical Care Medicine, The Second People's Hospital of Shenzhen, Shenzhen, 518035, China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing, 100048, China.
| |
Collapse
|
10
|
Lado-Abeal J, Martinez-Sánchez N, Cocho JA, Martín-Pastor M, Castro-Piedras I, Couce-Pico ML, Saha AK, López M. Lipopolysaccharide (LPS)-induced septic shock causes profound changes in myocardial energy metabolites in pigs. Metabolomics 2018; 14:131. [PMID: 30830414 DOI: 10.1007/s11306-018-1433-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/21/2018] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Energy deficiency is a cause for myocardial dysfunction during septic shock. In rodents, septic shock decreases the oxidation of long-chain fatty acids and glucose in the myocardium causing energy deficiency. However, the effect of septic shock on myocardial energy metabolites in large animals and human is unknown. OBJECTIVES Investigate the effects of septic shock on myocardial energy metabolites in domestic pigs. METHODS Seventeen female pigs divided into control and lipopolysaccharide (LPS)-induced septic shock groups. Myocardial metabolites were analyzed ex vivo by 1H nuclear magnetic resonance spectroscopy and liquid chromatography-tandem mass spectrometry. Gene and protein expression analysis were analyzed by real-time PCR and western blot. RESULTS Septic shock was associated with an increase in myocardial levels of short- and medium-chain acylcarnitines, lactate, alanine, and pyruvate dehydrogenase kinase 4 gene expression. COX-2 and prostaglandin E4 receptor gene expression also increased in the septic myocardium, although the only elevated eicosanoid in the septic animals was thromboxane B2. Myocardial levels of niacin, taurine, glutamate, glutamine, and glutathione were higher, and hypoxanthine levels lower in septic pigs than controls. CONCLUSIONS In pigs, septic shock induced by LPS caused myocardial changes directed to decrease the oxidation of medium- and short-chain fatty acid without an effect on long-chain fatty acid oxidation. The increase in myocardial levels of lactate, alanine, and pyruvate dehydrogenase kinase 4 gene expression suggest that septic shock decreases pyruvate dehydrogenase complex activity and glucose oxidation. Homeostasis of niacin, taurine, glutamate, glutamine, glutathione, hypoxanthine and thromboxane B2 is also affected in the septic myocardium.
Collapse
Affiliation(s)
- Joaquin Lado-Abeal
- Division of Endocrinology, Department of Internal Medicine, Texas Tech University Health Sciences Center-School of Medicine, Lubbock, TX, USA.
- Unidade de Enfermedades Tiroideas e Metabolicas (UETeM), Department of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain.
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine Truman Medical Centers, University of Missouri-Kansas City School of Medicine, 2301 Holmes Street, Kansas City, MO, 64108, USA.
| | - Noelia Martinez-Sánchez
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Jose Angel Cocho
- Unidad de Diagnóstico y Tratamiento de las Enfermedades Metabólicas, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Martín-Pastor
- Unidade de Resonancia Magnética (RIAIDT), Edif, CACTUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Isabel Castro-Piedras
- Division of Endocrinology, Department of Internal Medicine, Texas Tech University Health Sciences Center-School of Medicine, Lubbock, TX, USA
| | - M Luz Couce-Pico
- Unidad de Diagnóstico y Tratamiento de las Enfermedades Metabólicas, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Asish K Saha
- Division of Endocrinology, Diabetes and Nutrition, Boston University Medical Center, Boston, MA, USA
| | - Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain
| |
Collapse
|
11
|
Anderson RJ, Jinadasa SP, Hsu L, Ghafouri TB, Tyagi S, Joshua J, Mueller A, Talmor D, Sell RE, Beitler JR. Shock subtypes by left ventricular ejection fraction following out-of-hospital cardiac arrest. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:162. [PMID: 29907120 PMCID: PMC6003130 DOI: 10.1186/s13054-018-2078-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022]
Abstract
Background Post-resuscitation hemodynamic instability following out-of-hospital cardiac arrest (OHCA) may occur from myocardial dysfunction underlying cardiogenic shock and/or inflammation-mediated distributive shock. Distinguishing the predominant shock subtype with widely available clinical metrics may have prognostic and therapeutic value. Methods A two-hospital cohort was assembled of patients in shock following OHCA. Left ventricular ejection fraction (LVEF) was assessed via echocardiography or cardiac ventriculography within 1 day post arrest and used to delineate shock physiology. The study evaluated whether higher LVEF, indicating distributive-predominant shock physiology, was associated with neurocognitive outcome (primary endpoint), survival, and duration of multiple organ failures. The study also investigated whether volume resuscitation exhibited a subtype-specific association with outcome. Results Of 162 patients with post-resuscitation shock, 48% had normal LVEF (> 40%), consistent with distributive shock physiology. Higher LVEF was associated with less favorable neurocognitive outcome (OR 0.74, 95% CI 0.58–0.94 per 10% increase in LVEF; p = 0.01). Higher LVEF also was associated with worse survival (OR 0.81, 95% CI 0.67–0.97; p = 0.02) and fewer organ failure-free days (β = – 0.67, 95% CI – 1.28 to − 0.06; p = 0.03). Only 51% of patients received a volume challenge of at least 30 ml/kg body weight in the first 6 h post arrest, and the volume received did not differ by LVEF. Greater volume resuscitation in the first 6 h post arrest was associated with favorable neurocognitive outcome (OR 1.59, 95% CI 0.99–2.55 per liter; p = 0.03) and survival (OR 1.44, 95% CI 1.02–2.04; p = 0.02) among patients with normal LVEF but not low LVEF. Conclusions In post-resuscitation shock, higher LVEF—indicating distributive shock physiology—was associated with less favorable neurocognitive outcome, fewer days without organ failure, and higher mortality. Greater early volume resuscitation was associated with more favorable neurocognitive outcome and survival in patients with this shock subtype. Additional studies with repeated measures of complementary hemodynamic parameters are warranted to validate the clinical utility for subtyping post-resuscitation shock. Electronic supplementary material The online version of this article (10.1186/s13054-018-2078-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ryan J Anderson
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Sayuri P Jinadasa
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Leeyen Hsu
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Tiffany Bita Ghafouri
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Sanjeev Tyagi
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Jisha Joshua
- Division of Pulmonary and Critical Care Medicine, University of California San Diego, San Diego, CA, USA
| | - Ariel Mueller
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Daniel Talmor
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Rebecca E Sell
- Division of Pulmonary and Critical Care Medicine, University of California San Diego, San Diego, CA, USA
| | - Jeremy R Beitler
- Center for Acute Respiratory Failure, Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians & Surgeons, 622 W. 168th Street, 8E101, New York, NY, 10032, USA.
| |
Collapse
|
12
|
Wei X, Meng X, Yuan Y, Shen F, Li C, Yang J. Quercetin exerts cardiovascular protective effects in LPS-induced dysfunction in vivo by regulating inflammatory cytokine expression, NF-κB phosphorylation, and caspase activity. Mol Cell Biochem 2018; 446:43-52. [PMID: 29322353 DOI: 10.1007/s11010-018-3271-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/04/2018] [Indexed: 12/29/2022]
Abstract
Impaired myocardial contractile function, one of the well-documented features of sepsis, contributes greatly to the high rate of mortality. Quercetin is widely accepted as a potential antioxidant and free radical scavenger. Epidemiologic studies have suggested that an increase in the intake of dietary Quercetin can reduce the risk of cardiac disease. However, presently there is no report yet on the influence of Quercetin on LPS-induced myocardial dysfunction in vivo. Cardiovascular protective effects of Quercetin on LPS-induced sepsis in mice were measured after intragastric administration, using normal saline as a positive control. Quercetin pretreatment significantly alleviated LPS-induced cardiac abnormalities in mice. The histopathologic findings in the present study justify the findings reported from the biochemical analyses. Our observation from the present research work reveals that Quercetin suppressed the production of proinflammatory cytokines at different levels, such as TNF-α and IL-1β, and inhibits the activation of I-κB phosphorylation, whereas the total content was not affected. Apoptotic pathways are related to Quercetin protection in the development of myocardial dysfunction. In conclusion, our findings demonstrate the adjuvant potentials of Quercetin for clinical sepsis treatment.
