1
|
Jozwiak M, Lim SY, Si X, Monnet X. Biomarkers in cardiogenic shock: old pals, new friends. Ann Intensive Care 2024; 14:157. [PMID: 39414666 PMCID: PMC11485002 DOI: 10.1186/s13613-024-01388-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/29/2024] [Indexed: 10/18/2024] Open
Abstract
In cardiogenic shock, biomarkers should ideally help make the diagnosis, choose the right therapeutic options and monitor the patient in addition to clinical and echocardiographic indices. Among "old" biomarkers that have been used for decades, lactate detects, quantifies, and follows anaerobic metabolism, despite its lack of specificity. Renal and liver biomarkers are indispensable for detecting the effect of shock on organ function and are highly predictive of poor outcomes. Direct biomarkers of cardiac damage such as cardiac troponins, B-type natriuretic and N-terminal pro-B-type natriuretic peptides have a good prognostic value, but they lack specificity to detect a cardiogenic cause of shock, as many factors influence their plasma concentrations in critically ill patients. Among the biomarkers that have been more recently described, dipeptidyl peptidase-3 is one of the most interesting. In addition to its prognostic value, it could represent a therapeutic target in cardiogenic shock in the future as a specific antibody inhibits its activity. Adrenomedullin is a small peptide hormone secreted by various tissues, including vascular smooth muscle cells and endothelium, particularly under pathological conditions. It has a vasodilator effect and has prognostic value during cardiogenic shock. An antibody inhibits its activity and so adrenomedullin could represent a therapeutic target in cardiogenic shock. An increasing number of inflammatory biomarkers are also of proven prognostic value in cardiogenic shock, reflecting the inflammatory reaction associated with the syndrome. Some of them are combined to form prognostic proteomic scores. Alongside clinical variables, biomarkers can be used to establish biological "signatures" characteristic of the pathophysiological pathways involved in cardiogenic shock. This helps describe patient subphenotypes, which could in the future be used in clinical trials to define patient populations responding specifically to a treatment.
Collapse
Affiliation(s)
- Mathieu Jozwiak
- Service de Médecine Intensive Réanimation, CHU de Nice, Hôpital L'Archet 1, 151 Route Saint Antoine de Ginestière, 06200, Nice, France.
- UR2CA, Unité de Recherche Clinique Côte d'Azur, Université Côte d'Azur, 06200, Nice, France.
| | - Sung Yoon Lim
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU 4 CORREVE, Inserm UMR S_999, FHU SEPSIS, CARMAS, Université Paris-Saclay, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicêtre, France
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Xiang Si
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU 4 CORREVE, Inserm UMR S_999, FHU SEPSIS, CARMAS, Université Paris-Saclay, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicêtre, France
- Department of Critical Care Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xavier Monnet
- AP-HP, Service de Médecine Intensive-Réanimation, Hôpital de Bicêtre, DMU 4 CORREVE, Inserm UMR S_999, FHU SEPSIS, CARMAS, Université Paris-Saclay, 78 Rue du Général Leclerc, 94270, Le Kremlin-Bicêtre, France
| |
Collapse
|
2
|
Zhang J, Huang Z, Lin Q, Hu W, Zhong H, Zhang F, Huang J. The correlation between serum angiopoietin-2 levels and acute kidney injury (AKI): a meta-analysis. Clin Chem Lab Med 2024; 0:cclm-2024-0365. [PMID: 38915251 DOI: 10.1515/cclm-2024-0365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION The correlation between serum angiopoietin-2 levels and acute kidney injury (AKI) is a topic of significant clinical interest. This meta-analysis aims to provide a comprehensive evaluation of this relationship. CONTENT A systematic search was conducted in PubMed, Embase, Web of Science, and Cochrane databases up to October 11, 2023. The included studies were evaluated using the Newcastle-Ottawa Scale (NOS) and Methodological Index for Non-Randomized Studies (MINORS). Weighted mean differences (WMD) and odds ratios (OR) were calculated using random-effects models. Sensitivity analysis, funnel plots, and Egger's test were used to assess the robustness and publication bias of the findings. Subgroup analyses were performed to explore potential variations between adults and children. SUMMARY Eighteen studies encompassing a total of 7,453 participants were included. The analysis revealed a significant elevation in serum angiopoietin-2 levels in patients with AKI compared to those without (WMD: 4.85; 95 % CI: 0.75 to 0.27; I²=93.2 %, p<0.001). Subgroup analysis indicated significantly higher angiopoietin-2 levels in adults with AKI (WMD: 5.17; 95 % CI: 3.51 to 6.83; I²=82.6 %, p<0.001), but not in children. Additionally, high serum angiopoietin-2 levels were associated with an increased risk of AKI (OR: 1.58; 95 % CI: 1.39 to 1.8; I²=89.1 %, p<0.001). Sensitivity analysis validated the robustness of these results, showing no substantial change in the overall effect size upon the exclusion of individual studies. OUTLOOK This meta-analysis supports a significant association between elevated serum angiopoietin-2 levels and increased risk of AKI. The observed differential association between adults and children highlights the need for further targeted research to understand these age-specific variations.
Collapse
Affiliation(s)
- Juncheng Zhang
- The School of Clinical Medicine, 74551 Fujian Medical University , Fuzhou, Fujian, China
| | - Zhengjie Huang
- The School of Clinical Medicine, 74551 Fujian Medical University , Fuzhou, Fujian, China
| | - Qin Lin
- The School of Clinical Medicine, 74551 Fujian Medical University , Fuzhou, Fujian, China
- Department of Nephrology, Xiamen Key Laboratory of Precision Diagnosis and Treatment of Chronic Kidney Disease, The Fifth Hospital of Xiamen, Xiamen, Fujian, China
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Weiping Hu
- The School of Clinical Medicine, 74551 Fujian Medical University , Fuzhou, Fujian, China
- Department of Nephrology, Xiamen Key Laboratory of Precision Diagnosis and Treatment of Chronic Kidney Disease, The Fifth Hospital of Xiamen, Xiamen, Fujian, China
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hongbin Zhong
- The School of Clinical Medicine, 74551 Fujian Medical University , Fuzhou, Fujian, China
- Department of Nephrology, Xiamen Key Laboratory of Precision Diagnosis and Treatment of Chronic Kidney Disease, The Fifth Hospital of Xiamen, Xiamen, Fujian, China
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Fengling Zhang
- The School of Clinical Medicine, 74551 Fujian Medical University , Fuzhou, Fujian, China
- Department of Nephrology, Xiamen Key Laboratory of Precision Diagnosis and Treatment of Chronic Kidney Disease, The Fifth Hospital of Xiamen, Xiamen, Fujian, China
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jiyi Huang
- The School of Clinical Medicine, 74551 Fujian Medical University , Fuzhou, Fujian, China
- Department of Nephrology, Xiamen Key Laboratory of Precision Diagnosis and Treatment of Chronic Kidney Disease, The Fifth Hospital of Xiamen, Xiamen, Fujian, China
- Department of Nephrology, Fujian Clinical Research Center for Chronic Glomerular Disease, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
3
|
Kim HD, Kim EN, Lim JH, Kim Y, Ban TH, Lee H, Kim YS, Park CW, Choi BS. Phosphodiesterase inhibitor ameliorates senescent changes of renal interstitial pericytes in aging kidney. Aging Cell 2024; 23:e14075. [PMID: 38155524 PMCID: PMC10928568 DOI: 10.1111/acel.14075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023] Open
Abstract
Pericytes are mesenchymal cells that surround endothelial cells, playing a crucial role in angiogenesis and vessel maturation. Additionally, they are associated with interstitial fibrosis as a major contributor to renal myofibroblasts. In this study, we aim to investigate whether the phosphodiesterase inhibitor, pentoxifylline (PTX), can ameliorate aging-related functional and histological deterioration in the kidney. We subjected aging C57BL/6 mice, dividing into young, aging, and PTX-treated aging groups. Renal function, albuminuria, and histological changes were assessed. Interstitial pericytes were assessed by immunohistochemistry analysis. We examined changes in pericytes in elderly patients using human kidney tissue obtained from healthy kidney donors for kidney transplantation. In vitro experiments with human pericytes and endothelial cells were performed. Aging mice exhibited declined renal function, increased albuminuria, and aging-related histological changes including mesangial expansion and tubulointerstitial fibrosis. Notably, number of pericytes declined in aging kidneys, and myofibroblasts increased. PTX treatment ameliorated albuminuria, histological alterations, and microvascular rarefaction, as well as modulated angiopoietin expression. In vitro experiments showed PTX reduced cellular senescence and inflammation. Human kidney analysis confirmed similar pericyte changes in aging kidneys. The phosphodiesterase inhibitor, PTX preserved microvascular density and improved renal interstitial fibrosis and inflammation in aging mice kidneys. These protective effects were suggested to be associated with the amelioration of pericytes reduction and the transition to myofibroblasts. Additionally, the upregulation of angiopoietin-1 expression may exert potential impacts. To the best of our knowledge, this is the first report on the changes in renal interstitial pericytes in aging human kidneys.
Collapse
Affiliation(s)
- Hyung Duk Kim
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Eun Nim Kim
- Department of Internal Medicine, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Ji Hee Lim
- Department of Internal Medicine, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Yaeni Kim
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Tae Hyun Ban
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Hajeong Lee
- Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
| | - Yon Su Kim
- Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
| | - Cheol Whee Park
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Bum Soon Choi
- Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| |
Collapse
|
4
|
Morici N, Frigerio G, Campolo J, Fustinoni S, Sacco A, Garatti L, Villanova L, Tavazzi G, Kapur NK, Pappalardo F. Cardiogenic Shock Integrated PHenotyping for Event Reduction: A Pilot Metabolomics Analysis. Int J Mol Sci 2023; 24:17607. [PMID: 38139435 PMCID: PMC10743901 DOI: 10.3390/ijms242417607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/10/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Cardiogenic shock (CS) portends a dismal prognosis if hypoperfusion triggers uncontrolled inflammatory and metabolic derangements. We sought to investigate metabolomic profiles and temporal changes in IL6, Ang-2, and markers of glycocalyx perturbation from admission to discharge in eighteen patients with heart failure complicated by CS (HF-CS). Biological samples were collected from 18 consecutive HF-CS patients at admission (T0), 48 h after admission (T1), and at discharge (T2). ELISA analytical techniques and targeted metabolomics were performed Seven patients (44%) died at in-hospital follow-up. Among the survivors, IL-6 and kynurenine were significantly reduced at discharge compared to baseline. Conversely, the amino acids arginine, threonine, glycine, lysine, and asparagine; the biogenic amine putrescine; multiple sphingolipids; and glycerophospholipids were significantly increased. Patients with HF-CS have a metabolomic fingerprint that might allow for tailored treatment strategies for the patients' recovery or stabilization.
Collapse
Affiliation(s)
- Nuccia Morici
- Dipartimento Cardio-Respiratorio, IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20100 Milan, Italy
| | - Gianfranco Frigerio
- Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20126 Milan, Italy; (G.F.); (S.F.)
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4365 Luxembourg, Luxembourg
| | - Jonica Campolo
- Institute of Clinical Physiology CNR, 20162 Milan, Italy
| | - Silvia Fustinoni
- Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20126 Milan, Italy; (G.F.); (S.F.)
| | - Alice Sacco
- Intensive Cardiac Care Unit and De Gasperis Cardio Center, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Laura Garatti
- Intensive Cardiac Care Unit and De Gasperis Cardio Center, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Luca Villanova
- Intensive Cardiac Care Unit and De Gasperis Cardio Center, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Guido Tavazzi
- Unit of Anaesthesia and Intensive Care, Department of Clinical-Surgical, Diagnostic and Paediatric Sciences, University of Pavia, 27100 Pavia, Italy
- Anesthesia and Intensive Care, Fondazione Policlinico San Matteo Hospital IRCCS, Anestesia e Rianimazione I, 27100 Pavia, Italy
| | - Navin K. Kapur
- The CardioVascular Center, Tufts Medical Center, Boston, MA 02111, USA;
| | - Federico Pappalardo
- Cardiothoracic and Vascular Anesthesia and Intensive Care, AO SS. Antonio e Biagio e Cesare Arrigo, 15100 Alessandria, Italy;
| |
Collapse
|
5
|
Buckel M, Maclean P, Knight JC, Lawler PR, Proudfoot AG. Extending the 'host response' paradigm from sepsis to cardiogenic shock: evidence, limitations and opportunities. Crit Care 2023; 27:460. [PMID: 38012789 PMCID: PMC10683227 DOI: 10.1186/s13054-023-04752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
Recent clinical and research efforts in cardiogenic shock (CS) have largely focussed on the restoration of the low cardiac output state that is the conditio sine qua non of the clinical syndrome. This approach has failed to translate into improved outcomes, and mortality has remained static at 30-50%. There is an unmet need to better delineate the pathobiology of CS to understand the observed heterogeneity of presentation and treatment effect and to identify novel therapeutic targets. Despite data in other critical illness syndromes, specifically sepsis, the role of dysregulated inflammation and immunity is hitherto poorly described in CS. High-dimensional molecular profiling, particularly through leukocyte transcriptomics, may afford opportunity to better characterise subgroups of patients with shared mechanisms of immune dysregulation. In this state-of-the-art review, we outline the rationale for considering molecular subtypes of CS. We describe how high-dimensional molecular technologies can be used to identify these subtypes, and whether they share biological features with sepsis and other critical illness states. Finally, we propose how the identification of molecular subtypes of patients may enrich future clinical trial design and identification of novel therapies for CS.
