1
|
Hynninen E, Tolppanen H, Rivas‐Lasarte M, Tarvasmäki T, Harjola V, Deniau B, Hongisto M, Jankowska EA, Jurkko R, Jäntti T, Kataja A, Mebazaa A, Sabell T, Sionis A, Lassus J. Validation of a biomarker-based mortality score for cardiogenic shock patients: Comparison with a clinical risk score. ESC Heart Fail 2025; 12:2157-2165. [PMID: 39895206 PMCID: PMC12055353 DOI: 10.1002/ehf2.15234] [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: 09/23/2024] [Revised: 12/12/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
AIMS Cardiogenic shock (CS) is the deadliest manifestation of acute heart failure, with persistently high mortality rates and a lack of recent therapeutic breakthroughs. Accurate risk prediction is crucial in clinical decision-making and the design of future clinical trials. We aimed to validate the CLIP score, a biomarker-based risk score comprising cystatin C, lactate, interleukin-6 and NT-proBNP, for predicting mortality in acute coronary syndrome (ACS) related CS, and to compare its predictive value with the previously published CardShock risk score. METHODS AND RESULTS The study is a post hoc analysis of the CardShock Study, a prospective, observational European multicentre study on CS. The CLIP score was calculated 12 h after hospital admission, and its ability to predict 90-day mortality was assessed using are under the curve (AUC) of the receiver-operating characteristics (ROC) curve analysis. The discriminative ability of the CLIP score was compared with the CardShock risk score by comparing the AUC's. The cohort was dichotomized into low and high risk groups by the optimal cut-off value derived from the ROC analysis of the CLIP score. Kaplan-Meier curves were constructed to evaluate risk stratification when combining the CLIP and CardShock risk scores. The cohort (n = 121) comprised 77% (n = 93) men and the median age was 67 years (IQR 61-76). A total of 21% (n = 25) of the patients had non-ACS related CS. The CLIP score demonstrated appropriate predictive accuracy for 90-day mortality (AUC 0.84, 95% CI 0.77-0.91), comparable with the CardShock risk score (AUC 0.77 [95% CI 0.69-0.85]; P = 0.064 for comparison). A CLIP score cut-off of 0.28 stratified patients into high risk (65% mortality) and low risk (16% mortality) groups. In addition, incorporating the CLIP score enhanced risk stratification in all CardShock risk score categories. CONCLUSIONS The CLIP score, calculated within 12 h of hospital admission, accurately predicted 90-day mortality in CS and complemented the CardShock risk score. The biomarker-based score has potential utility in dynamic mortality risk assessment and could inform clinical management and trial design.
Collapse
Affiliation(s)
- Elina Hynninen
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| | - Heli Tolppanen
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| | - Mercedes Rivas‐Lasarte
- Department of CardiologyHospital Universitario Puerta de Hierro, IDIPHISA, CIBER CVMajadahondaSpain
| | - Tuukka Tarvasmäki
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| | - Veli‐Pekka Harjola
- Department of Emergency Medicine, Department of Emergency Medicine and ServicesHelsinki University Hospital, University of HelsinkiHelsinkiFinland
| | - Benjamin Deniau
- Department of Anesthesiology, Critical Care and Burn UnitUniversity Hospitals Saint‐Louis—Lariboisière, AP‐HP, FHU PROMICEParisFrance
| | - Mari Hongisto
- Department of Emergency Medicine, Department of Emergency Medicine and ServicesHelsinki University Hospital, University of HelsinkiHelsinkiFinland
| | - Ewa A. Jankowska
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland, Institute of Heart Diseases, University Hospital in Wroclaw, Wroclaw, Poland Internal MedicineHelsinki University HospitalHelsinkiFinland
| | - Raija Jurkko
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| | - Toni Jäntti
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| | - Anu Kataja
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland, Institute of Heart Diseases, University Hospital in Wroclaw, Wroclaw, Poland Internal MedicineHelsinki University HospitalHelsinkiFinland
| | - Alexandre Mebazaa
- Department of Anesthesia & Critical CareUniversité Paris Cité, APHP, Inserm MASCOT, FHU PROMICEParisFrance
| | - Tuija Sabell
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| | - Alessandro Sionis
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain and Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Johan Lassus
- Department of Cardiology, Heart and Lung CenterHelsinki University HospitalHelsinkiFinland
| |
Collapse
|
2
|
Ataman S, Girişgin AS, Ayranci MK, Küçükceran K. Relationship of serum phosphorus level with acute cerebrovascular disease. Sci Rep 2025; 15:16229. [PMID: 40346077 PMCID: PMC12064689 DOI: 10.1038/s41598-025-00704-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
There are no sufficient studies on laboratory parameters associated with acute stroke. Laboratory parameters associated with acute stroke may become important in the prevention or follow-up of the disease. To investigate the relationship between cerebrovascular disease (CVD) and serum phosphorus levels. This study was conducted prospectively to evaluate the relationship between cerebrovascular disease and phosphorus in patients who applied to the Emergency Medicine Clinic of the Faculty of Medicine Hospital and underwent central imaging between March 1, 2023, and December 31, 2023. 299 patients who applied to the university emergency department were prospectively included in the study with brain computed tomography (CT) and diffusion magnetic resonance imaging (MRI). The lactate, white blood cell, hgb, neutrophil, lymphocyte, urea, creatine and phosphorus values of the patients at their first admission were recorded. The patients were divided into two groups: CVD group and control group. The laboratory parameters of the patients were compared according to the groups. In the ROC analysis performed to evaluate the power of laboratory parameters in predicting the diagnosis of CVD, the AUC value of phosphorus was obtained as 0.935, lactate as 0.620 and urea as 0.596. The optimum cut-off value for phosphorus was calculated as 3.47 (mg/dl) and the sensitivity of this cut-off value was found to be 89%, specifically 86.9%, positive predictive value (ppv) as 77.4% and negative predictive value (npv) as 94%. Lactate, urea and phosphorus values were found to be significantly higher in the CVD group compared to the control group. With this study, we found that serum phosphorus levels were higher than our cut-off value in patients with acute ischemic CVD. We believe that serum phosphorus level will be useful in excluding acute cerebrovascular disease due to its high negative predictive value when evaluated according to the cut-off value of 3.47 (mg/dl).