Collapse
Affiliation(s)
- Xiqing Wei
- Qingdao University, Qingdao, 266071, Shandong, People's Republic of China.,Department of Cardiology, Affiliated Hospital of Jining Medical University, Jining, 273500, Shandong, People's Republic of China
| | - Xiangli Meng
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, Jining, 273500, Shandong, People's Republic of China
| | - Yuxiang Yuan
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, Jining, 273500, Shandong, People's Republic of China
| | - Fengjuan Shen
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, Jining, 273500, Shandong, People's Republic of China
| | - Chengqiu Li
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, Jining, 273500, Shandong, People's Republic of China.
| | - Jun Yang
- Qingdao University, Qingdao, 266071, Shandong, People's Republic of China. .,Department of Cardiology, Yantai Yuhuangding Hospital, Affiliated Hospital of Medical College Qingdao University, 20 Yudong Road, Yantai, 264000, Shandong, People's Republic of China.
| |
Collapse
|
13
|
Involvement of Mitochondrial Disorders in Septic Cardiomyopathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4076348. [PMID: 29201271 PMCID: PMC5671744 DOI: 10.1155/2017/4076348] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/11/2017] [Accepted: 09/28/2017] [Indexed: 12/29/2022]
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. It remains a leading cause of death worldwide, despite the development of various therapeutic strategies. Cardiac dysfunction, also referred to as septic cardiomyopathy, is a frequent and well-described complication of sepsis and associated with worse clinical outcomes. Recent research has increased our understanding of the role of mitochondrial dysfunction in the pathophysiology of septic cardiomyopathy. The purpose of this review is to present this evidence as a coherent whole and to highlight future research directions.
Collapse
|
14
|
Abstract
Although it is generally believed that oxidative phosphorylation and adequate oxygenation are essential for life, human development occurs in a profoundly hypoxic environment and "normal" levels of oxygen during embryogenesis are even harmful. The ability of embryos not only to survive but also to thrive in such an environment is made possible by adaptations related to metabolic pathways. Similarly, cancerous cells are able not only to survive but also to grow and spread in environments that would typically be fatal for healthy adult cells. Many biological states, both normal and pathological, share underlying similarities related to metabolism, the electron transport chain, and reactive species. The purpose of Part I of this review is to review the similarities among embryogenesis, mammalian adaptions to hypoxia (primarily driven by hypoxia-inducible factor-1), ischemia-reperfusion injury (and its relationship with reactive oxygen species), hibernation, diving animals, cancer, and sepsis, with a particular focus on the common characteristics that allow cells and organisms to survive in these states.
Collapse
Affiliation(s)
- Robert H Thiele
- From the Department of Anesthesiology, University of Virginia, Charlottesville, VA
| |
Collapse
|
15
|
Abstract
Part I of this review discussed the similarities between embryogenesis, mammalian adaptions to hypoxia (primarily driven by hypoxia-inducible factor-1 [HIF-1]), ischemia-reperfusion injury (and its relationship with reactive oxygen species), hibernation, diving animals, cancer, and sepsis, and it focused on the common characteristics that allow cells and organisms to survive in these states. Part II of this review describes techniques by which researchers gain insight into subcellular energetics and identify potential future tools for clinicians. In particular, P nuclear magnetic resonance to measure high-energy phosphates, serum lactate measurements, the use of near-infrared spectroscopy to measure the oxidation state of cytochrome aa3, and the ability of the protoporphyrin IX-triplet state lifetime technique to measure mitochondrial oxygen tension are discussed. In addition, this review discusses novel treatment strategies such as hyperbaric oxygen, preconditioning, exercise training, therapeutic gases, as well as inhibitors of HIF-1, HIF prolyl hydroxylase, and peroxisome proliferator-activated receptors.
Collapse
Affiliation(s)
- Robert H Thiele
- From the Department of Anesthesiology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
16
|
Kakihana Y, Ito T, Nakahara M, Yamaguchi K, Yasuda T. Sepsis-induced myocardial dysfunction: pathophysiology and management. J Intensive Care 2016; 4:22. [PMID: 27011791 PMCID: PMC4804632 DOI: 10.1186/s40560-016-0148-1] [Citation(s) in RCA: 310] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/04/2016] [Indexed: 12/29/2022] Open
Abstract
Sepsis is aggravated by an inappropriate immune response to invading microorganisms, which occasionally leads to multiple organ failure. Several lines of evidence suggest that the ventricular myocardium is depressed during sepsis with features of diastolic dysfunction. Potential candidates responsible for septic cardiomyopathy include pathogen-associated molecular patterns (PAMPs), cytokines, and nitric oxide. Extracellular histones and high-mobility group box 1 that function as endogenous damage-associated molecular patterns (DAMPs) also contribute to the myocardial dysfunction associated with sepsis. If untreated, persistent shock causes cellular injury and the liberation of further DAMPs. Like PAMPs, DAMPs have the potential to activate inflammation, creating a vicious circle. Early infection control with adequate antibiotic care is important during septic shock to decrease PAMPs arising from invasive microorganisms. Early aggressive fluid resuscitation as well as the administration of vasopressors and inotropes is also important to reduce DAMPs generated by damaged cells although excessive volume loading, and prolonged administration of catecholamines might be harmful. This review delineates some features of septic myocardial dysfunction, assesses its most common underlying mechanisms, and briefly outlines current therapeutic strategies and potential future approaches.
Collapse
Affiliation(s)
- Yasuyuki Kakihana
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520 Japan
| | - Takashi Ito
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520 Japan ; Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mayumi Nakahara
- Department of Anesthesiology and Critical Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Keiji Yamaguchi
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520 Japan
| | - Tomotsugu Yasuda
- Department of Emergency and Intensive Care Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8520 Japan
| |
Collapse
|
17
|
Langhorn R, Willesen JL. Cardiac Troponins in Dogs and Cats. J Vet Intern Med 2015; 30:36-50. [PMID: 26681537 PMCID: PMC4913658 DOI: 10.1111/jvim.13801] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 09/04/2015] [Accepted: 11/03/2015] [Indexed: 11/26/2022] Open
Abstract
Cardiac troponins are sensitive and specific markers of myocardial injury. The troponin concentration can be thought of as a quantitative measure of the degree of injury sustained by the heart, however, it provides no information on the cause of injury or the mechanism of troponin release. Conventionally, the cardiac troponins have been used for diagnosis of acute myocardial infarction in humans and have become the gold standard biomarkers for this indication. They have become increasingly recognized as an objective measure of cardiomyocyte status in both cardiac and noncardiac disease, supplying additional information to that provided by echocardiography and ECG. Injury to cardiomyocytes can occur through a variety of mechanisms with subsequent release of troponins. Independent of the underlying disease or the mechanism of troponin release, the presence of myocardial injury is associated with an increased risk of death. As increasingly sensitive assays are introduced, the frequent occurrence of myocardial injury is becoming apparent, and our understanding of its causes and importance is constantly evolving. Presently troponins are valuable for detecting a subgroup of patients with higher risk of death. Future research is needed to clarify whether troponins can serve as monitoring tools guiding treatment, whether administering more aggressive treatment to patients with evidence of myocardial injury is beneficial, and whether normalizing of troponin concentrations in patients presenting with evidence of myocardial injury is associated with reduced risk of death.
Collapse
Affiliation(s)
- R Langhorn
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| | - J L Willesen
- Department of Veterinary Clinical and Animal Sciences, University of Copenhagen, Frederiksberg C, Denmark
| |
Collapse
|
18
|
Berk J, Wade R, Baser HD, Lado J. Case report: severe reversible cardiomyopathy associated with systemic inflammatory response syndrome in the setting of diabetic hyperosmolar hyperglycemic non-ketotic syndrome. BMC Cardiovasc Disord 2015; 15:123. [PMID: 26466591 PMCID: PMC4606557 DOI: 10.1186/s12872-015-0112-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/24/2015] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND This case study features a woman who presented with clinical and laboratory findings consistent with hyperosmolar hyperglycemic non-ketotic syndrome (HHNS), systemic inflammatory response syndrome (SIRS), and non-thyroidal illness syndrome (NTIS) who was noted to have a transient decrease in myocardial function. To our knowledge, this is the first case discussing the overlapping pathophysiological mechanisms could increase susceptibility to SIRS-induced cardiomyopathy. It is imperative that this clinical question be investigated further as such a relationship may have significant clinical implications for prevention and future treatments, particularly in patients similar to the one presented in this clinical case. CASE PRESENTATION A 53-year old Caucasian female presented to the Emergency Department for cough, nausea, vomiting and "feeling sick for 3 weeks." Labs were indicative of diabetic ketoacidosis. Initial electrocardiograms were suggestive of possible myocardial infarction and follow-up echocardiogram showed severely depressed left ventricular systolic function which resolved upon treatment of ketoacidosis. CONCLUSION We suggest that her cardiomyopathy could have three synergistic sources: SIRS, HHNS and NTIS. Overlapping mechanisms suggest uncontrolled diabetes mellitus and NTIS could increase susceptibility to SIRS-induced cardiomyopathy as seen in this case. HHNS and SIRS cause cardiac tissue injury through mechanisms including impairment of fatty acid oxidation and formation of reactive oxygen species, as well as modifying the function of membrane calcium channels. As a result, it is conceivable that diabetes may amplify the deleterious effects of inflammatory stressors on cardiac myocytes. This novel case report offers a path for future research into prevention and treatment of SIRS-induced cardiomyopathy in, but not exclusive to, the setting of diabetes.