Collapse
Affiliation(s)
- Marie Buckel
- Department of Perioperative Medicine, Bart's Heart Centre, St. Bartholomew's Hospital, London, UK
| | - Patrick Maclean
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, ON, Canada
- McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Alastair G Proudfoot
- Department of Perioperative Medicine, Bart's Heart Centre, St. Bartholomew's Hospital, London, UK.
- Queen Mary University of London, London, UK.
| |
Collapse
|
6
|
Vahldieck C, Fels B, Löning S, Nickel L, Weil J, Kusche-Vihrog K. Prolonged Door-to-Balloon Time Leads to Endothelial Glycocalyx Damage and Endothelial Dysfunction in Patients with ST-Elevation Myocardial Infarction. Biomedicines 2023; 11:2924. [PMID: 38001925 PMCID: PMC10669223 DOI: 10.3390/biomedicines11112924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Damage to the endothelial glycocalyx (eGC) has been reported during acute ischemic events like ST-elevation myocardial infarction (STEMI). In STEMI, a door-to-balloon time (D2B) of <60 min was shown to reduce mortality and nonfatal complications. Here, we hypothesize that eGC condition is associated with D2B duration and endothelial function during STEMI. One hundred and twenty-six individuals were analyzed in this study (STEMI patients vs. age-/sex-matched healthy volunteers). After stimulating endothelial cells with patient/control sera, the eGC's nanomechanical properties (i.e., height/stiffness) were analyzed using the atomic force microscopy-based nanoindentation technique. eGC components were determined via ELISA, and measurements of nitric oxide levels (NO) were based on chemiluminescence. eGC height/stiffness (both p < 0.001), as well as NO concentration (p < 0.001), were reduced during STEMI. Notably, the D2B had a strong impact on the endothelial condition: a D2B > 60 min led to significantly higher serum concentrations of eGC components (syndecan-1: p < 0.001/heparan sulfate: p < 0.001/hyaluronic acid: p < 0.0001). A D2B > 60 min led to the pronounced loss of eGC height/stiffness (both, p < 0.001) with reduced NO concentrations (p < 0.01), activated the complement system (p < 0.001), and prolonged the hospital stay (p < 0.01). An increased D2B led to severe eGC shedding, with endothelial dysfunction in a temporal context. eGC components and pro-inflammatory mediators correlated with a prolonged D2B, indicating a time-dependent immune reaction during STEMI, with a decreased NO concentration. Thus, D2B is a crucial factor for eGC damage during STEMI. Clinical evaluation of the eGC condition might serve as an important predictor for the endothelial function of STEMI patients in the future.
Collapse
Affiliation(s)
- Carl Vahldieck
- Department of Anesthesiology and Intensive Care Medicine, University Medical Centre Schleswig-Holstein Campus Luebeck, 23538 Luebeck, Germany
- Institute of Physiology, University of Luebeck, 23562 Luebeck, Germany; (B.F.); (K.K.-V.)
| | - Benedikt Fels
- Institute of Physiology, University of Luebeck, 23562 Luebeck, Germany; (B.F.); (K.K.-V.)
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, 23562 Luebeck, Germany
| | - Samuel Löning
- Institute of Physiology, University of Luebeck, 23562 Luebeck, Germany; (B.F.); (K.K.-V.)
| | - Laura Nickel
- Medizinische Klinik II, Sana Kliniken Luebeck, 23560 Luebeck, Germany (J.W.)
| | - Joachim Weil
- Medizinische Klinik II, Sana Kliniken Luebeck, 23560 Luebeck, Germany (J.W.)
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Luebeck, 23562 Luebeck, Germany; (B.F.); (K.K.-V.)
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Luebeck/Kiel, 23562 Luebeck, Germany
| |
Collapse
|
7
|
Nakamura M, Imamura T, Ueno H, Kinugawa K. Impact of the elevated angiopoietin-2 levels during Impella support on the short-term prognosis. J Artif Organs 2023; 26:184-191. [PMID: 35932355 DOI: 10.1007/s10047-022-01347-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 10/15/2022]
Abstract
Elevated serum angiopoietin-2 levels in patients with acute myocardial infarction-related cardiogenic shock with and without intra-aortic balloon pump as well as acute decompensated heart failure are associated with short-term mortality. However, its prognostic impact in patients with cardiogenic shock supported by Impella-incorporated mechanical circulatory support (MCS) remains unknown. Patients who received temporary MCS (Impella alone or Impella and veno-arterial extracorporeal membrane oxygenation) in our institute between August 2018 and January 2022 were included in this prospective study. The serum levels of angiopoietin-2 were measured just before and following the initiation of temporary MCS therapy. Association between the levels of serum angiopoietin-2 and 30-day mortality was investigated. A total of 38 patients (median 72 years old, 63% men) were included. The median levels of serum angiopoetin-2 tended to decrease from baseline to 4 days following the initiation of temporary MCS from 5.2 (3.3, 10.5) ng/mL to 4.8 (2.7, 6.8) ng/mL (p = 0.132). A higher angiopoietin-2 (> 6.8 ng/mL) following the initiation of temporary MCS was associated with higher 30-day mortality (89.7% versus 44.4%, p = 0.0048) with an odds ratio 18.946 (95% confidence interval 1.624-218.695, p = 0.018) adjusted for potential confounders. A higher serum angiopoietin-2 level following the initiation of Impella-incorporated temporary MCS, instead of baseline angiopoetin-2 level, was associated with higher short-term mortality.
Collapse
Affiliation(s)
- Makiko Nakamura
- Second Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Teruhiko Imamura
- Second Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.
| | - Hiroshi Ueno
- Second Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Koichiro Kinugawa
- Second Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| |
Collapse
|
8
|
Soussi S, Dos Santos C, Jentzer JC, Mebazaa A, Gayat E, Pöss J, Schaubroeck H, Billia F, Marshall JC, Lawler PR. Distinct host-response signatures in circulatory shock: a narrative review. Intensive Care Med Exp 2023; 11:50. [PMID: 37592121 PMCID: PMC10435428 DOI: 10.1186/s40635-023-00531-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 07/01/2023] [Indexed: 08/19/2023] Open
Abstract
Circulatory shock is defined syndromically as hypotension associated with tissue hypoperfusion and often subcategorized according to hemodynamic profile (e.g., distributive, cardiogenic, hypovolemic) and etiology (e.g., infection, myocardial infarction, trauma, among others). These shock subgroups are generally considered homogeneous entities in research and clinical practice. This current definition fails to consider the complex pathophysiology of shock and the influence of patient heterogeneity. Recent translational evidence highlights previously under-appreciated heterogeneity regarding the underlying pathways with distinct host-response patterns in circulatory shock syndromes. This heterogeneity may confound the interpretation of trial results as a given treatment may preferentially impact distinct subgroups. Re-analyzing results of major 'neutral' treatment trials from the perspective of biological mechanisms (i.e., host-response signatures) may reveal treatment effects in subgroups of patients that share treatable traits (i.e., specific biological signatures that portend a predictable response to a given treatment). In this review, we discuss the emerging literature suggesting the existence of distinct biomarker-based host-response patterns of circulatory shock syndrome independent of etiology or hemodynamic profile. We further review responses to newly prescribed treatments in the intensive care unit designed to personalize treatments (biomarker-driven or endotype-driven patient selection in support of future clinical trials).
Collapse
Affiliation(s)
- Sabri Soussi
- Department of Anesthesia and Pain Management, University Health Network (UHN), Women's College Hospital, University of Toronto, Toronto Western Hospital, 399 Bathurst St, ON, M5T 2S8, Toronto, Canada.
- St Michael's Hospital, Keenan Research Centre for Biomedical Science and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.
| | - Claudia Dos Santos
- St Michael's Hospital, Keenan Research Centre for Biomedical Science and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Jacob C Jentzer
- Department of Cardiovascular Medicine, Mayo Clinic Rochester, Rochester, MN, 55905, USA
| | - Alexandre Mebazaa
- Department of Anesthesiology, Critical Care, Lariboisière-Saint-Louis Hospitals, DMU Parabol, AP-HP Nord; Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, Paris, France
| | - Etienne Gayat
- Department of Anesthesiology, Critical Care, Lariboisière-Saint-Louis Hospitals, DMU Parabol, AP-HP Nord; Inserm UMR-S 942, Cardiovascular Markers in Stress Conditions (MASCOT), University of Paris, Paris, France
| | - Janine Pöss
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at the University of Leipzig, Strümpellstraße, 39 04289, Leipzig, Germany
| | - Hannah Schaubroeck
- Department of Intensive Care Medicine, Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Filio Billia
- Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, ON, Canada
- Ted Roger's Center for Heart Research, University Health Network, University of Toronto, Toronto, ON, Canada
| | - John C Marshall
- St Michael's Hospital, Keenan Research Centre for Biomedical Science and Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, ON, Canada
- McGill University Health Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
9
|
Polyzogopoulou E, Bezati S, Karamasis G, Boultadakis A, Parissis J. Early Recognition and Risk Stratification in Cardiogenic Shock: Well Begun Is Half Done. J Clin Med 2023; 12:2643. [PMID: 37048727 PMCID: PMC10095596 DOI: 10.3390/jcm12072643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Cardiogenic shock is a complex syndrome manifesting with distinct phenotypes depending on the severity of the primary cardiac insult and the underlying status. As long as therapeutic interventions fail to divert its unopposed rapid evolution, poor outcomes will continue challenging health care systems. Thus, early recognition in the emergency setting is a priority, in order to avoid delays in appropriate management and to ensure immediate initial stabilization. Since advanced therapeutic strategies and specialized shock centers may provide beneficial support, it seems that directing patients towards the recently described shock network may improve survival rates. A multidisciplinary approach strategy commands the interconnections between the strategic role of the ED in affiliation with cardiac shock centers. This review outlines critical features of early recognition and initial therapeutic management, as well as the utility of diagnostic tools and risk stratification models regarding the facilitation of patient trajectories through the shock network. Further, it proposes the implementation of precise criteria for shock team activation and the establishment of definite exclusion criteria for streaming the right patient to the right place at the right time.