Collapse
Affiliation(s)
- Justin Berk
- Department of Internal Medicine, Texas Tech University Health Sciences Center, School of Medicine, 3601 4th St Stop 9410, Lubbock, TX, 79416, USA.
| | - Raymond Wade
- Department of Internal Medicine, Texas Tech University Health Sciences Center, School of Medicine, 3601 4th St Stop 9410, Lubbock, TX, 79416, USA.
| | - Hatice Duygu Baser
- Department of Internal Medicine, Texas Tech University Health Sciences Center, School of Medicine, 3601 4th St Stop 9410, Lubbock, TX, 79416, USA.
| | - Joaquin Lado
- Department of Internal Medicine, Texas Tech University Health Sciences Center, School of Medicine, 3601 4th St Stop 9410, Lubbock, TX, 79416, USA.
| |
Collapse
|
19
|
Moore J, Dyson A, Singer M, Fraser J. Microcirculatory dysfunction and resuscitation: why, when, and how. Br J Anaesth 2015; 115:366-75. [DOI: 10.1093/bja/aev163] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
20
|
Farrag M, Laufenberg LJ, Steiner JL, Weller GE, Lang CH, Ruiz-Velasco V. Modulation of voltage-gated Ca2+ channels by G protein-coupled receptors in celiac-mesenteric ganglion neurons of septic rats. PLoS One 2015; 10:e0125566. [PMID: 26017846 PMCID: PMC4446366 DOI: 10.1371/journal.pone.0125566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/25/2015] [Indexed: 12/25/2022] Open
Abstract
Septic shock, the most severe complication associated with sepsis, is manifested by tissue hypoperfusion due, in part, to cardiovascular and autonomic dysfunction. In many cases, the splanchnic circulation becomes vasoplegic. The celiac-superior mesenteric ganglion (CSMG) sympathetic neurons provide the main autonomic input to these vessels. We used the cecal ligation puncture (CLP) model, which closely mimics the hemodynamic and metabolic disturbances observed in septic patients, to examine the properties and modulation of Ca2+ channels by G protein-coupled receptors in acutely dissociated rat CSMG neurons. Voltage-clamp studies 48 hr post-sepsis revealed that the Ca2+ current density in CMSG neurons from septic rats was significantly lower than those isolated from sham control rats. This reduction coincided with a significant increase in membrane surface area and a negligible increase in Ca2+ current amplitude. Possible explanations for these findings include either cell swelling or neurite outgrowth enhancement of CSMG neurons from septic rats. Additionally, a significant rightward shift of the concentration-response relationship for the norepinephrine (NE)-mediated Ca2+ current inhibition was observed in CSMG neurons from septic rats. Testing for the presence of opioid receptor subtypes in CSMG neurons, showed that mu opioid receptors were present in ~70% of CSMG, while NOP opioid receptors were found in all CSMG neurons tested. The pharmacological profile for both opioid receptor subtypes was not significantly affected by sepsis. Further, the Ca2+ current modulation by propionate, an agonist for the free fatty acid receptors GPR41 and GPR43, was not altered by sepsis. Overall, our findings suggest that CSMG function is affected by sepsis via changes in cell size and α2-adrenergic receptor-mediated Ca2+ channel modulation.
Collapse
Affiliation(s)
- Mohamed Farrag
- Department of Anesthesiology, Penn State College of Medicine, Hershey, PA, United States of America
| | - Lacee J. Laufenberg
- Department of Surgery, Penn State College of Medicine, Hershey, PA, United States of America
| | - Jennifer L. Steiner
- Department of Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, PA, United States of America
| | - Gregory E. Weller
- Department of Anesthesiology, Penn State College of Medicine, Hershey, PA, United States of America
| | - Charles H. Lang
- Department of Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, PA, United States of America
| | - Victor Ruiz-Velasco
- Department of Anesthesiology, Penn State College of Medicine, Hershey, PA, United States of America
- * E-mail:
| |
Collapse
|
21
|
Lee I, Hüttemann M. Energy crisis: the role of oxidative phosphorylation in acute inflammation and sepsis. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1579-86. [PMID: 24905734 PMCID: PMC4147665 DOI: 10.1016/j.bbadis.2014.05.031] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/17/2014] [Accepted: 05/27/2014] [Indexed: 12/13/2022]
Abstract
Mitochondrial dysfunction is increasingly recognized as an accomplice in most of the common human diseases including cancer, neurodegeneration, diabetes, ischemia/reperfusion injury as seen in myocardial infarction and stroke, and sepsis. Inflammatory conditions, both acute and chronic, have recently been shown to affect mitochondrial function. We here discuss the role of oxidative phosphorylation (OxPhos), focusing on acute inflammatory conditions, in particular sepsis and experimental sepsis models. We discuss mitochondrial alterations, specifically the suppression of oxidative metabolism and the role of mitochondrial reactive oxygen species in disease pathology. Several signaling pathways including metabolic, proliferative, and cytokine signaling affect mitochondrial function and appear to be important in inflammatory disease conditions. Cytochrome c oxidase (COX) and cytochrome c, the latter of which plays a central role in apoptosis in addition to mitochondrial respiration, serve as examples for the entire OxPhos system since they have been studied in more detail with respect to cell signaling. We propose a model in which inflammatory signaling leads to changes in the phosphorylation state of mitochondrial proteins, including Tyr304 phosphorylation of COX catalytic subunit I. This results in an inhibition of OxPhos, a reduction of the mitochondrial membrane potential, and consequently a lack of energy, which can cause organ failure and death as seen in septic patients.
Collapse
Affiliation(s)
- Icksoo Lee
- College of Medicine, Dankook University, Cheonan-si, Chungcheongnam-do 330-714, Republic of Korea
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA; Cardiovascular Research Institute, Wayne State University, Detroit, MI 48201, USA; Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, MI 48201, USA; Karmanos Cancer Institute, Detroit, MI 48201, USA.
| |
Collapse
|
22
|
Quinones QJ, Ma Q, Zhang Z, Barnes BM, Podgoreanu MV. Organ protective mechanisms common to extremes of physiology: a window through hibernation biology. Integr Comp Biol 2014; 54:497-515. [PMID: 24848803 DOI: 10.1093/icb/icu047] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Supply and demand relationships govern survival of animals in the wild and are also key determinants of clinical outcomes in critically ill patients. Most animals' survival strategies focus on the supply side of the equation by pursuing territory and resources, but hibernators are able to anticipate declining availability of nutrients by reducing their energetic needs through the seasonal use of torpor, a reversible state of suppressed metabolic demand and decreased body temperature. Similarly, in clinical medicine the majority of therapeutic interventions to care for critically ill or trauma patients remain focused on elevating physiologic supply above critical thresholds by increasing the main determinants of delivery of oxygen to the tissues (cardiac output, perfusion pressure, hemoglobin concentrations, and oxygen saturation), as well as increasing nutritional support, maintaining euthermia, and other general supportive measures. Techniques, such as induced hypothermia and preconditioning, aimed at diminishing a patient's physiologic requirements as a short-term strategy to match reduced supply and to stabilize their condition, are few and underutilized in clinical settings. Consequently, comparative approaches to understand the mechanistic adaptations that suppress metabolic demand and alter metabolic use of fuel as well as the application of concepts gleaned from studies of hibernation, to the care of critically ill and injured patients could create novel opportunities to improve outcomes in intensive care and perioperative medicine.