Collapse
Affiliation(s)
- Effie Polyzogopoulou
- Emergency Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece
| | - Sofia Bezati
- Emergency Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece
| | - Grigoris Karamasis
- Second Department of Cardiology, Medical School, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Antonios Boultadakis
- Emergency Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece
| | - John Parissis
- Emergency Department, Attikon University Hospital, National and Kapodistrian University of Athens, Rimini 1, Chaidari, 12462 Athens, Greece
| |
Collapse
|
10
|
Circulating Galectin-3 in Patients with Cardiogenic Shock Complicating Acute Myocardial Infarction Treated with Mild Hypothermia: A Biomarker Sub-Study of the SHOCK-COOL Trial. J Clin Med 2022; 11:jcm11237168. [PMID: 36498742 PMCID: PMC9740246 DOI: 10.3390/jcm11237168] [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: 11/06/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
Background: Galectin-3 (Gal-3) is considered a potential cardiovascular inflammatory marker that may provide additional risk stratification for patients with acute heart failure. It is unknown whether mild therapeutic hypothermia (MTH) impacts Gal-3 levels. Therefore, this biomarker study aimed to investigate the effect of MTH on Gal-3. Methods: In the randomized SHOCK-COOL trial, 40 patients with cardiogenic shock (CS) complicating acute myocardial infraction (AMI) were randomly assigned to the MTH (33 °C) or control group in a 1:1 ratio. Blood samples were collected on the day of admission/day 1, day 2, and day 3. Gal-3 level kinetics throughout these time points were compared between the MTH and control groups. Additionally, potential correlations between Gal-3 and clinical patient characteristics were assessed. Multiple imputations were performed to account for missing data. Results: In the control group, Gal-3 levels were significantly lower on day 3 than on day 1 (day 1 vs. day 3: 3.84 [IQR 2.04−13.3] vs. 1.79 [IQR 1.23−3.50] ng/mL; p = 0.049). Gal-3 levels were not significantly different on any day between the MTH and control groups (p for interaction = 0.242). Spearman’s rank correlation test showed no significant correlation between Gal-3 levels and sex, age, smoking, body mass index (BMI), and levels of creatine kinase-MB, creatine kinase, C-reactive protein, creatinine, and white blood cell counts (all p > 0.05). Patients with lower Gal-3 levels on the first day after admission demonstrated a higher risk of all-cause mortality at 30 days (hazard ratio, 2.67; 95% CI, 1.11−6.42; p = 0.029). In addition, Gal-3 levels on day 1 had a good predictive value for 30-day all-cause mortality with an area under the receiver operating characteristic curve of 0.696 (95% CI: 0.513−0.879), with an optimal cut-off point of less than 3651 pg/mL. Conclusions: MTH has no effect on Gal-3 levels in patients with CS complicating AMI compared to the control group. In addition, Gal-3 is a relatively stable biomarker, independent of age, sex, and BMI, and Gal-3 levels at admission might predict the risk of 30-day all-cause mortality.
Collapse
|
11
|
Collazos-Alemán JD, Gnecco-González S, Jaramillo-Zarama B, Jiménez-Mora MA, Mendivil CO. The Role of Angiopoietins in Neovascular Diabetes-Related Retinal Diseases. Diabetes Ther 2022; 13:1811-1821. [PMID: 36331711 PMCID: PMC9663771 DOI: 10.1007/s13300-022-01326-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Diabetic retinopathy is a devastating and frequent complication of poorly controlled diabetes, whose pathogenesis is still only partially understood. Advances in basic research over the last two decades have led to the discovery of angiopoietins, proteins that strongly influence the growth and integrity of blood vessels in many vascular beds, with particular importance in the retina. Angiopoietin 1 (Ang1), produced mostly by pericytes and platelets, and angiopoietin 2 (Ang2), produced mainly by endothelial cells, bind to the same receptor (Tie2), but exert opposing effects on target cells. Ang1 maintains the stability of the mature vasculature, while Ang2 promotes vessel wall destabilization and disruption of the connections between endothelial cells and pericytes. Human retinal endothelial cells exposed to Ang2 show reduced membrane expression of the adhesion molecule VE-cadherin, and patients with proliferative diabetic retinopathy or diabetic macular edema have markedly increased vitreal concentrations of Ang2. Faricimab, a bi-specific antibody simultaneously directed against Ang2 and VEGF, has shown promising results in clinical trials among patients with diabetic retinopathy, and other agents targeting the angiopoietin system are currently in development.
Collapse
Affiliation(s)
| | - Sofía Gnecco-González
- School of Medicine, Universidad de los Andes, Carrera 7 No 116-05, Of 413, Bogotá, Colombia
| | | | - Mario A Jiménez-Mora
- Department of Ophthalmology, Faculty of Medicine, National University of Colombia, Bogotá, Colombia
| | - Carlos O Mendivil
- School of Medicine, Universidad de los Andes, Carrera 7 No 116-05, Of 413, Bogotá, Colombia.
- Section of Endocrinology, Fundación Santa Fe de Bogotá, Bogotá, Colombia.
| |
Collapse
|
12
|
Varricchi G, Poto R, Ferrara AL, Gambino G, Marone G, Rengo G, Loffredo S, Bencivenga L. Angiopoietins, vascular endothelial growth factors and secretory phospholipase A 2 in heart failure patients with preserved ejection fraction. Eur J Intern Med 2022; 106:111-119. [PMID: 36280524 DOI: 10.1016/j.ejim.2022.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Heart failure (HF) is a growing public health burden, with high prevalence and mortality rates. A proportion of patients with HF have a normal ventricular ejection fraction (EF), referred to as HF with preserved EF (HFpEF), as opposed to patients with HF with reduced ejection fraction (HFrEF). HFpEF currently accounts for about 50% of all HF patients, and its prevalence is rising. Angiopoietins (ANGPTs), vascular endothelial growth factors (VEGFs) and secretory phospholipases A2 (sPLA2s) are proinflammatory mediators and key regulators of endothelial cells. METHODS The aim of this study was to analyze the plasma concentrations of angiogenic (ANGPT1, ANGPT2, VEGF-A) and lymphangiogenic (VEGF-C, VEGF-D) factors and the plasma activity of sPLA2 in patients with HFpEF and HFrEF compared to healthy controls. RESULTS The concentration of ANGPT1 was reduced in HFrEF compared to HFpEF patients and healthy controls. ANGPT2 levels were increased in both HFrEF and HFpEF subjects compared to controls. The ANGPT2/ANGPT1 ratio was increased in HFrEF patients compared to controls. The concentrations of both VEGF-A and VEGF-C did not differ among the three groups examined. VEGF-D was increased in both HFrEF and HFpEF patients compared to controls. Plasma activity of sPLA2 was increased in HFrEF but not in HFpEF patients compared to controls. CONCLUSIONS Our results indicate that three different classes of proinflammatory regulators of vascular permeability and smoldering inflammation are selectively altered in HFrEF or HFpEF patients. Studies involving larger cohorts of these patients will be necessary to demonstrate the clinical implications of our findings.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuseppina Gambino
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Istituti Clinici Scientifici Maugeri SpA Società Benefit, 82037, Telese, (BN), Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy; World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy; Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Leonardo Bencivenga
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80131, Naples, Italy; Gèrontopole de Toulouse, Institut du Vieillissement, CHU de Toulouse, 31000, Toulouse, France
| |
Collapse
|
13
|
Szekely Y, Luk A, Lawler PR. Emerging biomarkers for risk stratification in cardiogenic shock: steps closer to precision? EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:739-741. [PMID: 36166348 DOI: 10.1093/ehjacc/zuac112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Affiliation(s)
- Yishay Szekely
- Peter Munk Cardiac Centre and Ted Rogers Centre for Heart Research, University Health Network, 585 University Ave, Toronto, ON M5G 2N2, Canada
- Division of Cardiology, Department of Medicine, University of Toronto, 6 Queen's Park Crescent West, Toronto, ON, M5S 3H2, Canada
- Tel Aviv Sourasky Medical Center and Sackler School of Medicine, Tel Aviv University, 6 Weizmann St, Tel Aviv, 6423906, Israel
| | - Adriana Luk
- Peter Munk Cardiac Centre and Ted Rogers Centre for Heart Research, University Health Network, 585 University Ave, Toronto, ON M5G 2N2, Canada
- Division of Cardiology, Department of Medicine, University of Toronto, 6 Queen's Park Crescent West, Toronto, ON, M5S 3H2, Canada
| | - Patrick R Lawler
- Peter Munk Cardiac Centre and Ted Rogers Centre for Heart Research, University Health Network, 585 University Ave, Toronto, ON M5G 2N2, Canada
- Division of Cardiology, Department of Medicine, University of Toronto, 6 Queen's Park Crescent West, Toronto, ON, M5S 3H2, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, 6 Queen's Park Crescent West, Toronto, ON, M5S 3H2, Canada
| |
Collapse
|
14
|
Krychtiuk KA, Vrints C, Wojta J, Huber K, Speidl WS. Basic mechanisms in cardiogenic shock: part 2 - biomarkers and treatment options. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:366-374. [PMID: 35218355 DOI: 10.1093/ehjacc/zuac022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/02/2022] [Accepted: 02/11/2022] [Indexed: 06/14/2023]
Abstract
Cardiogenic shock mortality rates remain high despite significant advances in cardiovascular medicine and the widespread uptake of mechanical circulatory support systems. Except for early invasive angiography and percutaneous coronary intervention of the infarct-related artery, all other widely used therapeutic measures are based on low-quality evidence. The grim prognosis and lack of high-quality data warrant further action. Within Part 2 of this two-part educational review on basic mechanisms in cardiogenic shock, we aimed to highlight the current status of translating our understanding of the pathophysiology of cardiogenic shock into clinical practice. We summarize the current status of biomarker research in risk stratification and therapy guidance. In addition, we summarized the current status of translating the findings from bench-, bedside, and biomarker studies into treatment options. Several large randomized controlled trials (RCTs) are underway, providing a huge opportunity to study contemporary cardiogenic shock patients. Finally, we call for translational, homogenous, biomarker-based, international RCTs testing novel treatment approaches to improve the outcome of our patients.
Collapse
Affiliation(s)
- Konstantin A Krychtiuk
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Duke Clinical Research Institute, 300 W Morgan Street, 27701 Durham, NC, USA
| | - Christiaan Vrints
- Research Group Cardiovascular Diseases, Department GENCOR, University of Antwerp, Prinsstraat 13, 2000 Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, 2650 Edegem, Belgium
| | - Johann Wojta
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Core Facilities, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Kurt Huber
- Ludwig Boltzmann Institute for Cardiovascular Research, Waehringer Guertel 18-20, 1090 Vienna, Austria
- 3rd Department of Internal Medicine, Cardiology and Intensive Care Unit, Wilhelminenhospital, Montleartstraße 37, 1160 Vienna, Austria
- Medical School, Sigmund Freud University, Freudplatz 1, 1020 Vienna, Austria
| | - Walter S Speidl
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
15
|
Muzafarova T, Motovska Z. Laboratory Predictors of Prognosis in Cardiogenic Shock Complicating Acute Myocardial Infarction. Biomedicines 2022; 10:1328. [PMID: 35740350 PMCID: PMC9220203 DOI: 10.3390/biomedicines10061328] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
Cardiogenic shock is a state of reduced cardiac output leading to hypotension, pulmonary congestion, and hypoperfusion of tissues and vital organs. Despite the advances in intensive care over the last years, the morbidity and mortality of patients remain high. The available studies of patients with cardiogenic shock suggest a connection between clinical variables, the level of biomarkers, the results of imaging investigations, strategies of management and the outcome of this group of patients. The management of patients with cardiogenic shock initially complicating acute myocardial infarction is challenging, and the number of studies in this area is growing fast. The purpose of this review is to summarize the currently available evidence on cardiogenic shock initially complicating acute myocardial infarction with particular attention to predictors of prognosis, focusing on laboratory variables (established and new), and to discuss the practical implementation. Currently available scoring systems developed during the past few decades predict the clinical outcome of this group of patients using some of the established biomarkers among other variables. With the new laboratory biomarkers that have shown their predictive value in cardiogenic shock outcomes, a new design of scoring systems would be of interest. Identifying high-risk patients offers the opportunity for early decision-making.
Collapse
Affiliation(s)
| | - Zuzana Motovska
- Cardiocenter, Third Faculty of Medicine Charles University and University Hospital Kralovske Vinohrady, 10034 Prague, Czech Republic;
| |
Collapse
|
16
|
Mathew R, Fernando SM, Hu K, Parlow S, Di Santo P, Brodie D, Hibbert B. Optimal Perfusion Targets in Cardiogenic Shock. JACC. ADVANCES 2022; 1:100034. [PMID: 38939320 PMCID: PMC11198174 DOI: 10.1016/j.jacadv.2022.100034] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 06/29/2024]
Abstract
Cardiology shock is a syndrome of low cardiac output resulting in end-organ dysfunction. Few interventions have demonstrated meaningful clinical benefit, and cardiogenic shock continues to carry significant morbidity with mortality rates that have plateaued at upwards of 40% over the past decade. Clinicians must rely on clinical, biochemical, and hemodynamic parameters to guide resuscitation. Several features, including physical examination, renal function, serum lactate metabolism, venous oxygen saturation, and hemodynamic markers of right ventricular function, may be useful both as prognostic markers and to guide therapy. This article aims to review these targets, their utility in the care of patients with cardiology shock, and their association with outcomes.