Collapse
Affiliation(s)
- Quintin J Quinones
- *Department of Anesthesiology, Systems Modeling of Perioperative Organ Injury Laboratory, Duke University, Box 3094, Durham, NC 27710, USA; Institute for Arctic Biology, University of Alaska, Fairbanks, AK, USA
| | - Qing Ma
- *Department of Anesthesiology, Systems Modeling of Perioperative Organ Injury Laboratory, Duke University, Box 3094, Durham, NC 27710, USA; Institute for Arctic Biology, University of Alaska, Fairbanks, AK, USA
| | - Zhiquan Zhang
- *Department of Anesthesiology, Systems Modeling of Perioperative Organ Injury Laboratory, Duke University, Box 3094, Durham, NC 27710, USA; Institute for Arctic Biology, University of Alaska, Fairbanks, AK, USA
| | - Brian M Barnes
- *Department of Anesthesiology, Systems Modeling of Perioperative Organ Injury Laboratory, Duke University, Box 3094, Durham, NC 27710, USA; Institute for Arctic Biology, University of Alaska, Fairbanks, AK, USA
| | - Mihai V Podgoreanu
- *Department of Anesthesiology, Systems Modeling of Perioperative Organ Injury Laboratory, Duke University, Box 3094, Durham, NC 27710, USA; Institute for Arctic Biology, University of Alaska, Fairbanks, AK, USA*Department of Anesthesiology, Systems Modeling of Perioperative Organ Injury Laboratory, Duke University, Box 3094, Durham, NC 27710, USA; Institute for Arctic Biology, University of Alaska, Fairbanks, AK, USA
| |
Collapse
|
23
|
Romero-Bermejo FJ, Ruiz-Bailen M, Gil-Cebrian J, Huertos-Ranchal MJ. Sepsis-induced cardiomyopathy. Curr Cardiol Rev 2013; 7:163-83. [PMID: 22758615 PMCID: PMC3263481 DOI: 10.2174/157340311798220494] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 02/24/2011] [Accepted: 02/24/2011] [Indexed: 01/20/2023] Open
Abstract
Myocardial dysfunction is one of the main predictors of poor outcome in septic patients, with mortality rates next to 70%. During the sepsis-induced myocardial dysfunction, both ventricles can dilate and diminish its ejection fraction, having less response to fluid resuscitation and catecholamines, but typically is assumed to be reversible within 7-10 days. In the last 30 years, It´s being subject of substantial research; however no explanation of its etiopathogenesis or effective treatment have been proved yet. The aim of this manuscript is to review on the most relevant aspects of the sepsis-induced myocardial dysfunction, discuss its clinical presentation, pathophysiology, etiopathogenesis, diagnostic tools and therapeutic strategies proposed in recent years.
Collapse
Affiliation(s)
- Francisco J Romero-Bermejo
- Intensive Care Unit, Critical Care and Emergency Department, Puerto Real University Hospital, Cadiz, Spain.
| | | | | | | |
Collapse
|
24
|
Early functional and transcriptomic changes in the myocardium predict outcome in a long-term rat model of sepsis. Clin Sci (Lond) 2013; 124:391-401. [PMID: 22988837 DOI: 10.1042/cs20120334] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Myocardial function is depressed in sepsis and is an important prognosticator in the human condition. Using echocardiography in a long-term fluid-resuscitated Wistar rat model of faecal peritonitis we investigated whether depressed myocardial function could be detected at an early stage of sepsis and, if so, whether the degree of depression could predict eventual outcome. At 6 h post-insult, a stroke volume <0.17 ml prognosticated 3-day mortality with positive and negative predictive values of 93 and 80%, respectively. Subsequent fluid loading studies demonstrated intrinsic myocardial depression with poor-prognosis animals tolerating less fluid than either good-prognosis or sham-operated animals. Cardiac gene expression analysis at 6 h detected 527 transcripts significantly up- or down-regulated by the septic process, including genes related to inflammatory and cell cycle pathways. Predicted mortality was associated with significant differences in transcripts of genes expressing proteins related to the TLR2/MyD88 (Toll-like receptor 2/myeloid differentiation factor 88) and JAK/STAT (Janus kinase/signal transducer and activator of transcription) inflammatory pathways, β-adrenergic signalling and intracellular calcium cycling. Our findings highlight the presence of myocardial depression in early sepsis and its prognostic significance. Transcriptomic analysis in heart tissue identified changes in signalling pathways that correlated with clinical dysfunction. These pathways merit further study to both better understand and potentially modify the disease process.
Collapse
|
25
|
Takasu O, Gaut JP, Watanabe E, To K, Fagley RE, Sato B, Jarman S, Efimov IR, Janks DL, Srivastava A, Bhayani SB, Drewry A, Swanson PE, Hotchkiss RS. Mechanisms of cardiac and renal dysfunction in patients dying of sepsis. Am J Respir Crit Care Med 2013; 187:509-17. [PMID: 23348975 DOI: 10.1164/rccm.201211-1983oc] [Citation(s) in RCA: 341] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
RATIONALE The mechanistic basis for cardiac and renal dysfunction in sepsis is unknown. In particular, the degree and type of cell death is undefined. OBJECTIVES To evaluate the degree of sepsis-induced cardiomyocyte and renal tubular cell injury and death. METHODS Light and electron microscopy and immunohistochemical staining for markers of cellular injury and stress, including connexin-43 and kidney-injury-molecule-1 (Kim-1), were used in this study. MEASUREMENTS AND MAIN RESULTS Rapid postmortem cardiac and renal harvest was performed in 44 septic patients. Control hearts were obtained from 12 transplant and 13 brain-dead patients. Control kidneys were obtained from 20 trauma patients and eight patients with cancer. Immunohistochemistry demonstrated low levels of apoptotic cardiomyocytes (<1-2 cells per thousand) in septic and control subjects and revealed redistribution of connexin-43 to lateral membranes in sepsis (P < 0.020). Electron microscopy showed hydropic mitochondria only in septic specimens, whereas mitochondrial membrane injury and autophagolysosomes were present equally in control and septic specimens. Control kidneys appeared relatively normal by light microscopy; 3 of 20 specimens showed focal injury in approximately 1% of renal cortical tubules. Conversely, focal acute tubular injury was present in 78% of septic kidneys, occurring in 10.3 ± 9.5% and 32.3 ± 17.8% of corticomedullary-junction tubules by conventional light microscopy and Kim-1 immunostains, respectively (P < 0.01). Electron microscopy revealed increased tubular injury in sepsis, including hydropic mitochondria and increased autophagosomes. CONCLUSIONS Cell death is rare in sepsis-induced cardiac dysfunction, but cardiomyocyte injury occurs. Renal tubular injury is common in sepsis but presents focally; most renal tubular cells appear normal. The degree of cell injury and death does not account for severity of sepsis-induced organ dysfunction.
Collapse
Affiliation(s)
- Osamu Takasu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Protective effects of magnesium supplementation on metabolic energy derangements in lipopolysaccharide-induced cardiotoxicity in mice. Eur J Pharmacol 2012; 694:75-81. [PMID: 22939974 DOI: 10.1016/j.ejphar.2012.07.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Revised: 07/11/2012] [Accepted: 07/27/2012] [Indexed: 12/27/2022]
Abstract
Metabolic derangements and bioenergetic failure are major contributors to sepsis-induced multiple organ dysfunctions. Due to the well known role of magnesium (Mg) as a cofactor in many enzymatic reactions that involve energy creation and utilization, the present investigation was directed to estimate the cardioprotective effect of Mg supplementation in lipopolysaccharide (LPS)-induced metabolic energy changes in mice. Oral doses of Mg aspartate (20 or 40 mg/kg) were administered once daily for 7 day. Mice were then subjected to a single intraperitoneal injection of LPS (2 mg/kg). Plasma was separated 3 h after LPS injection for determination of creatine kinase-MB activity. Animals were then sacrificed and the hearts were separated for estimation of tissue thiobarbituric acid reactive substances, reduced glutathione, lactate, pyruvate, adenine nucleotides, creatine phosphate and cardiac Na(+),K(+)-ATPase activity. Finally, electron microscopic examination was performed to visualize the protective effects of Mg pretreatment on mitochondrial ultrastructure. In general, the higher dose of Mg was more effective than the lower dose in ameliorating creatine kinase-MB elevation and the state of oxidative stress, lactate accumulation, pyruvate reduction as well as preserving creatine phosphate, adenine nucleotides and Na(+),K(+)-ATPase activity. Moreover, the higher dose of Mg provided a significant cardioprotection against the mitochondrial ultrastructural changes. Mg therapy can afford a significant protection against metabolic energy derangements and mitochondrial ultrastructural changes induced by LPS cardiotoxicity in mice.
Collapse
|
27
|
Thaliporphine preserves cardiac function of endotoxemic rabbits by both directly and indirectly attenuating NFκB signaling pathway. PLoS One 2012; 7:e39174. [PMID: 22761733 PMCID: PMC3382609 DOI: 10.1371/journal.pone.0039174] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 05/16/2012] [Indexed: 11/19/2022] Open
Abstract
Cardiac depression in sepsis is associated with the increased morbidity and mortality. Although myofilaments damage, autonomic dysfunction, and apoptosis play roles in sepsis-induced myocardial dysfunction, the underlying mechanism is not clear. All of these possible factors are related to NFκB signaling, which plays the main role in sepsis signaling. Thaliporphine was determined to possess anti-inflammatory and cardioprotective activity by suppressing NFκB signaling in rodents. The purpose of this study is to further prove this protective effect in larger septic animals, and try to find the underlying mechanisms. The systolic and diastolic functions were evaluated in vivo by pressure-volume analysis at different preloads. Both preload-dependent and -independent hemodynamic parameters were performed. Inflammatory factors of whole blood and serum samples were analyzed. Several sepsis-related signaling pathways were also determined at protein level. Changes detected by conductance catheter showed Thaliporphine could recover impaired left ventricular systolic function after 4 hours LPS injection. It could also reverse the LPS induced steeper EDPVR and gentler ESPVR, thus improve Ees, Ea, and PRSW. Thaliporphine may exert this protective effect by decreasing TNFα and caspase3 dependent cell apoptosis, which was consistent with the decreased serum cTnI and LDH concentration. Thaliporphine could protect sepsis-associated myocardial dysfunction in both preload-dependent and -independent ways. It may exert these protective effects by both increase of "good"-PI3K/Akt/mTOR and decrease of "bad"-p38/NFκB pathways, which followed by diminishing TNFα and caspase3 dependent cell apoptosis.