Collapse
Affiliation(s)
- Rebecca Mathew
- Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Shannon M. Fernando
- Division of Critical Care, Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Kira Hu
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Simon Parlow
- Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Pietro Di Santo
- Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Daniel Brodie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
- Center for Acute Respiratory Failure, New York-Presbyterian Hospital, New York, New York, USA
| | - Benjamin Hibbert
- Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada
- CAPITAL Research Group, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
17
|
Araki T, Okumura T, Hiraiwa H, Mizutani T, Kimura Y, Kazama S, Shibata N, Oishi H, Kuwayama T, Kondo T, Morimoto R, Takefuji M, Murohara T. Serum autotaxin as a novel prognostic marker in patients with non-ischaemic dilated cardiomyopathy. ESC Heart Fail 2022; 9:1304-1313. [PMID: 35112500 PMCID: PMC8934986 DOI: 10.1002/ehf2.13817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/26/2021] [Accepted: 01/11/2022] [Indexed: 11/22/2022] Open
Abstract
Aims Autotaxin (ATX) promotes myocardial inflammation, fibrosis, and the subsequent cardiac remodelling through lysophosphatidic acid production. However, the prognostic impact of serum ATX in non‐ischaemic dilated cardiomyopathy (NIDCM) has not been clarified. We investigated the prognostic impact of serum ATX in patients with NIDCM. Methods and results We enrolled 104 patients with NIDCM (49.8 ± 13.4 years, 76 men). We divided the patients into two groups using different cutoffs of median serum ATX levels for men and women: high‐ATX group and low‐ATX group. Cardiac events were defined as a composite of cardiac death and heart failure resulting in hospitalization. Median ATX level was 203.5 ng/mL for men and 257.0 ng/mL for women. Brain natriuretic peptide levels [224.0 (59.6–689.5) pg/mL vs. 96.5 (40.8–191.5) pg/mL, P = 0.010] were higher in the high‐ATX group than low‐ATX group, whereas high‐sensitivity C‐reactive protein and collagen volume fraction levels in endomyocardial biopsy samples were not significantly different between the two groups. Kaplan–Meier survival analysis revealed that the event‐free survival rate was significantly lower in the high‐ATX group than low‐ATX group (log‐rank; P = 0.007). Cox proportional hazard analysis revealed that high‐ATX was an independent determinant of composite cardiac events. In both sexes, serum ATX levels did not correlate with high‐sensitivity C‐reactive protein levels and collagen volume fraction but had a weak correlation with brain natriuretic peptide levels (men; spearman's rank: 0.274, P = 0.017, women; spearman's rank: 0.378, P = 0.048). Conclusion High serum ATX levels can be associated with increasing adverse clinical outcomes in patients with NIDCM. These results indicate serum ATX may be a novel biomarker or therapeutic target in NIDCM.
Collapse
Affiliation(s)
- Takashi Araki
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hiroaki Hiraiwa
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Takashi Mizutani
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Yuki Kimura
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Shingo Kazama
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Naoki Shibata
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hideo Oishi
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Tasuku Kuwayama
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Toru Kondo
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Ryota Morimoto
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
18
|
Huang B, Tian L, Chen Z, Zhang L, Su W, Lu T, Yang Y, Hui R, Wang X, Fan X. Angiopoietin 2 as a Novel Potential Biomarker for Acute Aortic Dissection. Front Cardiovasc Med 2022; 8:743519. [PMID: 35004874 PMCID: PMC8733161 DOI: 10.3389/fcvm.2021.743519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/05/2021] [Indexed: 01/16/2023] Open
Abstract
Biomarker-assisted diagnosis of acute aortic dissection (AAD) is important for initiation of treatment and improved survival. However, identification of biomarkers for AAD in blood is a challenging task. The present study aims to find the potential AAD biomarkers using a transcriptomic strategy. Arrays based genome-wide gene expression profiling were performed using ascending aortic tissues which were collected from AAD patients and healthy donors. The differentially expressed genes were validated using quantitative reverse transcriptase PCR (qRT-PCR) and western blot. The plasma levels of a potential biomarker, angiopoietin 2 (ANGPT2) were determined in case-control cohort (77 AAD patients and 82 healthy controls) by enzyme linked immunosorbent assay. Receiver operating characteristic curve (ROC) was used to evaluate the diagnostic power of ANGPT2 for AAD. Transcriptome data demonstrated that a total of 18 genes were significantly up-regulated and 28 genes were significantly down-regulated among AAD tissues (foldchange>3.0, p < 0.01). By bioinformatic analysis, we identified ANGPT2 as a candidate biomarker for blood-based detection of AAD. The qRT-PCR and protein expression demonstrated that ANGPT2 increased 2.4- and 4.2 folds, respectively in aortic tissue of AAD patients. Immunohistochemical staining demonstrated that ANGPT2 was markedly increased in intima of the aortic wall in AAD. Furthermore, ANGPT2 was significantly elevated in AAD patients as compared with controls (median 1625 vs. 383 pg/ml, p < 1E-6). ROC curve analysis showed that ANGPT2 was highly predictive of a diagnosis of type A AAD (area under curve 0.93, p < 1E-6). Sensitivity and specificity were 81 and 90%, respectively at the cutoff value of 833 pg/ml. In conclusion, ANGPT2 could be a promising biomarker for diagnosis of AAD; however, more studies are still needed to verify its specificity in diagnosing of AAD.
Collapse
Affiliation(s)
- Bi Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Tian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhaoran Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Geriatrics and Gerontology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Liang Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjun Su
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianyi Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanmin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rutai Hui
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaojian Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohan Fan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
Lassus J, Tarvasmäki T, Tolppanen H. Biomarkers in cardiogenic shock. Adv Clin Chem 2022; 109:31-73. [DOI: 10.1016/bs.acc.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
20
|
Thakkar AB, Ma Y, Dela Cruz M, Wu Y, Arechiga V, Swaminathan S, Ganz P, Wu AHB, Scherzer R, Deeks S, Hsue PY. Effect of HIV-1 Infection on Angiopoietin 1 and 2 Levels and Measures of Microvascular and Macrovascular Endothelial Dysfunction. J Am Heart Assoc 2021; 10:e021397. [PMID: 34726064 PMCID: PMC8751943 DOI: 10.1161/jaha.121.021397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Individuals infected with HIV have an increased risk of developing cardiovascular disease; yet, the underlying mechanisms remain unknown. Recent evidence has implicated the Tie-2 tyrosine kinase receptor system and its associated ligands ANG1 (angiopoietin 1) and ANG2 (angiopoietin 2) in maintaining vascular homeostasis. In the general population, lower ANG1 levels and higher ANG2 levels are strongly correlated with the development of cardiovascular disease. In this study, we aim to investigate the associations of HIV infection with angiopoietin levels and endothelial dysfunction. Methods and Results In this cross-sectional study, we compared measures of ANG1, ANG2, and endothelial dysfunction using flow-mediated vasodilation of the brachial artery in 39 untreated subjects infected with HIV, 47 treated subjects infected with HIV, and 46 uninfected subjects from the SCOPE (Observational Study of the Consequences of the Protease Inhibitor Era) cohort. Compared with uninfected controls, treated individuals infected with HIV had 53.1% lower mean ANG1 levels (P<0.01) and similar ANG2 levels. On the other hand, untreated individuals infected with HIV had similar ANG1 levels, and 29.2% had higher ANG2 levels (P<0.01) compared with uninfected controls. When compared with individuals with untreated HIV infection, those with treated HIV infection had 56% lower ANG1 levels (P<0.01) and 22% lower ANG2 levels (P<0.01).Both treated and untreated HIV infection were associated with significant impairment in hyperemic velocity, a key measure of microvascular dysfunction (median 61 versus 72 cm/s, P<0.01), compared with uninfected controls (median 73 cm/s). This difference persisted after adjustment for ANG1 and ANG2 levels. Interestingly, when compared with untreated individuals infected with HIV, treated individuals infected with HIV had worse hyperemic velocity (-12.35 cm/s, P=0.05). In contrast, HIV status, ANG1 levels, and ANG2 levels were not associated with macrovascular dysfunction as measured by flow-mediated dilatation and brachial artery diameter, 2 other measures of vascular homeostasis. Conclusions HIV infection affects the balance between levels of ANG1 and ANG2 and may disturb endothelial homeostasis through disruption of vascular homeostasis. Individuals with treated HIV had decreased ANG1 levels and similar ANG2 levels, whereas individuals with untreated HIV had similar ANG1 levels and increased ANG2 levels, suggesting that treatment status may alter the balance between ANG1 and ANG2. HIV also promotes endothelial dysfunction via impairment of microvascular dysfunction, independent of the Tie-2 receptor system; the finding of worse microvascular dysfunction in the setting of treated HIV infection may reflect the impact of viral persistence on the microvasculature or toxicities of specific antiretroviral regimens. Further research to clarify the mechanism of HIV-mediated endothelial dysfunction is necessary to advance treatment of cardiovascular complications of HIV infection.
Collapse
Affiliation(s)
- Anjali B Thakkar
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Yifei Ma
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Mark Dela Cruz
- Section of Cardiology Department of Medicine University of Chicago Medical Center Chicago IL
| | - Yuaner Wu
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Victor Arechiga
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Shreya Swaminathan
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Peter Ganz
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Alan H B Wu
- Division of Clinical Chemistry Department of Laboratory Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Rebecca Scherzer
- San Francisco Veteran's Affairs Medical Center San Francisco CA.,Department of Medicine University of California San Francisco CA
| | - Steven Deeks
- Positive Health Program Zuckerberg San Francisco General HospitalUniversity of California San Francisco CA
| | - Priscilla Y Hsue
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| |
Collapse
|
21
|
Aarsetøy R, Ueland T, Aukrust P, Michelsen AE, de la Fuente RL, Pönitz V, Brügger-Andersen T, Grundt H, Staines H, Nilsen DWT. Angiopoietin-2 and angiopoietin-like 4 protein provide prognostic information in patients with suspected acute coronary syndrome. J Intern Med 2021; 290:894-909. [PMID: 34237166 DOI: 10.1111/joim.13339] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Plasma levels of angiopoietin-2 (ANGPT2) and angiopoietin-like 4 protein (ANGPTL4) reflect different pathophysiological aspects of cardiovascular disease. We evaluated their association with outcome in a hospitalized Norwegian patient cohort (n = 871) with suspected acute coronary syndrome (ACS) and validated our results in a similar Argentinean cohort (n = 982). METHODS A cox regression model, adjusting for traditional cardiovascular risk factors, was fitted for ANGPT2 and ANGPTL4, respectively, with all-cause mortality and cardiac death within 24 months and all-cause mortality within 60 months as the dependent variables. RESULTS At 24 months follow-up, 138 (15.8%) of the Norwegian and 119 (12.1%) of the Argentinian cohort had died, of which 86 and 66 deaths, respectively, were classified as cardiac. At 60 months, a total of 259 (29.7%) and 173 (17.6%) patients, respectively, had died. ANGPT2 was independently associated with all-cause mortality in both cohorts at 24 months [hazard ratio (HR) 1.27 (95% confidence interval (CI), 1.08-1.50) for Norway, and HR 1.57 (95% CI, 1.27-1.95) for Argentina], with similar results at 60 months [HR 1.19 (95% CI, 1.05-1.35) (Norway), and HR 1.56 (95% CI, 1.30-1.88) (Argentina)], and was also significantly associated with cardiac death [HR 1.51 (95% CI, 1.14-2.00)], in the Argentinean population. ANGPTL4 was significantly associated with all-cause mortality in the Argentinean cohort at 24 months [HR 1.39 (95% CI, 1.15-1.68)] and at 60 months [HR 1.43 (95% CI, 1.23-1.67)], enforcing trends in the Norwegian population. CONCLUSIONS ANGPT2 and ANGPTL4 were significantly associated with outcome in similar ACS patient cohorts recruited on two continents. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT00521976. ClinicalTrials.gov Identifier: NCT01377402.