Collapse
|
28
|
Ishikawa K, Wan L, Calzavacca P, Bellomo R, Bailey M, May CN. The effects of terlipressin on regional hemodynamics and kidney function in experimental hyperdynamic sepsis. PLoS One 2012; 7:e29693. [PMID: 22355305 PMCID: PMC3280248 DOI: 10.1371/journal.pone.0029693] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 12/02/2011] [Indexed: 01/29/2023] Open
Abstract
Background and Aims Although terlipressin (TP) may improve renal function in cirrhotic patients, its use in sepsis remains controversial due to concerns about regional ischemia. We investigated the effects of TP on regional hemodynamics and kidney function in experimental hyperdynamic sepsis. Methods We studied thirteen merino ewes in a university physiology laboratory using a randomized controlled cross over design. We implanted flow probes around the pulmonary, circumflex coronary, superior mesenteric, renal and iliac arteries. We injected live Escherichia coli and induced hyperdynamic sepsis. We treated animals with either bolus vehicle or a single dose of TP (sTP = 1 mg). In a second group, after 1 mg of TP, two additional bolus injections (mTP) of 0.5 mg were given at 2 hourly intervals. Main Results sTP (1 mg) significantly increased mean arterial pressure (MAP) (74 to 89 mmHg; P<0.0001) creatinine clearance (31 to 85 mL/min; P<0.0001) and urine output (24 to 307 mL/hr) (P<0.0001). However, it decreased CO (5.7 to 3.9 L/min; p<0.0001), coronary blood flow (CBF) (43 to 32 mL/min; p<0.0001) and mesenteric blood flow (MBF) (944 to 625 mL/min; p = 0.004) and increased blood lactate (2.1 to 4.0 mmol/L; p<0.0001). Extra doses of TP caused little additional effect. Conclusions In hyperdynamic sepsis, bolus TP transiently improves MAP and renal function, but reduces CO, CBF and MBF, and increases blood lactate. Caution should be applied when prescribing bolus TP in septic patients at risk of coronary or mesenteric ischemia.
Collapse
Affiliation(s)
- Ken Ishikawa
- Howard Florey Institute, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
29
|
Alders DJC, Groeneveld ABJ, Binsl TW, de Kanter FJ, van Beek JHGM. Endotoxemia decreases matching of regional blood flow and O2 delivery to O2 uptake in the porcine left ventricle. Am J Physiol Heart Circ Physiol 2011; 300:H1459-66. [PMID: 21297021 DOI: 10.1152/ajpheart.00287.2010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heterogeneity of regional coronary blood flow is caused in part by heterogeneity in O(2) demand in the normal heart. We investigated whether myocardial O(2) supply/demand mismatching is associated with the myocardial depression of sepsis. Regional blood flow (microspheres) and O(2) uptake ([(13)C]acetate infusion and analysis of resultant NMR spectra) were measured in about nine contiguous tissue samples from the left ventricle (LV) in each heart. Endotoxemic pigs (n = 9) showed hypotension at unchanged cardiac output with a fall in LV stroke work and first derivative of LV pressure relative to controls (n = 4). Global coronary blood flow and O(2) delivery were maintained. Lactate accumulated in arterial blood, but net lactate extraction across the coronary bed was unchanged during endotoxemia. When LV O(2) uptake based on blood gas versus NMR data were compared, the correlation was 0.73 (P = 0.007). While stable over time in controls, regional blood flows were strongly redistributed during endotoxin shock, with overall flow heterogeneity unchanged. A stronger redistribution of blood flow with endotoxin was associated with a larger fall in LV function parameters. Moreover, the correlation of regional O(2) delivery to uptake fell from r = 0.73 (P < 0.001) in control to r = 0.18 (P = 0.25, P = 0.009 vs. control) in endotoxemic hearts. The results suggest a redistribution of LV regional coronary blood flow during endotoxin shock in pigs, with regional O(2) delivery mismatched to O(2) demand. Mismatching may underlie, at least in part, the myocardial depression of sepsis.
Collapse
Affiliation(s)
- David J C Alders
- Department of Intensive Care, Vrije Universiteit, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
30
|
Shi W, Cui N, Wu Z, Yang Y, Zhang S, Gai H, Zhu D, Jiang C. Lipopolysaccharides up-regulate Kir6.1/SUR2B channel expression and enhance vascular KATP channel activity via NF-kappaB-dependent signaling. J Biol Chem 2010; 285:3021-9. [PMID: 19959479 PMCID: PMC2823456 DOI: 10.1074/jbc.m109.058313] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 12/01/2009] [Indexed: 12/31/2022] Open
Abstract
Sepsis is a severe medical condition causing a large number of deaths worldwide. Recent studies indicate that the septic susceptibility is attributable to the vascular ATP-sensitive K(+) (K(ATP)) channel. However, the mechanisms underlying the channel modulation in sepsis are still unclear. Here we show evidence for the modulation of vascular K(ATP) channel by septic pathogen lipopolysaccharides (LPS). In isolated mesenteric arterial rings, phenylephrine (PE) produced concentration-dependent vasoconstriction that was relaxed by pinacidil, a selective K(ATP) channel opener. The PE response was disrupted with a LPS treatment. In acutely dissociated aortic smooth myocytes the LPS treatment augmented K(ATP) channel activity, and hyperpolarized the cells. Quantitative PCR analysis showed that LPS raised Kir6.1 and SUR2B transcripts in a concentration-dependent manner, which was suppressed by transcriptional inhibition. Consistently, the same LPS treatment did not affect Kir6.1/SUR2B channels in a heterologous expression system. The LPS effect on Kir6.1 and SUR2B expression was abolished in the presence of NF-kappaB inhibitors. Several other Toll-like receptor ligands also stimulated Kir6.1 and SUR2B expression to a similar degree as LPS. Thus, the effect of LPS on vasodilation involves up-regulation of K(ATP) channel expression, in which the NF-kappaB-dependent signaling plays an important role.
Collapse
Affiliation(s)
- Weiwei Shi
- From the Department of Biology, Georgia State University, Atlanta, Georgia 30303 and
| | - Ningren Cui
- From the Department of Biology, Georgia State University, Atlanta, Georgia 30303 and
| | - Zhongying Wu
- From the Department of Biology, Georgia State University, Atlanta, Georgia 30303 and
| | - Yang Yang
- From the Department of Biology, Georgia State University, Atlanta, Georgia 30303 and
| | - Shuang Zhang
- the School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hongyu Gai
- From the Department of Biology, Georgia State University, Atlanta, Georgia 30303 and
| | - Daling Zhu
- the School of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Chun Jiang
- From the Department of Biology, Georgia State University, Atlanta, Georgia 30303 and
| |
Collapse
|
31
|
Ceylan-Isik AF, Zhao P, Zhang B, Xiao X, Su G, Ren J. Cardiac overexpression of metallothionein rescues cardiac contractile dysfunction and endoplasmic reticulum stress but not autophagy in sepsis. J Mol Cell Cardiol 2009; 48:367-78. [PMID: 19914257 DOI: 10.1016/j.yjmcc.2009.11.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 10/21/2009] [Accepted: 11/06/2009] [Indexed: 01/01/2023]
Abstract
Sepsis is characterized by systematic inflammation where oxidative damage plays a key role in organ failure. This study was designed to examine the impact of the antioxidant metallothionein (MT) on lipopolysaccharide (LPS)-induced cardiac contractile and intracellular Ca(2+) dysfunction, oxidative stress, endoplasmic reticulum (ER) stress and autophagy. Mechanical and intracellular Ca(2+) properties were examined in hearts from FVB and cardiac-specific MT overexpression mice treated with LPS. Oxidative stress, activation of mitogen-activated protein kinase pathways (ERK, JNK and p38), ER stress, autophagy and inflammatory markers iNOS and TNFalpha were evaluated. Our data revealed enlarged end systolic diameter, decreased fractional shortening, myocyte peak shortening and maximal velocity of shortening/relengthening as well as prolonged duration of relengthening in LPS-treated FVB mice associated with reduced intracellular Ca(2+) release and decay. LPS treatment promoted oxidative stress (reduced glutathione/glutathione disulfide ratio and ROS generation). Western blot analysis revealed greater iNOS and TNFalpha, activation of ERK, JNK and p38, upregulation of ER stress markers GRP78, Gadd153, PERK and IRE1alpha, as well as the autophagy markers Beclin-1, LCB3 and Atg7 in LPS-treated mouse hearts without any change in total ERK, JNK and p38. Interestingly, these LPS-induced changes in echocardiographic, cardiomyocyte mechanical and intracellular Ca(2+) properties, ROS, stress signaling and ER stress (but not autophagy, iNOS and TNFalpha) were ablated by MT. Antioxidant N-acetylcysteine and the ER stress inhibitor tauroursodeoxycholic acid reversed LPS-elicited depression in cardiomyocyte contractile function. LPS activated AMPK and its downstream signaling ACC in conjunction with an elevated AMP/ATP ratio, which was unaffected by MT. Taken together, our data favor a beneficial effect of MT in the management of cardiac dysfunction in sepsis.
Collapse
Affiliation(s)
- Asli F Ceylan-Isik
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | | | | | | | | | | |
Collapse
|
32
|
Supinski GS, Murphy MP, Callahan LA. MitoQ administration prevents endotoxin-induced cardiac dysfunction. Am J Physiol Regul Integr Comp Physiol 2009; 297:R1095-102. [PMID: 19657095 DOI: 10.1152/ajpregu.90902.2008] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Sepsis elicits severe alterations in cardiac function, impairing cardiac mitochondrial and pressure-generating capacity. Currently, there are no therapies to prevent sepsis-induced cardiac dysfunction. We tested the hypothesis that administration of a mitochondrially targeted antioxidant, 10-(6'-ubiquinonyl)-decyltriphenylphosphonium (MitoQ), would prevent endotoxin-induced reductions in cardiac mitochondrial and contractile function. Studies were performed on adult rodents (n = 52) given either saline, endotoxin (8 mg x kg(-1) x day(-1)), saline + MitoQ (500 microM), or both endotoxin and MitoQ. At 48 h animals were killed and hearts were removed for determination of either cardiac mitochondrial function (using polarography) or cardiac pressure generation (using the Langendorf technique). We found that endotoxin induced reductions in mitochondrial state 3 respiration rates, the respiratory control ratio, and ATP generation. Moreover, MitoQ administration prevented each of these endotoxin-induced abnormalities, P < 0.001. We also found that endotoxin produced reductions in cardiac pressure-generating capacity, reducing the systolic pressure-diastolic relationship. MitoQ also prevented endotoxin-induced reductions in cardiac pressure generation, P < 0.01. One potential link between mitochondrial and contractile dysfunction is caspase activation; we found that endotoxin increased cardiac levels of active caspases 9 and 3 (P < 0.001), while MitoQ prevented this increase (P < 0.01). These data demonstrate that MitoQ is a potent inhibitor of endotoxin-induced mitochondrial and cardiac abnormalities. We speculate that this agent may prove a novel therapy for sepsis-induced cardiac dysfunction.
Collapse
Affiliation(s)
- G S Supinski
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Kentucky, Lexington, Kentucky 40536-0284, USA.
| | | | | |
Collapse
|
33
|
Flierl MA, Rittirsch D, Huber-Lang MS, Sarma JV, Ward PA. Molecular events in the cardiomyopathy of sepsis. Mol Med 2008; 14:327-36. [PMID: 18256728 DOI: 10.2119/2007-00130.flierl] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 01/28/2008] [Indexed: 01/22/2023] Open
Abstract
Septic cardiomyopathy is a well-described complication of severe sepsis and septic shock. However, the interplay of its underlying mechanisms remains enigmatic. Consequently, we constantly add to our pathophysiological understanding of septic cardiomyopathy. Various cardiosuppressive mediators have been discovered, as have multiple molecular mechanisms (alterations of myocardial calcium homeostasis, mitochondrial dysfunction, and myocardial apoptosis) that may be involved in myocardial dysfunction during sepsis. Finally, the detrimental roles of nitric oxide and peroxynitrite have been unraveled. Here, we describe our present understanding of systemic, supracellular, and cellular molecular mechanisms involved in sepsis-induced myocardial suppression.
Collapse
Affiliation(s)
- Michael A Flierl
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0602, United States of America
| | | | | | | | | |
Collapse
|
34
|
Chew MS, Johansson A, Anderson C, Ersson A, Tønnesen E. Decreases in myocardial glucose and increases in pyruvate but not ischaemia are observed during porcine endotoxaemia. Acta Anaesthesiol Scand 2008; 52:959-68. [PMID: 18494850 DOI: 10.1111/j.1399-6576.2008.01603.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Myocardial dysfunction occurs commonly in septic shock. It is not known whether this is due to local ischaemia and metabolic disturbances. Our hypothesis was that endotoxaemic myocardial dysfunction may be associated with interstitial ischaemic and metabolic changes, measured using interstitial microdialysis (MD). METHODS Eighteen pigs were randomized to control (n=6) or endotoxin infusion (n=12). MD catheters were inserted into the myocardium for measurement of interstitial glucose, pyruvate and lactate concentrations. Plasma glucose and lactate concentrations and systemic haemodynamic parameters were measured simultaneously. RESULTS Compared with the control group, the endotoxaemic animals had significantly decreased left ventricular stroke work and venous oxygen saturation (SvO2), and increased mean pulmonary artery pressure and plasma lactate. In the endotoxaemic group, decreases in interstitial glucose were observed, occurring simultaneously with increases in interstitial pruvate. Interstitial lactate: pyruvate ratios decreased with time in all animals. CONCLUSIONS Despite severe systemic and pulmonary haemodynamic changes, interstitial MD measurements revealed no evidence of anaerobic metabolism in the myocardium of endotoxaemic pigs. There were, however, changes in glucose and pyruvate concentrations, suggesting local energy metabolic disturbances.
Collapse
Affiliation(s)
- M S Chew
- Department of Intensive Care Medicine, Malmö University Hospital, Malmö, Sweden.
| | | | | | | | | |
Collapse
|
35
|
Levy RJ, Deutschman CS. Deficient mitochondrial biogenesis in critical illness: cause, effect, or epiphenomenon? CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 11:158. [PMID: 17764588 PMCID: PMC2206497 DOI: 10.1186/cc6098] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Recent studies indicate that mitochondrial dysfunction plays a role in the pathogenesis of a number of disease states. The importance of these organelles in shock and multiple organ dysfunction is of particular interest to those caring for the critically ill. Mitochondria have their own unique DNA (mtDNA) that encodes 13 essential subunits of electron transport chain enzymes, two ribosomal RNAs and 22 transfer RNAs. Importantly, mtDNA is especially susceptible to deletions, rearrangements and mutations because it is not bound by histones and lacks the extensive repair machinery present in the nucleus. The study by Côté et al. in this issue of Critical Care examines changes in mtDNA in critically ill patients. The results support further investigation into the role of mtDNA in the critically ill.
Collapse
Affiliation(s)
- Richard J Levy
- Maria Fareri Children's Hospital of Westchester Medical Center, New York Medical College, Valhalla, New York, USA
| | - Clifford S Deutschman
- Department of Anesthesiology and Critical Care and the Stavropoulos Sepsis Research Program, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
36
|
Abstract
Sepsis is generally viewed as a disease aggravated by an inappropriate immune response encountered in the afflicted individual. As an important organ system frequently compromised by sepsis and always affected by septic shock, the cardiovascular system and its dysfunction during sepsis have been studied in clinical and basic research for more than 5 decades. Although a number of mediators and pathways have been shown to be associated with myocardial depression in sepsis, the precise cause remains unclear to date. There is currently no evidence supporting global ischemia as an underlying cause of myocardial dysfunction in sepsis; however, in septic patients with coexistent and possibly undiagnosed coronary artery disease, regional myocardial ischemia or infarction secondary to coronary artery disease may certainly occur. A circulating myocardial depressant factor in septic shock has long been proposed, and potential candidates for a myocardial depressant factor include cytokines, prostanoids, and nitric oxide, among others. Endothelial activation and induction of the coagulatory system also contribute to the pathophysiology in sepsis. Prompt and adequate antibiotic therapy accompanied by surgical removal of the infectious focus, if indicated and feasible, is the mainstay and also the only strictly causal line of therapy. In the presence of severe sepsis and septic shock, supportive treatment in addition to causal therapy is mandatory. The purpose of this review is to delineate some characteristics of septic myocardial dysfunction, to assess the most commonly cited and reported underlying mechanisms of cardiac dysfunction in sepsis, and to briefly outline current therapeutic strategies and possible future approaches.