Collapse
Affiliation(s)
- Reidun Aarsetøy
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | | | - Volker Pönitz
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway
| | | | - Heidi Grundt
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Respiratory Medicine, Stavanger University Hospital, Stavanger, Norway
| | | | - Dennis W T Nilsen
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
22
|
Zhang Z, Hu Q, Hu T. Association of Lymphocyte to Monocyte Ratio and Risk of in-Hospital Mortality in Patients with Cardiogenic Shock: A Propensity Score Matching Study. Int J Gen Med 2021; 14:4459-4468. [PMID: 34408483 PMCID: PMC8367081 DOI: 10.2147/ijgm.s325907] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 07/21/2021] [Indexed: 11/23/2022] Open
Abstract
Background Lymphocyte to monocyte ratio (LMR) has been long implicated in the prediction of many inflammatory-related diseases. However, the possible value as prognostic marker of LMR have not been evaluated in cardiogenic shock (CS) patients. The aim of the study was to assess the relationship between LMR on admission and in-hospital mortality in CS patients. Methods Data on patients diagnosed with CS were extracted from the Medical Information Mart for Intensive Care-IV (MIMIC-IV) database. We performed a single-institution, retrospective study of 1487 CS patients and determined the optimal cut-off for LMR by X-tile software. Propensity score matching (PSM) and inverse probabilities of treatment weighting (IPTW) were conducted to control confounders. Cox proportional hazards model was performed to evaluate the relationship between LMR and in-hospital mortality. Kaplan-Meier curves and receiver operating characteristics (ROC) analysis were applied to assess the prognostic value of LMR. Results The optimal cut-off value for LMR was 0.9. Cox proportional hazards model demonstrated that lower LMR (< 0.9) was independently associated with in-hospital mortality with hazard ratio (HR) of 1.40 (1.12-1.74, P = 0.003). The results were consistent with survival analyses (P < 0.001, Log rank test). Adding LMR< 0.9 to the sequential organ failure assessment (SOFA) score improved discrimination and risk stratification for in-hospital mortality. Conclusion Lower level of LMR is related to higher risk of in-hospital mortality of patients with CS. As an easily available biomarker, LMR can independently predict the in-hospital mortality in CS patients.
Collapse
Affiliation(s)
- Zhengwei Zhang
- Department of Critical Care Medicine, Chengdu Second People's Hospital, Chengdu, People's Republic of China
| | - Qionghua Hu
- Department of Critical Care Medicine, Chengdu Second People's Hospital, Chengdu, People's Republic of China
| | - Tianyang Hu
- Department of Cardiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
23
|
Singh S, Kanwar A, Sundaragiri PR, Cheungpasitporn W, Truesdell AG, Rab ST, Singh M, Vallabhajosyula S. Acute Kidney Injury in Cardiogenic Shock: An Updated Narrative Review. J Cardiovasc Dev Dis 2021; 8:88. [PMID: 34436230 PMCID: PMC8396972 DOI: 10.3390/jcdd8080088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/19/2022] Open
Abstract
Acute myocardial infarction with cardiogenic shock (AMI-CS) is associated with high mortality and morbidity despite advancements in cardiovascular care. AMI-CS is associated with multiorgan failure of non-cardiac organ systems. Acute kidney injury (AKI) is frequently seen in patients with AMI-CS and is associated with worse mortality and outcomes compared to those without. The pathogenesis of AMI-CS associated with AKI may involve more factors than previously understood. Early use of renal replacement therapies, management of comorbid conditions and judicious fluid administration may help improve outcomes. In this review, we seek to address the etiology, pathophysiology, management, and outcomes of AKI complicating AMI-CS.
Collapse
Affiliation(s)
- Sohrab Singh
- Department of Medicine, The Brooklyn Hospital, Brooklyn, NY 11201, USA;
| | - Ardaas Kanwar
- Department of Medicine, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA;
| | - Pranathi R. Sundaragiri
- Section of Primary Care Internal Medicine, Wake Forest Baptist Health, High Point, NC 27262, USA;
| | - Wisit Cheungpasitporn
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | | | - Syed Tanveer Rab
- Section of Interventional Cardiology, Division of Cardiovascular Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Mandeep Singh
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Saraschandra Vallabhajosyula
- Section of Cardiovascular Medicine, Department of Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27262, USA
| |
Collapse
|
24
|
Sciaccaluga C, Mandoli GE, Ghionzoli N, Anselmi F, Dini CS, Righini F, Cesareo F, D'Ascenzi F, Focardi M, Valente S, Cameli M. Risk stratification in cardiogenic shock: a focus on the available evidence. Heart Fail Rev 2021; 27:1105-1117. [PMID: 34263413 PMCID: PMC9197897 DOI: 10.1007/s10741-021-10140-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 11/25/2022]
Abstract
Cardiogenic shock is a clinical syndrome which is defined as the presence of primary cardiac disorder that results in hypotension together with signs of organ hypoperfusion in the state of normovolaemia or hypervolaemia. It represents a complex life-threatening condition, characterized by a high mortality rate, that requires urgent diagnostic assessment as well as treatment; therefore, it is of paramount important to advocate for a thorough risk stratification. In fact, the early identification of patients that could benefit the most from more aggressive and invasive approaches could facilitate a more efficient resource allocation. This review attempts to critically analyse the current evidence on prognosis in cardiogenic shock, focusing in particular on clinical, laboratoristic and echocardiographic prognostic parameters. Furthermore, it focuses also on the available prognostic scores, highlighting the strengths and the possible pitfalls. Finally, it provides insights into future direction that could be followed in order to ameliorate risk stratification in this delicate subset of patients.
Collapse
Affiliation(s)
- C Sciaccaluga
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy.
| | - G E Mandoli
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - N Ghionzoli
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - F Anselmi
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - C Sorini Dini
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - F Righini
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - F Cesareo
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - F D'Ascenzi
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - M Focardi
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - S Valente
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| | - M Cameli
- Department of Medical Biotechnologies, Section of Cardiology, University of Siena, Siena, Italy
| |
Collapse
|
25
|
Ceglarek U, Schellong P, Rosolowski M, Scholz M, Willenberg A, Kratzsch J, Zeymer U, Fuernau G, de Waha-Thiele S, Büttner P, Jobs A, Freund A, Desch S, Feistritzer HJ, Isermann B, Thiery J, Pöss J, Thiele H. The novel cystatin C, lactate, interleukin-6, and N-terminal pro-B-type natriuretic peptide (CLIP)-based mortality risk score in cardiogenic shock after acute myocardial infarction. Eur Heart J 2021; 42:2344-2352. [PMID: 33647946 DOI: 10.1093/eurheartj/ehab110] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/21/2020] [Accepted: 02/10/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Cardiogenic shock (CS) complicating acute myocardial infarction (AMI) still reaches excessively high mortality rates. This analysis is aimed to develop a new easily applicable biomarker-based risk score. METHODS AND RESULTS A biomarker-based risk score for 30-day mortality was developed from 458 patients with CS complicating AMI included in the randomized CULPRIT-SHOCK trial. The selection of relevant predictors and the coefficient estimation for the prognostic model were performed by a penalized multivariate logistic regression analysis. Validation was performed internally, internally externally as well as externally in 163 patients with CS included in the randomized IABP-SHOCK II trial. Blood samples were obtained at randomization. The two trials are registered with ClinicalTrials.gov (NCT01927549 and NCT00491036), are closed to new participants, and follow-up is completed. Out of 58 candidate variables, the four strongest predictors for 30-day mortality were included in the CLIP score (cystatin C, lactate, interleukin-6, and N-terminal pro-B-type natriuretic peptide). The score was well calibrated and yielded high c-statistics of 0.82 [95% confidence interval (CI) 0.78-0.86] in internal validation, 0.82 (95% CI 0.75-0.89) in internal-external (temporal) validation, and 0.73 (95% CI 0.65-0.81) in external validation. Notably, it outperformed the Simplified Acute Physiology Score II and IABP-SHOCK II risk score in prognostication (0.83 vs 0.62; P < 0.001 and 0.83 vs. 0.76; P = 0.03, respectively). CONCLUSIONS A biomarker-only score for 30-day mortality risk stratification in infarct-related CS was developed, extensively validated and calibrated in a prospective cohort of contemporary patients with CS after AMI. The CLIP score outperformed other clinical scores and may be useful as an early decision tool in CS.
Collapse
Affiliation(s)
- Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, P.-List-Str. 13, 04103 Leipzig, Germany
| | - Paul Schellong
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, P.-List-Str. 13, 04103 Leipzig, Germany
| | - Maciej Rosolowski
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, P.-List-Str. 13, 04103 Leipzig, Germany
| | - Markus Scholz
- Institute of Medical Informatics, Statistics and Epidemiology, University Leipzig, Germany
| | - Anja Willenberg
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, P.-List-Str. 13, 04103 Leipzig, Germany
| | - Jürgen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, P.-List-Str. 13, 04103 Leipzig, Germany
| | - Uwe Zeymer
- Klinikum Ludwigshafen und Institut für Herzinfarktforschung, Ludwigshafen, Germany
| | | | - Suzanne de Waha-Thiele
- University Heart Center Lübeck, Lübeck, Germany.,German Center for Cardiovascular Research (DZHK), Lübeck, Germany
| | - Petra Büttner
- Heart Center Leipzig at University of Leipzig, Department of Internal Medicine/Cardiology, Strümpellstr.. 39, 04289 Leipzig, and Leipzig Heart Institute, Leipzig, Germany
| | - Alexander Jobs
- German Center for Cardiovascular Research (DZHK), Lübeck, Germany.,Heart Center Leipzig at University of Leipzig, Department of Internal Medicine/Cardiology, Strümpellstr.. 39, 04289 Leipzig, and Leipzig Heart Institute, Leipzig, Germany
| | - Anne Freund
- Heart Center Leipzig at University of Leipzig, Department of Internal Medicine/Cardiology, Strümpellstr.. 39, 04289 Leipzig, and Leipzig Heart Institute, Leipzig, Germany
| | - Steffen Desch
- German Center for Cardiovascular Research (DZHK), Lübeck, Germany.,Heart Center Leipzig at University of Leipzig, Department of Internal Medicine/Cardiology, Strümpellstr.. 39, 04289 Leipzig, and Leipzig Heart Institute, Leipzig, Germany
| | - Hans-Josef Feistritzer
- Heart Center Leipzig at University of Leipzig, Department of Internal Medicine/Cardiology, Strümpellstr.. 39, 04289 Leipzig, and Leipzig Heart Institute, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, P.-List-Str. 13, 04103 Leipzig, Germany
| | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, P.-List-Str. 13, 04103 Leipzig, Germany
| | - Janine Pöss
- Heart Center Leipzig at University of Leipzig, Department of Internal Medicine/Cardiology, Strümpellstr.. 39, 04289 Leipzig, and Leipzig Heart Institute, Leipzig, Germany
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig, Department of Internal Medicine/Cardiology, Strümpellstr.. 39, 04289 Leipzig, and Leipzig Heart Institute, Leipzig, Germany
| |
Collapse
|
26
|
Circulating biomarkers to assess cardiovascular function in critically ill. Curr Opin Crit Care 2021; 27:261-268. [PMID: 33899816 DOI: 10.1097/mcc.0000000000000829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Circulatory shock is one of the most common reasons for ICU admission. Mortality rates in excess of 40% necessitate the rapid identification of high-risk patients, as well as the early assessment of effects of initiated treatments. There is an unmet medical need for circulating biomarkers that may improve patient stratification, predict responses to treatment interventions and may even be a target for novel therapies, enabling a better biological rationale to personalize therapy. RECENT FINDINGS Apart from established biomarkers such as lactate, ScvO2 or NT-pro-BNP, novel biomarkers, including adrenomedullin, angiopoietins, angiotensin I/II ratios, renin and DPP3 show promise, as they are all associated with well defined, therapeutically addressable molecular pathways that are dysregulated during circulatory shock. Although some of the therapies related to these biomarkers are still in preclinical stages of development, they may represent personalized treatment opportunities for patients in circulatory shock. SUMMARY From a molecular perspective, shock represents a highly heterologous syndrome, in which multiple unique pathways are dysregulated. Assessment of the status of these pathways with circulating biomarkers may provide a unique opportunity to detect specific phenotypes and implement personalized medicine in the treatment of circulatory shock.