Collapse
Affiliation(s)
- M W Merx
- Department of Medicine, RWTH Aachen University, Aachen, Germany.
| | | |
Collapse
|
37
|
Minneci PC, Deans KJ, Hansen B, Parent C, Romines C, Gonzales DA, Ying SX, Munson P, Suffredini AF, Feng J, Solomon MA, Banks SM, Kern SJ, Danner RL, Eichacker PQ, Natanson C, Solomon SB. A canine model of septic shock: balancing animal welfare and scientific relevance. Am J Physiol Heart Circ Physiol 2007; 293:H2487-500. [PMID: 17644570 DOI: 10.1152/ajpheart.00589.2007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A shock canine pneumonia model that permitted relief of discomfort with the use of objective criteria was developed and validated. After intrabronchial Staphylococcus aureus challenge, mechanical ventilation, antibiotics, fluids, vasopressors, sedatives, and analgesics were titrated based on algorithms for 96 h. Increasing S. aureus (1 to 8 x 10(9) colony-forming units/kg) produced decreasing survival rates (P = 0.04). From 4 to 96 h, changes in arterial-alveolar oxygen gradients, mean pulmonary artery pressure, IL-1, serum sodium levels, mechanical ventilation, and vasopressor support were ordered based on survival time [acute nonsurvivors (< or =24 h until death, n = 8) > or = subacute nonsurvivors (>24 to 96 h until death, n = 8) > or = survivors (> or =96 h until death, n = 22) (all P < 0.05)]. In the first 12 h, increases in lactate and renal abnormalities were greatest in acute nonsurvivors (all P < 0.05). Compared with survivors, subacute nonsurvivors had greater rises in cytokines and liver enzymes and greater falls in platelets, white cell counts, pH, and urine output from 24 to 96 h (all P < 0.05). Importantly, these changes were not attributable to dosages of sedation, which decreased in nonsurvivors [survivors vs. nonsurvivors: 5.0 +/- 1.0 vs. 3.8 +/- 0.7 ml x h(-1) x (fentanyl/midazolam/ medetomidine)(-1); P = 0.02]. In this model, the pain control regimen did not mask changes in metabolic function and lung injury or the need for more hemodynamic and pulmonary support related to increasing severity of sepsis. The integration into this model of both specific and supportive titrated therapies routinely used in septic patients may provide a more realistic setting to evaluate therapies for sepsis.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animal Welfare
- Animals
- Anti-Bacterial Agents/pharmacology
- Biomedical Research/methods
- Blood Chemical Analysis
- Blood Proteins/metabolism
- Cytokines/blood
- Disease Models, Animal
- Dogs
- Fluid Therapy
- Hematologic Tests
- Hypnotics and Sedatives/pharmacology
- Kidney Diseases/microbiology
- Kidney Function Tests
- Liver Diseases/microbiology
- Liver Function Tests
- Pneumonia, Staphylococcal/blood
- Pneumonia, Staphylococcal/complications
- Pneumonia, Staphylococcal/microbiology
- Pneumonia, Staphylococcal/physiopathology
- Pneumonia, Staphylococcal/therapy
- Reproducibility of Results
- Respiration, Artificial
- Severity of Illness Index
- Shock, Septic/blood
- Shock, Septic/complications
- Shock, Septic/microbiology
- Shock, Septic/physiopathology
- Shock, Septic/therapy
- Staphylococcus aureus
- Time Factors
- Vasoconstrictor Agents
Collapse
Affiliation(s)
- Peter C Minneci
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
OBJECTIVES To review mechanisms underlying sepsis-induced cardiac dysfunction in general and intrinsic myocardial depression in particular. DATA SOURCE MEDLINE database. DATA SYNTHESIS Myocardial depression is a well-recognized manifestation of organ dysfunction in sepsis. Due to the lack of a generally accepted definition and the absence of large epidemiologic studies, its frequency is uncertain. Echocardiographic studies suggest that 40% to 50% of patients with prolonged septic shock develop myocardial depression, as defined by a reduced ejection fraction. Sepsis-related changes in circulating volume and vessel tone inevitably affect cardiac performance. Although the coronary circulation during sepsis is maintained or even increased, alterations in the microcirculation are likely. Mitochondrial dysfunction, another feature of sepsis-induced organ dysfunction, will also place the cardiomyocytes at risk of adenosine triphosphate depletion. However, clinical studies have demonstrated that myocardial cell death is rare and that cardiac function is fully reversible in survivors. Hence, functional rather than structural changes seem to be responsible for intrinsic myocardial depression during sepsis. The underlying mechanisms include down-regulation of beta-adrenergic receptors, depressed postreceptor signaling pathways, impaired calcium liberation from the sarcoplasmic reticulum, and impaired electromechanical coupling at the myofibrillar level. Most, if not all, of these changes are regulated by cytokines and nitric oxide. CONCLUSIONS Integrative studies are needed to distinguish the hierarchy of the various mechanisms underlying septic cardiac dysfunction. As many of these changes are related to severe inflammation and not to infection per se, a better understanding of septic myocardial dysfunction may be usefully extended to other systemic inflammatory conditions encountered in the critically ill. Myocardial depression may be arguably viewed as an adaptive event by reducing energy expenditure in a situation when energy generation is limited, thereby preventing activation of cell death pathways and allowing the potential for full functional recovery.
Collapse
Affiliation(s)
- Alain Rudiger
- Bloomsbury Institute of Intensive Care Medicine, Wolfson Institute for Biomedical Research and Department of Medicine, University College London, UK
| | | |
Collapse
|
39
|
Maeder M, Fehr T, Rickli H, Ammann P. Sepsis-associated myocardial dysfunction: diagnostic and prognostic impact of cardiac troponins and natriuretic peptides. Chest 2006; 129:1349-66. [PMID: 16685029 DOI: 10.1378/chest.129.5.1349] [Citation(s) in RCA: 202] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Myocardial dysfunction, which is characterized by transient biventricular impairment of intrinsic myocardial contractility, is a common complication in patients with sepsis. Left ventricular systolic dysfunction is reflected by a reduced left ventricular stroke work index or, less accurately, by an impaired left ventricular ejection fraction (LVEF). Early recognition of myocardial dysfunction is crucial for the administration of the most appropriate therapy. Cardiac troponins and natriuretic peptides are biomarkers that were previously introduced for diagnosis and risk stratification in patients with acute coronary syndrome and congestive heart failure, respectively. However, their prognostic and diagnostic impact in critically ill patients warrants definition. The elevation of cardiac troponin levels in patients with sepsis, severe sepsis, or septic shock has been shown to indicate left ventricular dysfunction and a poor prognosis. Troponin release in this population occurs in the absence of flow-limiting coronary artery disease, suggesting the presence of mechanisms other than thrombotic coronary artery occlusion, probably a transient loss in membrane integrity with subsequent troponin leakage or microvascular thrombotic injury. In contrast to the rather uniform results of studies dealing with cardiac troponins, the impact of raised B-type natriuretic peptide (BNP) levels in patients with sepsis is less clear. The relationship between BNP and both LVEF and left-sided filling pressures is weak, and data on the prognostic impact of high BNP levels in patients with sepsis are conflicting. Mechanisms other than left ventricular wall stress may contribute to BNP release, including right ventricular overload, catecholamine therapy, renal failure, diseases of the CNS, and cytokine up-regulation. Whereas cardiac troponins may be integrated into the monitoring of myocardial dysfunction in patients with severe sepsis or septic shock to identify those patients requiring early and aggressive supportive therapy, the routine use of BNP and other natriuretic peptides in this setting is discouraged at the moment.
Collapse
Affiliation(s)
- Micha Maeder
- Division of Cardiology, University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland.
| | | | | | | |
Collapse
|
40
|
Joshi MS, Julian MW, Huff JE, Bauer JA, Xia Y, Crouser ED. Calcineurin regulates myocardial function during acute endotoxemia. Am J Respir Crit Care Med 2006; 173:999-1007. [PMID: 16424445 PMCID: PMC2662919 DOI: 10.1164/rccm.200411-1507oc] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
RATIONALE Cyclosporin A (CsA) is known to preserve cardiac contractile function during endotoxemia, but the mechanism is unclear. Increased nitric oxide (NO) production and altered mitochondrial function are implicated as mechanisms contributing to sepsis-induced cardiac dysfunction, and CsA has the capacity to reduce NO production and inhibit mitochondrial dysfunction relating to the mitochondrial permeability transition (MPT). OBJECTIVES We hypothesized that CsA would protect against endotoxin-mediated cardiac contractile dysfunction by attenuating NO production and preserving mitochondrial function. METHODS Left ventricular function was measured continuously over 4 h in cats assigned as follows: control animals (n = 7); LPS alone (3 mg/kg, n = 8); and CsA (6 mg/kg, n = 7), a calcineurin inhibitor that blocks the MPT, or tacrolimus (FK506, 0.1 mg/kg, n = 7), a calcineurin inhibitor lacking MPT activity, followed in 30 min by LPS. Myocardial tissue was then analyzed for NO synthase-2 expression, tissue nitration, protein carbonylation, and mitochondrial morphology and function. MEASUREMENTS AND MAIN RESULTS LPS treatment resulted in impaired left ventricular contractility, altered mitochondrial morphology and function, and increased protein nitration. As hypothesized, CsA pretreatment normalized cardiac performance and mitochondrial respiration and reduced myocardial protein nitration. Unexpectedly, FK506 pretreatment had similar effects, normalizing both cardiac and mitochondrial parameters. However, CsA and FK506 pretreatments markedly increased protein carbonylation in the myocardium despite elevated manganese superoxide dismutase activity during endotoxemia. CONCLUSIONS Our data indicate that calcineurin is a critical regulator of mitochondrial respiration, tissue nitration, protein carbonylation, and contractile function in the heart during acute endotoxemia.