Collapse
|
27
|
Relationship between markers of inflammation and hemodynamic stress and death in patients with out-of-hospital cardiac arrest. Sci Rep 2021; 11:9954. [PMID: 33976254 PMCID: PMC8113496 DOI: 10.1038/s41598-021-88474-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/12/2021] [Indexed: 11/21/2022] Open
Abstract
Biomarkers that reflect hemodynamic stress, inflammation, extracellular matrix remodeling, angiogenesis, and endothelial dysfunction may improve risk stratification and add valuable pathobiological insight in patients with out-of-hospital cardiac arrest (OHCA). In total, 120 patients with OHCA who survived at least 48 h after return of spontaneous circulation were consecutively included in the present analysis. Concentrations of 30 biomarkers were measured simultaneously using a multi-panel biomarker assay. Cox regression models were adjusted for age, sex, estimated glomerular filtration rate, lactate concentration, bystander resuscitation, initial cardiac rhythm, and type of targeted temperature management. Overall, 57 patients (47.5%) had a favorable neurological outcome (Cerebral Performance Category ≤ 2) at 30 days, while palliative care was initiated in 49 patients (40.8%), and 52 patients (43.3%) died. After correction for multiple testing with Bonferroni-Holm, 8 biomarkers (including Angiopoietin-2, Procalcitonin, Resistin, IL-4Rα, MMP-8, TNFα, Renin, and IL-1α) were significantly associated with all-cause death. After multivariable adjustment, only angiopoietin-2 (Adjusted (Adj) hazard ratio (HR) per 1-unit increase in standardized biomarker concentrations 1.52 (95% CI 1.16–1.99)) and renin (Adj HR 1.32 (95% CI 1.06–1.65) remained independently associated with an increased risk of death. The discriminatory performance indicated good performance for angiopoietin-2 (area under the curve (AUC): 0.75 (95% CI 0.66–0.75) and was significantly higher (P = 0.011) as compared with renin (AUC: 0.60, 95% CI 0.50–0.60). In conclusion, angiopoietin-2 was significantly associated with all-cause mortality in patients with OHCA who survived the first 48 h and may prove to be useful for risk stratification of these patients.
Collapse
|
28
|
Newman J, Brailovsky Y, Allen S, Bontekoe E, Masic D, Walenga J, Fareed J, Darki A. Angiopoietin-2 correlates with pulmonary embolism severity, right ventricular dysfunction, and intensive care unit admission. Vasc Med 2021; 26:556-560. [PMID: 33840325 DOI: 10.1177/1358863x211002276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Risk stratification of acute pulmonary embolism (PE) is important to identify patients at risk for hemodynamic collapse who would benefit from more aggressive therapies. Angiopoietin-2 (Ang-2) is a signaling molecule involved in angiogenesis and is upregulated in response to tissue hypoxia. We aimed to assess the association of Ang-2 with (1) PE severity, (2) echocardiographic and invasive hemodynamic markers of right ventricular (RV) dysfunction, and (3) need for intensive treatment. Patients presenting to our institution with acute PE were included in a prospective database and blood samples were collected and stored for later analysis. A total of 65 patients were included in the study. Ang-2 correlated with PE risk stratification and echocardiographic and invasive hemodynamic markers of RV dysfunction and pulmonary hypertension. An Ang-2 level of > 4101 pg/mL had an odds ratio of 7.4 (95% CI: 1.53-12.5, p < 0.01) for intensive care unit (ICU) admission. In conclusion, Ang-2 correlates with PE severity, RV dysfunction, and need for ICU admission. Ang-2 holds promise as a novel marker that can aid in risk stratification for this patient population.
Collapse
Affiliation(s)
- Joshua Newman
- Department of Medicine, Division of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Yevgeniy Brailovsky
- Advanced Heart Failure, Mechanical Circulatory Support, Heart Transplantation, Jefferson Heart Institute, Sidney Kimmel School of Medicine, Philadelphia, PA, USA
| | - Sorcha Allen
- Department of Medicine, Division of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Emily Bontekoe
- Cardiovascular Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Dalila Masic
- Department of Medicine, Division of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Jeanine Walenga
- Cardiovascular Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Jawed Fareed
- Cardiovascular Research Institute, Loyola University Chicago Health Sciences Campus, Maywood, IL, USA
| | - Amir Darki
- Department of Medicine, Division of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
29
|
Morici N, Marini C, Sacco A, Tavazzi G, Cipriani M, Oliva F, Rota M, De Ferrari GM, Campolo J, Frigerio G, Valente S, Leonardi S, Corrada E, Bottiroli M, Grosseto D, Cacciavillani L, Frigerio M, Pappalardo F. Early intra-aortic balloon pump in acute decompensated heart failure complicated by cardiogenic shock: Rationale and design of the randomized Altshock-2 trial. Am Heart J 2021; 233:39-47. [PMID: 33338464 DOI: 10.1016/j.ahj.2020.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cardiogenic shock (CS) is a systemic disorder associated with dismal short-term prognosis. Given its time-dependent nature, mechanical circulatory support may improve survival. Intra-aortic balloon pump (IABP) had gained widespread use because of the easiness to implant and the low rate of complications; however, a randomized trial failed to demonstrate benefit on mortality in the setting of acute myocardial infarction. Acute decompensated heart failure with cardiogenic shock (ADHF-CS) represents a growing resource-intensive scenario with scant data and indications on the best management. However, a few data suggest a potential benefit of IABP in this setting. We present the design of a study aimed at addressing this research gap. METHODS AND DESIGN The Altshock-2 trial is a prospective, randomized, multicenter, open-label study with blinded adjudicated evaluation of outcomes. Patients with ADHF-CS will be randomized to early IABP implantation or to vasoactive treatments. The primary end point will be 60 days patients' survival or successful bridge to heart replacement therapy. The key secondary end point will be 60-day overall survival; 60-day need for renal replacement therapy; in-hospital maximum inotropic score, maximum duration of inotropic/vasopressor therapy, and maximum sequential organ failure assessment score. Safety end points will be in-hospital occurrence of bleeding events (Bleeding Academic Research Consortium >3), vascular access complications and systemic (noncerebral) embolism. The sample size for the study is 200 patients. IMPLICATIONS The Altshock-2 trial will provide evidence on whether IABP should be implanted early in ADHF-CS patients to improve their clinical outcomes.
Collapse
Affiliation(s)
- Nuccia Morici
- Intensive Cardiac Care Unit, De Gasperis Cardio Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| | - Claudia Marini
- Intensive Cardiac Care Unit, De Gasperis Cardio Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alice Sacco
- Intensive Cardiac Care Unit, De Gasperis Cardio Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Guido Tavazzi
- Emergency Department, Anaesthesia and Intensive Care Unit, Pavia, Italy
| | - Manlio Cipriani
- Heart Failure and Transplant Unit, De Gasperis Cardio Center and Transplant Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Fabrizio Oliva
- Intensive Cardiac Care Unit, De Gasperis Cardio Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Matteo Rota
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Jonica Campolo
- CNR Institute of Clinical Physiology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Gianfranco Frigerio
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Serafina Valente
- Cardiovascular and Thoracic Department, Azienda Ospedaliera Universitaria Senese, Policlinico Santa Maria alle Scotte, Siena, Italy
| | - Sergio Leonardi
- Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology - Fondazione IRCCS Policlinico San Matteo, and Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Elena Corrada
- Cardiovascular Department, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Maurizio Bottiroli
- Cardiothoracic Anesthesiology Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Luisa Cacciavillani
- Department of Cardiac, Thoracic, and Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Maria Frigerio
- Heart Failure and Transplant Unit, De Gasperis Cardio Center and Transplant Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federico Pappalardo
- Department of Anesthesia and Intensive Care, IRCCS ISMETT, UPMC Italy, Palermo, Italy
| |
Collapse
|
30
|
Hayashi SI, Rakugi H, Morishita R. Insight into the Role of Angiopoietins in Ageing-Associated Diseases. Cells 2020; 9:E2636. [PMID: 33302426 PMCID: PMC7762563 DOI: 10.3390/cells9122636] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
Angiopoietin (Ang) and its receptor, TIE signaling, contribute to the development and maturation of embryonic vasculature as well as vascular remodeling and permeability in adult tissues. Targeting both this signaling pathway and the major pathway with vascular endothelial growth factor (VEGF) is expected to permit clinical applications, especially in antiangiogenic therapies against tumors. Several drugs targeting the Ang-TIE signaling pathway in cancer patients are under clinical development. Similar to how cancer increases with age, unsuitable angiogenesis or endothelial dysfunction is often seen in other ageing-associated diseases (AADs) such as atherosclerosis, Alzheimer's disease, type 2 diabetes, chronic kidney disease and cardiovascular diseases. Thus, the Ang-TIE pathway is a possible molecular target for AAD therapy. In this review, we focus on the potential role of the Ang-TIE signaling pathway in AADs, especially non-cancer-related AADs. We also suggest translational insights and future clinical applications of this pathway in those AADs.
Collapse
Affiliation(s)
- Shin-ichiro Hayashi
- Department of Clinical Gene Therapy, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan;
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| |
Collapse
|
31
|
Wu X, Reboll MR, Korf-Klingebiel M, Wollert KC. Angiogenesis after acute myocardial infarction. Cardiovasc Res 2020; 117:1257-1273. [PMID: 33063086 DOI: 10.1093/cvr/cvaa287] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/09/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Acute myocardial infarction (MI) inflicts massive injury to the coronary microcirculation leading to vascular disintegration and capillary rarefication in the infarct region. Tissue repair after MI involves a robust angiogenic response that commences in the infarct border zone and extends into the necrotic infarct core. Technological advances in several areas have provided novel mechanistic understanding of postinfarction angiogenesis and how it may be targeted to improve heart function after MI. Cell lineage tracing studies indicate that new capillary structures arise by sprouting angiogenesis from pre-existing endothelial cells (ECs) in the infarct border zone with no meaningful contribution from non-EC sources. Single-cell RNA sequencing shows that ECs in infarcted hearts may be grouped into clusters with distinct gene expression signatures, likely reflecting functionally distinct cell populations. EC-specific multicolour lineage tracing reveals that EC subsets clonally expand after MI. Expanding EC clones may arise from tissue-resident ECs with stem cell characteristics that have been identified in multiple organs including the heart. Tissue repair after MI involves interactions among multiple cell types which occur, to a large extent, through secreted proteins and their cognate receptors. While we are only beginning to understand the full complexity of this intercellular communication, macrophage and fibroblast populations have emerged as major drivers of the angiogenic response after MI. Animal data support the view that the endogenous angiogenic response after MI can be boosted to reduce scarring and adverse left ventricular remodelling. The improved mechanistic understanding of infarct angiogenesis therefore creates multiple therapeutic opportunities. During preclinical development, all proangiogenic strategies should be tested in animal models that replicate both cardiovascular risk factor(s) and the pharmacotherapy typically prescribed to patients with acute MI. Considering that the majority of patients nowadays do well after MI, clinical translation will require careful selection of patients in need of proangiogenic therapies.
Collapse
Affiliation(s)
- Xuekun Wu
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Marc R Reboll
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Mortimer Korf-Klingebiel
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Kai C Wollert
- Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| |
Collapse
|
32
|
Varricchi G, Loffredo S, Bencivenga L, Ferrara AL, Gambino G, Ferrara N, de Paulis A, Marone G, Rengo G. Angiopoietins, Vascular Endothelial Growth Factors and Secretory Phospholipase A 2 in Ischemic and Non-Ischemic Heart Failure. J Clin Med 2020; 9:jcm9061928. [PMID: 32575548 PMCID: PMC7356305 DOI: 10.3390/jcm9061928] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a growing public health burden, with high prevalence and mortality rates. In contrast to ischemic heart failure (IHF), the diagnosis of non-ischemic heart failure (NIHF) is established in the absence of coronary artery disease. Angiopoietins (ANGPTs), vascular endothelial growth factors (VEGFs) and secretory phospholipases A2 (sPLA2s) are proinflammatory mediators and key regulators of endothelial cells. In the present manuscript, we analyze the plasma concentrations of angiogenic (ANGPT1, ANGPT2, VEGF-A) and lymphangiogenic (VEGF-C, VEGF-D) factors and the plasma activity of sPLA2 in patients with IHF and NIHF compared to healthy controls. The concentrations of ANGPT1, ANGPT2 and their ratio significantly differed between HF patients and healthy controls. Similarly, plasma levels of VEGF-D and sPLA2 activity were higher in HF as compared to controls. Concentrations of ANGPT2 and the ANGPT2/ANGPT1 ratio (an index of vascular permeability) were increased in NIHF patients. VEGF-A and VEGF-C concentrations did not differ among the three examined groups. Interestingly, VEGF-D was selectively increased in IFH patients compared to controls. Plasma activity of sPLA2 was increased in IHF and NIHF patients compared to controls. Our results indicate that several regulators of vascular permeability and smoldering inflammation are specifically altered in IHF and NIHF patients. Studies involving larger cohorts of these patients will be necessary to demonstrate the clinical implications of our findings.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80100 Naples, Italy
- Correspondence: (G.V.); (S.L.)
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80100 Naples, Italy
- Correspondence: (G.V.); (S.L.)
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80100 Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
| | - Giuseppina Gambino
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80100 Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80100 Naples, Italy
- Institute of Experimental Endocrinology and Oncology “G. Salvatore” (IEOS), National Research Council (CNR), 80100 Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy; (L.B.); (A.L.F.); (G.G.); (N.F.); (A.d.P.); (G.M.); (G.R.)
- Istituti Clinici Scientifici Maugeri SpA Società Benefit, Via Bagni Vecchi, 1, 82037 Telese BN, Italy
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Cardiogenic shock remains beside sudden cardiac death the most outcome relevant complication of acute myocardial infarction. Over the last two decades as confirmation of the benefit of early revascularization no further relevant improvement in outcome could be achieved. Biomarkers are important for diagnosis, monitoring, and management in cardiogenic shock patients. RECENT FINDINGS A bunch of different biomarkers have been associated with prognosis in patients with cardiogenic shock. In routine use standard parameters such as serum lactate or serum creatinine are still most important in monitoring these patients. These established markers outperformed novel markers in prognostic impact in recent trials. SUMMARY Biomarkers serve as important treatment targets and may help physicians in therapeutic decision-making. Furthermore, the complex pathophysiology of cardiogenic shock may be better understood by investigation of different biomarkers.
Collapse
|
34
|
Cardiogenic Shock: Reflections at the Crossroad Between Perfusion, Tissue Hypoxia, and Mitochondrial Function. Can J Cardiol 2020; 36:184-196. [PMID: 32036863 DOI: 10.1016/j.cjca.2019.11.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/19/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiogenic shock is classically defined by systemic hypotension with evidence of hypoperfusion and end organ dysfunction. In modern practice, however, these metrics often incompletely describe cardiogenic shock because patients present with more advanced cardiovascular disease and greater degrees of multiorgan dysfunction. Understanding how perfusion, congestion, and end organ dysfunction contribute to hypoxia at the cellular level are central to the diagnosis and management of cardiogenic shock. Although, in clinical practice, increased lactate level is often equated with hypoxia, several other factors might contribute to an elevated lactate level including mitochondrial dysfunction, impaired hepatic and renal clearance, as well as epinephrine use. To this end, we present the evidence underlying the value of lactate to pyruvate ratio as a potential discriminator of cellular hypoxia. We will then discuss the physiological implications of hypoxia and congestion on hepatic, intestinal, and renal physiology. Organ-specific susceptibility to hypoxia is presented in the context of their functional architecture. We discuss how the concepts of contractile reserve, fluid responsiveness, tissue oxygenation, and cardiopulmonary interactions can help personalize the management of cardiogenic shock. Finally, we highlight the limitations of using lactate for tailoring therapy in cardiogenic shock.
Collapse
|
35
|
Shirali AS, Lluri G, Guihard PJ, Conrad MB, Kim H, Pawlikowska L, Boström KI, Iruela-Arispe ML, Aboulhosn JA. Angiopoietin-2 predicts morbidity in adults with Fontan physiology. Sci Rep 2019; 9:18328. [PMID: 31797976 PMCID: PMC6892891 DOI: 10.1038/s41598-019-54776-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
Morbidity in patients with single-ventricle Fontan circulation is common and includes arrhythmias, edema, and pulmonary arteriovenous malformations (PAVM) among others. We sought to identify biomarkers that may predict such complications. Twenty-five patients with Fontan physiology and 12 control patients with atrial septal defects (ASD) that underwent cardiac catheterization were included. Plasma was collected from the hepatic vein and superior vena cava and underwent protein profiling for a panel of 20 analytes involved in angiogenesis and endothelial dysfunction. Ten (40%) of Fontan patients had evidence of PAVM, eighteen (72%) had a history of arrhythmia, and five (20%) were actively in arrhythmia or had a recent arrhythmia. Angiopoietin-2 (Ang-2) was higher in Fontan patients (8,875.4 ± 3,336.9 pg/mL) versus the ASD group (1,663.6 ± 587.3 pg/mL, p < 0.0001). Ang-2 was higher in Fontan patients with active or recent arrhythmia (11,396.0 ± 3,457.7 vs 8,118.2 ± 2,795.1 pg/mL, p < 0.05). A threshold of 8,500 pg/mL gives Ang-2 a negative predictive value of 100% and positive predictive value of 42% in diagnosing recent arrhythmia. Ang-2 is elevated among adults with Fontan physiology. Ang-2 level is associated with active or recent arrhythmia, but was not found to be associated with PAVM.
Collapse
Affiliation(s)
- Aditya S Shirali
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Gentian Lluri
- Ahmanson/UCLA Adult Congenital Heart Disease Center, Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA.
| | - Pierre J Guihard
- Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Miles B Conrad
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Helen Kim
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Ludmila Pawlikowska
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Kristina I Boström
- Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell & Developmental Biology, Molecular Biology Institute and Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Jamil A Aboulhosn
- Ahmanson/UCLA Adult Congenital Heart Disease Center, Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
36
|
Rueda F, Borràs E, García-García C, Iborra-Egea O, Revuelta-López E, Harjola VP, Cediel G, Lassus J, Tarvasmäki T, Mebazaa A, Sabidó E, Bayés-Genís A. Protein-based cardiogenic shock patient classifier. Eur Heart J 2019; 40:2684-2694. [DOI: 10.1093/eurheartj/ehz294] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/04/2019] [Accepted: 04/19/2019] [Indexed: 11/15/2022] Open
Abstract
Abstract
Aims
Cardiogenic shock (CS) is associated with high short-term mortality and a precise CS risk stratification could guide interventions to improve patient outcome. Here, we developed a circulating protein-based score to predict short-term mortality risk among patients with CS.
Methods and results
Mass spectrometry analysis of 2654 proteins was used for screening in the Barcelona discovery cohort (n = 48). Targeted quantitative proteomics analyses (n = 51 proteins) were used in the independent CardShock cohort (n = 97) to derive and cross-validate the protein classifier. The combination of four circulating proteins (Cardiogenic Shock 4 proteins—CS4P), discriminated patients with low and high 90-day risk of mortality. CS4P comprises the abundances of liver-type fatty acid-binding protein, beta-2-microglobulin, fructose-bisphosphate aldolase B, and SerpinG1. Within the CardShock cohort used for internal validation, the C-statistic was 0.78 for the CardShock risk score, 0.83 for the CS4P model, and 0.84 (P = 0.033 vs. CardShock risk score) for the combination of CardShock risk score with the CS4P model. The CardShock risk score with the CS4P model showed a marked benefit in patient reclassification, with a net reclassification improvement (NRI) of 0.49 (P = 0.020) compared with CardShock risk score. Similar reclassification metrics were observed in the IABP-SHOCK II risk score combined with CS4P (NRI =0.57; P = 0.032). The CS4P patient classification power was confirmed by enzyme-linked immunosorbent assay (ELISA).
Conclusion
A new protein-based CS patient classifier, the CS4P, was developed for short-term mortality risk stratification. CS4P improved predictive metrics in combination with contemporary risk scores, which may guide clinicians in selecting patients for advanced therapies.
Collapse
Affiliation(s)
- Ferran Rueda
- Heart Institute, Hospital Universitari Germans Trias i Pujol, c/ Canyet SN, 08916 Badalona, Spain
- Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Eva Borràs
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Dr Aiguader 88, Barcelona, Spain
| | - Cosme García-García
- Heart Institute, Hospital Universitari Germans Trias i Pujol, c/ Canyet SN, 08916 Badalona, Spain
- Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Oriol Iborra-Egea
- Heart Institute, Hospital Universitari Germans Trias i Pujol, c/ Canyet SN, 08916 Badalona, Spain
- Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Elena Revuelta-López
- Heart Institute, Hospital Universitari Germans Trias i Pujol, c/ Canyet SN, 08916 Badalona, Spain
- Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Veli-Pekka Harjola
- Emergency Medicine, Department of Emergency Medicine and Services, University of Helsinki, Helsinki University Hospital, Finland
| | - Germán Cediel
- Heart Institute, Hospital Universitari Germans Trias i Pujol, c/ Canyet SN, 08916 Badalona, Spain
- Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Johan Lassus
- Cardiology, University of Helsinki, Heart and Lung Center, Helsinki University Hospital, Finland
| | - Tuukka Tarvasmäki
- Cardiology, University of Helsinki, Heart and Lung Center, Helsinki University Hospital, Finland
| | - Alexandre Mebazaa
- U942 Inserm, University Paris Diderot, APHP Hôpitaux Universitaires Saint-Louis-Lariboisière, INI-CRCT, Paris, France
| | - Eduard Sabidó
- Proteomics Unit, Centre de Regulació Genòmica (CRG), Barcelona Institute of Science and Technology (BIST), Dr Aiguader 88, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Dr Aiguader 88, Barcelona, Spain
| | - Antoni Bayés-Genís
- Heart Institute, Hospital Universitari Germans Trias i Pujol, c/ Canyet SN, 08916 Badalona, Spain
- Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
37
|
Baran DA, Grines CL, Bailey S, Burkhoff D, Hall SA, Henry TD, Hollenberg SM, Kapur NK, O'Neill W, Ornato JP, Stelling K, Thiele H, van Diepen S, Naidu SS. SCAI clinical expert consensus statement on the classification of cardiogenic shock: This document was endorsed by the American College of Cardiology (ACC), the American Heart Association (AHA), the Society of Critical Care Medicine (SCCM), and the Society of Thoracic Surgeons (STS) in April 2019. Catheter Cardiovasc Interv 2019; 94:29-37. [PMID: 31104355 DOI: 10.1002/ccd.28329] [Citation(s) in RCA: 367] [Impact Index Per Article: 73.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The outcome of cardiogenic shock complicating myocardial infarction has not appreciably changed in the last 30 years despite the development of various percutaneous mechanical circulatory support options. It is clear that there are varying degrees of cardiogenic shock but there is no robust classification scheme to categorize this disease state. METHODS A multidisciplinary group of experts convened by the Society for Cardiovascular Angiography and Interventions was assembled to derive a proposed classification schema for cardiogenic shock. Representatives from cardiology (interventional, advanced heart failure, noninvasive), emergency medicine, critical care, and cardiac nursing all collaborated to develop the proposed schema. RESULTS A system describing stages of cardiogenic shock from A to E was developed. Stage A is "at risk" for cardiogenic shock, stage B is "beginning" shock, stage C is "classic" cardiogenic shock, stage D is "deteriorating", and E is "extremis". The difference between stages B and C is the presence of hypoperfusion which is present in stages C and higher. Stage D implies that the initial set of interventions chosen have not restored stability and adequate perfusion despite at least 30 minutes of observation and stage E is the patient in extremis, highly unstable, often with cardiovascular collapse. CONCLUSION This proposed classification system is simple, clinically applicable across the care spectrum from pre-hospital providers to intensive care staff but will require future validation studies to assess its utility and potential prognostic implications.
Collapse
Affiliation(s)
- David A Baran
- Sentara Heart Hospital, Division of Cardiology, Advanced Heart Failure Center and Eastern Virginia Medical School, Norfolk, Virginia
| | - Cindy L Grines
- Department of Cardiology, Zucker School of Medicine at Hofstra/Northwell, North Shore University Hospital, Manhasset, New York
| | - Steven Bailey
- Department of Internal Medicine, LSU Health School of Medicine, Shreveport, Louisiana
| | | | | | - Timothy D Henry
- Lindner Research Center at the Christ Hospital, Cincinnati, Ohio
| | | | - Navin K Kapur
- The CardioVascular Center, Tufts Medical Center, Boston, Massachusetts
| | | | - Joseph P Ornato
- Virginia Commonwealth University Health System, Richmond, Virginia
| | - Kelly Stelling
- Sentara Heart Hospital, Division of Cardiology, Advanced Heart Failure Center and Eastern Virginia Medical School, Norfolk, Virginia
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig, Department of Internal Medicine/Cardiology, Leipzig, Germany
| | - Sean van Diepen
- Department of Critical Care Medicine and Division of Cardiology, University of Alberta, Edmonton, Canada
| | - Srihari S Naidu
- Westchester Medical Center and New York Medical College, Valhalla, New York
| |
Collapse
|
38
|
Jian W, Li L, Wei XM, Wu CQ, Gui C. Prognostic value of angiopoietin-2 for patients with coronary heart disease after elective PCI. Medicine (Baltimore) 2019; 98:e14216. [PMID: 30702576 PMCID: PMC6380730 DOI: 10.1097/md.0000000000014216] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Patients with coronary heart disease (CHD) frequently have cardiovascular complications after undergoing PCI. Angiopoietin-2 (Ang-2) is an important proangiogenic factor that also plays an important role in atherosclerosis. This study aimed to evaluate the value of Ang-2 in predicting cardiovascular events after elective PCI.This prospective study enrolled 97 patients with CHD who underwent elective PCI from 2013 to 2014. Blood samples were collected in the first morning after admission and within 24 to 48 h after PCI. The primary endpoint was cardiovascular events, defined as a composite of cardiac death, nonfatal myocardial infarction/repeat revascularization, readmission for severe deterioration of angina and readmission for new onset heart failure. Based on the median level of pre-PCI or post-PCI Ang-2, the patients were divided into a low level group and a high level group.During the whole follow-up period (mean, 53 ± 13 months), Kaplan-Meier curves of cardiovascular events showed that there was no significant difference between the two pre-PCI groups (χ = 2.22, P = .137, and log-rank test) or the two post-PCI groups (χ = 2.83, P = .093, and log-rank test). However, in a multivariable Cox regression model, landmark analysis showed that the patients in high level group of post-PCI, not pre-PCI, were associated with remarkable higher risks of cardiovascular events compared to the low level group during the first 1.5 years of follow-up (adjusted HR = 9.99, 95%CI = 1.99-50.13, P = .005). However, that was of no significance from 1.5 years to maximum follow-up years (adjusted HR = 0.82, 95%CI = 0.26-2.59, P = .733).High Ang-2 levels of post-PCI can predict the occurrence of cardiovascular events in the short to medium term.