Collapse
Affiliation(s)
- Mandar S Joshi
- Center for Cardiovascular Medicine, Columbus Children's Research Institute, Ohio State University Medical Center, Columbus, OH 43210-1252, USA
| | | | | | | | | | | |
Collapse
|
41
|
Lang CH, Pruznak AM, Frost RA. TNFalpha mediates sepsis-induced impairment of basal and leucine-stimulated signaling via S6K1 and eIF4E in cardiac muscle. J Cell Biochem 2005; 94:419-31. [PMID: 15534870 DOI: 10.1002/jcb.20311] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Decreased translation initiation adversely impacts protein synthesis and contributes to the myocardial dysfunction produced by sepsis. Therefore, the purpose of the present study was to identify sepsis-induced changes in signal transduction pathways known to regulate translation initiation in cardiac muscle and to determine whether the stimulatory effects of leucine can reverse the observed defects. To address this aim, sepsis was produced by cecal ligation and puncture (CLP) in anesthetized rats and the animals studied in the fasted condition 24 h later. Separate groups of septic and time-matched control rats also received an oral gavage of leucine. To identify potential mechanisms responsible for regulating cap-dependent mRNA translation in cardiac muscle, several eukaryotic initiation factors (eIFs) were examined. Under basal conditions, hearts from septic rats demonstrated a redistribution of the rate-limiting factor eIF4E due to increased binding of the translational repressor 4E-BP1 with eIF4E. However, this change was independent of an alteration in the phosphorylation state of 4E-BP1. The phosphorylation of mTOR, S6K1, the ribosomal protein (rp) S6, and eIF4G was not altered in hearts from septic rats under basal conditions. In control rats, leucine failed to alter eIF4E distribution but increased the phosphorylation of S6K1 and S6. In contrast, in hearts from septic rats leucine acutely reversed the alterations in eIF4E distribution. However, the ability of leucine to increase S6K1 and rpS6 phosphorylation in septic hearts was blunted. Sepsis increased the content of tumor necrosis factor (TNF)-alpha in heart and pre-treatment of rats with a TNF antagonist prevented the above-mentioned sepsis-induced changes. These data indicate that oral administration of leucine acutely reverses sepsis-induced alterations eIF4E distribution observed under basal conditions but the anabolic actions of this amino acid on S6K1 and rpS6 phosphorylation remain blunted, providing evidence for a leucine resistance. Finally, TNFalpha, either directly or indirectly, appears to mediate the sepsis-induced defects in myocardial translation initiation.
Collapse
Affiliation(s)
- Charles H Lang
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA.
| | | | | |
Collapse
|
42
|
Lang CH, Frost RA, Vary TC. Thermal injury impairs cardiac protein synthesis and is associated with alterations in translation initiation. Am J Physiol Regul Integr Comp Physiol 2003; 286:R740-50. [PMID: 14695116 DOI: 10.1152/ajpregu.00661.2003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of the present study was to determine whether burn injury decreases myocardial protein synthesis and potential contributing mechanisms for this impairment. To address this aim, thermal injury was produced by a 40% total body surface area full-thickness scald burn in anesthetized rats, and the animals were studied 24 h late. Burn decreased the in vivo-determined rate of myocardial protein synthesis and translation efficiency by 25% but did not alter the protein synthetic rate in skeletal muscle. To identify potential mechanisms responsible for regulating mRNA translation in cardiac muscle, we examined several eukaryotic initiation factors (eIFs) and elongation factors (eEFs). Burn failed to alter eIF2B activity or the total amount or phosphorylation status of either eIF2 alpha or eIF2B epsilon in heart. In contrast, hearts from burned rats demonstrated 1) an increased binding of the translational repressor 4E-BP1 with eIF4E, 2) a decreased amount of eIF4E associated with eIF4G, and 3) a decreased amount of the hyperphosphorylated gamma-form of 4E-BP1. These changes in eIF4E availability were not seen in gastrocnemius muscle where burn injury did not decrease protein synthesis. Furthermore, constitutive phosphorylation of mTOR, S6K1, the ribosomal protein S6, and eIF4G were also decreased in hearts from burned rats. Burn did not appear to adversely affect elongation because there was no significant difference in the myocardial content of eEF1 alpha or eEF2 or the phosphorylation state of eEF2. The above-mentioned burn-induced changes in mRNA translation were associated with an impairment of in vitro myocardial performance. Finally, 24 h postburn, the cardiac mRNA content of IL-1 beta, IL-6, and high-mobility group protein B1 (but not TNF-alpha) was increased. In summary, these data suggest that thermal injury specifically decreases cardiac protein synthesis in part by decreasing mRNA translation efficiency resulting from an impairment in translation initiation associated with alterations in eIF4E availability and S6K1 activity.
Collapse
Affiliation(s)
- Charles H Lang
- Department of Cellular and Molecular Physiology H166, Penn State College of Medicine, Hershey, PA 17033, USA.
| | | | | |
Collapse
|
43
|
Abstract
The nature of myocardial dysfunction during sepsis and septic shock has been investigated for more than half a century. This review traces the evolution of scientific thought regarding this phenomenon during this period with particular emphasis on the current understanding of both the clinical manifestations and the molecular/cellular basis of septic myocardial dysfunction in critically ill patients. Current data suggest, contrary to older literature, that patients with septic shock develop a hyperdynamic circulatory state after fluid resuscitation and maintain this hyperdynamic circulatory state until death or recovery. Overt myocardial depression, as manifested by decreased cardiac output, is decidedly uncommon, even in the preterminal phase. Nonetheless, myocardial depression, as evidenced by biventricular dilation and depression of the ejection fraction, can be demonstrated in most patients with septic shock by using either radionuclide cineangiography or echocardiography. Depression is reversible over the course of 7 to 10 days in survivors. Available evidence suggests that myocardial hypoperfusion is not responsible for septic myocardial depression, because examination of humans with septic shock demonstrates increased myocardial perfusion, and animal models of septic shock appear to maintain myocardial high-energy phosphates. A circulating factor or factors, including the cytokines tumor necrosis factor alpha and interleukin-1beta, appear to have a significant role in the phenomenon. In addition, septic myocardial depression appears to be mediated in part through combinations of nitric oxide-dependent and -independent alterations of basal and catecholamine-stimulated cardiac myocyte contractility.
Collapse
Affiliation(s)
- Sreenandh Krishnagopalan
- Section of Critical Care Medicine, Rush-Presbyterian-St. Luke's Medical Center, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
44
|
Abstract
Myocardial dysfunction frequently accompanies severe sepsis and septic shock. Whereas myocardial depression was previously considered a preterminal event, it is now clear that cardiac dysfunction as evidenced by biventricular dilatation and reduced ejection fraction is present in most patients with severe sepsis and septic shock. Myocardial depression exists despite a fluid resuscitation-dependent hyperdynamic state that typically persists in septic shock patients until death or recovery. Cardiac function usually recovers within 7-10 days in survivors. Myocardial dysfunction does not appear to be due to myocardial hypoperfusion but due to circulating depressant factors, including the cytokines tumor necrosis factor alpha and IL-1beta. At a cellular level, reduced myocardial contractility seems to be induced by both nitric oxide-dependent and nitric oxide-independent mechanisms. The present paper reviews both the clinical manifestations and the molecular/cellular mechanisms of sepsis-induced myocardial depression.
Collapse
Affiliation(s)
- Olivier Court
- Fellow, Section of Critical Care Medicine, Health Sciences Center, University of Manitoba, Winnipeg, Canada
| | - Aseem Kumar
- Assistant Professor of Medicine, Section of Critical Care Medicine, Rush–Presbyterian–St Luke's Medical Center, Chicago, IL, USA
| | - Joseph E Parrillo
- Director, Division of Cardiovascular Diseases and CCM, Cooper Hospital, Camden, NJ, USA
| | - Anand Kumar
- Assistant Professor of Medicine, Section of Critical Care Medicine, Health Sciences Center, University of Manitoba, Winnipeg, Canada
| |
Collapse
|