Collapse
Affiliation(s)
- Wen Jian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| | - Xiao-Min Wei
- Department of Cardiology, Gongren Hospital of Wuzhou, Wuzhou
| | - Cheng-Qiang Wu
- Department of Cardiology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, People's Republic of China
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| |
Collapse
|
39
|
Jian W, Li L, Wei XM, Guan JH, Yang GL, Gui C. Serum angiopoietin-2 concentrations of post-PCI are correlated with the parameters of renal function in patients with coronary artery disease. Medicine (Baltimore) 2019; 98:e13960. [PMID: 30608432 PMCID: PMC6344115 DOI: 10.1097/md.0000000000013960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Patients with coronary artery disease (CAD) frequently have comorbidity of chronic kidney disease (CKD). Their renal function may deteriorate because of the use of contrast agent after percutaneous coronary intervention (PCI). Angiopoietin-2 (Ang-2), which is highly expressed in the site of angiogenesis, plays an important role in both CAD and CKD. This study aimed to investigate the relation of serum Ang-2 concentrations with the renal function after PCI.This study enrolled 57 patients with CAD undergoing PCI. Blood samples for Ang-2 were collected in the first morning after admission and within 24 to 48 h after PCI. The parameters of renal function (serum creatinine, cystatin C and eGFR) were tested on the first day after admission and within 72 h after PCI.Overall, serum Ang-2 levels of post-PCI were significantly lower than those of pre-PCI [median, 1733 (IQR, 1100-2568) vs median, 2523 (IQR, 1702-3640) pg/mL; P < .001]. However, in patients with CKD (eGFR < 60 mL/min/1.73 m), there was no significant difference between serum Ang-2 levels of post-PCI and those of pre-PCI [median, 2851 (IQR, 1720-4286) vs. median, 2492 (IQR, 1434-4994) pg/mL; P = .925]. In addition, serum Ang-2 levels of post-PCI, but not pre-PCI, were significantly correlated with the post-PCI parameters of renal function.Serum Ang-2 concentrations of post-PCI are closely related to renal function in patients with CAD. It may have potential to be the early biomarker of contrast-induced nephropathy in the future.
Collapse
Affiliation(s)
- Wen Jian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| | - Xiao-Min Wei
- Department of Cardiology, Gongren Hospital of Wuzhou, Wuzhou
| | - Jia-Hui Guan
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Guo-Liang Yang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University
- Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning
| |
Collapse
|
40
|
Abstract
Myocardial infarction (MI) complicated by cardiogenic shock (MI-CS) is a major cause of cardiovascular morbidity and mortality. Predictors of outcomes in MI-CS include clinical, laboratory, radiologic variables, and management strategies. This article reviews the existing literature on short- and long-term predictors and risk stratification in MI complicated by CS.
Collapse
Affiliation(s)
- Deepak Acharya
- From the Section of Advanced Heart Failure, Mechanical Circulatory Support, and Pulmonary Vascular Disease, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
41
|
Lee SJ, Lee CK, Kang S, Park I, Kim YH, Kim SK, Hong SP, Bae H, He Y, Kubota Y, Koh GY. Angiopoietin-2 exacerbates cardiac hypoxia and inflammation after myocardial infarction. J Clin Invest 2018; 128:5018-5033. [PMID: 30295643 DOI: 10.1172/jci99659] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 08/21/2018] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence indicates that angiopoietin-2 (Angpt2), a well-recognized vascular destabilizing factor, is a biomarker of poor outcome in ischemic heart disease. However, its precise role in postischemic cardiovascular remodeling is poorly understood. Here, we show that Angpt2 plays multifaceted roles in the exacerbation of cardiac hypoxia and inflammation after myocardial ischemia. Angpt2 was highly expressed in endothelial cells at the infarct border zone after myocardial infarction (MI) or ischemia/reperfusion injury in mice. In the acute phase of MI, endothelial-derived Angpt2 antagonized Angpt1/Tie2 signaling, which was greatly involved in pericyte detachment, vascular leakage, increased adhesion molecular expression, degradation of the glycocalyx and extracellular matrix, and enhanced neutrophil infiltration and hypoxia in the infarct border area. In the chronic remodeling phase after MI, endothelial- and macrophage-derived Angpt2 continuously promoted abnormal vascular remodeling and proinflammatory macrophage polarization through integrin α5β1 signaling, worsening cardiac hypoxia and inflammation. Accordingly, inhibition of Angpt2 either by gene deletion or using an anti-Angpt2 blocking antibody substantially alleviated these pathological findings and ameliorated postischemic cardiovascular remodeling. Blockade of Angpt2 thus has potential as a therapeutic option for ischemic heart failure.
Collapse
Affiliation(s)
- Seung-Jun Lee
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea
| | - Choong-Kun Lee
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Seok Kang
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Intae Park
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Yoo Hyung Kim
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Seo Ki Kim
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Seon Pyo Hong
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Hosung Bae
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Yulong He
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Yoshiaki Kubota
- The Laboratory of Vascular Biology, School of Medicine, Keio University, Tokyo, Japan
| | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science, Daejeon, South Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| |
Collapse
|
42
|
Saharinen P, Eklund L, Alitalo K. Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov 2017; 16:635-661. [PMID: 28529319 DOI: 10.1038/nrd.2016.278] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelial angiopoietin (ANG)-TIE growth factor receptor pathway regulates vascular permeability and pathological vascular remodelling during inflammation, tumour angiogenesis and metastasis. Drugs that target the ANG-TIE pathway are in clinical development for oncological and ophthalmological applications. The aim is to complement current vascular endothelial growth factor (VEGF)-based anti-angiogenic therapies in cancer, wet age-related macular degeneration and macular oedema. The unique function of the ANG-TIE pathway in vascular stabilization also renders this pathway an attractive target in sepsis, organ transplantation, atherosclerosis and vascular complications of diabetes. This Review covers key aspects of the function of the ANG-TIE pathway in vascular disease and describes the recent development of novel therapeutics that target this pathway.
Collapse
Affiliation(s)
- Pipsa Saharinen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Aapistie 5A, University of Oulu, 90220 Oulu, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| |
Collapse
|
43
|
Lorusso R. Are two crutches better than one? The ongoing dilemma on the effects and need for left ventricular unloading during veno-arterial extracorporeal membrane oxygenation. Eur J Heart Fail 2016; 19:413-415. [DOI: 10.1002/ejhf.695] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/13/2016] [Accepted: 10/16/2016] [Indexed: 12/16/2022] Open
Affiliation(s)
- Roberto Lorusso
- Cardio-Thoracic Surgery Department, Heart & Vascular Centre; Maastricht University Medical Centre; Maastricht The Netherlands
| |
Collapse
|
44
|
Kataja A, Tarvasmäki T, Lassus J, Cardoso J, Mebazaa A, Køber L, Sionis A, Spinar J, Carubelli V, Banaszewski M, Marino R, Parissis J, Nieminen MS, Harjola VP. The association of admission blood glucose level with the clinical picture and prognosis in cardiogenic shock - Results from the CardShock Study. Int J Cardiol 2016; 226:48-52. [PMID: 27788389 DOI: 10.1016/j.ijcard.2016.10.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 10/09/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Critically ill patients often present with hyperglycemia, regardless of previous history of diabetes mellitus (DM). Hyperglycemia has been associated with adverse outcome in acute myocardial infarction and acute heart failure. We investigated the association of admission blood glucose level with the clinical picture and short-term mortality in cardiogenic shock (CS). METHODS Consecutively enrolled CS patients were divided into five categories according to plasma glucose level at the time of enrolment: hypoglycemia (glucose <4.0mmol/L), normoglycemia (4.0-7.9mmol/L), mild (8.0-11.9mmol/L), moderate (12.0-15.9mmol/L), and severe (≥16.0mmol/L) hyperglycemia. Clinical presentation, biochemistry, and short-term mortality were compared between the groups. RESULTS Plasma glucose level of 211 CS patients was recorded. Glucose levels were distributed equally between normoglycemia (26% of patients), mild (27%), moderate (19%) and severe (25%) hyperglycemia, while hypoglycemia (2%) was rare. Severe hyperglycemia was associated with higher blood leukocyte count (17.3 (5.8) E9/L), higher lactate level (4.4 (3.3-8.4) mmol/L) and lower arterial pH (7.23 (0.14)) compared with normoglycemia or mild to moderate hyperglycemia (p<0.001 for all). In-hospital mortality was highest among hypoglycemic (60%) and severely hyperglycemic (56%) patients, compared with 22% in normoglycemic group (p<0.01). Severe hyperglycemia was an independent predictor of in-hospital mortality (OR 3.7, 95% CI 1.19-11.7, p=0.02), when adjusted for age, gender, LVEF, lactate, and DM. CONCLUSIONS Admission blood glucose level has prognostic significance in CS. Mortality is highest among patients with severe hyperglycemia or hypoglycemia. Severe hyperglycemia is independently associated with high in-hospital mortality in CS. It is also associated with biomarkers of systemic hypoperfusion and stress response.
Collapse
Affiliation(s)
- Anu Kataja
- Emergency Medicine, University of Helsinki, Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland.
| | - Tuukka Tarvasmäki
- Emergency Medicine, University of Helsinki, Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland
| | - Johan Lassus
- Cardiology, University of Helsinki, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Jose Cardoso
- CINTESIS - Center for Health Technology and Services Research, Department of Cardiology, Faculty of Medicine, University of Porto, São João Medical Center, Porto, Portugal
| | - Alexandre Mebazaa
- INSERM U942, Hopital Lariboisiere, APHP and University Paris Diderot, Paris, France
| | - Lars Køber
- Rigshospitalet, Copenhagen University Hospital, Division of Heart Failure, Pulmonary Hypertension and Heart Transplantation, Copenhagen, Denmark
| | - Alessandro Sionis
- Intensive Cardiac Care Unit, Cardiology Department, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute IIB-Sant Pau, Universitat de Barcelona, Barcelona, Spain
| | - Jindrich Spinar
- Internal Cardiology Department, University Hospital Brno and Masaryk University, Brno, Czech republic
| | - Valentina Carubelli
- Division of Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | - Marek Banaszewski
- Intensive Cardiac Therapy Clinic, Institute of Cardiology, Warsaw, Poland
| | - Rossella Marino
- Department of Medical Sciences and Translational Medicine, University of Rome Sapienza, Emergency Department, Sant''Andrea Hospital, Rome, Italy
| | - John Parissis
- Heart Failure Clinic, Attikon University Hospital, Athens, Greece
| | - Markku S Nieminen
- Cardiology, University of Helsinki, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Veli-Pekka Harjola
- Emergency Medicine, University of Helsinki, Department of Emergency Medicine and Services, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
45
|
Metra M. November 2015 at a glance. Eur J Heart Fail 2015. [DOI: 10.1002/ejhf.447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health; University of Brescia; Brescia Italy
| |
Collapse
|