Collapse
Affiliation(s)
- Sami Ataman
- Emergency Department, Muradiye State Hospital, Van, Turkey.
| | | | - Mustafa Kürşat Ayranci
- Emergency Department, Meram School of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Kadir Küçükceran
- Emergency Department, Meram School of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
3
|
Grimmett ZW, Zhang R, Zhou HL, Chen Q, Miller D, Qian Z, Lin J, Kalra R, Gross SS, Koch WJ, Premont RT, Stamler JS. The denitrosylase SCoR2 controls cardioprotective metabolic reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642752. [PMID: 40161620 PMCID: PMC11952481 DOI: 10.1101/2025.03.12.642752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Acute myocardial infarction (MI) is a leading cause of morbidity and mortality, and therapeutic options remain limited. Endogenously generated nitric oxide (NO) is highly cardioprotective, but protection is not replicated by nitroso-vasodilators (e.g., nitrates, nitroprusside) used in clinical practice, highlighting specificity in NO-based signaling and untapped therapeutic potential. Signaling by NO is mediated largely by S-nitrosylation, entailing specific enzymes that form and degrade S-nitrosothiols in proteins (SNO-proteins), termed nitrosylases and denitrosylases, respectively. SNO-CoA Reductase 2 (SCoR2; product of the Akr1a1 gene) is a recently discovered protein denitrosylase. Genetic variants in SCoR2 have been associated with cardiovascular disease, but its function is unknown. Here we show that mice lacking SCoR2 exhibit robust protection in an animal model of MI. SCoR2 regulates ketolytic energy availability, antioxidant levels and polyol homeostasis via S-nitrosylation of key metabolic effectors. Human cardiomyopathy shows reduced SCoR2 expression and an S-nitrosylation signature of metabolic reprogramming, mirroring SCoR2-/- mice. Deletion of SCoR2 thus coordinately reprograms multiple metabolic pathways-ketone body utilization, glycolysis, pentose phosphate shunt and polyol metabolism-to limit infarct size, establishing SCoR2 as a novel regulator in the injured myocardium and a potential drug target. Impact statement Mice lacking the denitrosylase enzyme SCoR2/AKR1A1 demonstrate robust cardioprotection resulting from reprogramming of multiple metabolic pathways, revealing widespread, coordinated metabolic regulation by SCoR2.
Collapse
Affiliation(s)
- Zachary W. Grimmett
- Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland OH, 44106
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Rongli Zhang
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Hua-Lin Zhou
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065
| | - Dawson Miller
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065
| | - Zhaoxia Qian
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Justin Lin
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Riti Kalra
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Steven S. Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065
| | - Walter J. Koch
- Department of Surgery, Duke University School of Medicine, Durham NC, 27710
- Department of Medicine, Duke University School of Medicine, Durham NC, 27710
| | - Richard T. Premont
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland OH, 44106
| | - Jonathan S. Stamler
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland OH, 44106
| |
Collapse
|
4
|
Wang Y, Wu X, Liu Y, Zhang J, Wang L, Luo X. A wearable platform for biochemical sweat analysis using photonic crystal hydrogel. Anal Chim Acta 2025; 1338:343590. [PMID: 39832860 DOI: 10.1016/j.aca.2024.343590] [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: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Wearable systems for health monitoring are highly desired in personal diagnostics and precision medicine while challenges remain in constructing such wearable systems with reliability and high performance. Herein, we report a wearable platform for non-invasive monitoring biomarkers in sweat. The device is composed of a butterfly-shaped like microfluidic platform in which responsive photonic crystal hydrogels are embedded in each butterfly wing as sensors. Sweat lactate concentration and pH can be obtained via the color variation of the sensor with naked eyes. Accurate quantitative analysis of the two target analytes can be obtained via the image analysis on a mobile phone by integrating the linear range response to analytes within the physiological range. Moreover, the sensor exhibits good reproducibility, excellent selectivity and long-term stability. On-body trials have been conducted by attaching the device on volunteers' body, and the obtained results are consistent with those analyzed through standardized methods, demonstrating its potentiality in real-time and continuous monitoring sweat biomarkers. This non-invasive wearable sensor provides a new strategy for personal health monitoring and sports performance assessment.
Collapse
Affiliation(s)
- Yingli Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Xiao Wu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Yun Liu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Jincheng Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China
| | - Lei Wang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| | - Xiliang Luo
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, PR China.
| |
Collapse
|
5
|
Ocariza MGC, Paton LN, Templeton EM, Pemberton CJ, Pilbrow AP, Appleby S. CNDP2: An Enzyme Linking Metabolism and Cardiovascular Diseases? J Cardiovasc Transl Res 2025; 18:48-57. [PMID: 39349903 PMCID: PMC11885389 DOI: 10.1007/s12265-024-10560-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/12/2024] [Indexed: 03/09/2025]
Abstract
The heart requires a substantial amount of energy to function, utilising various substrates including lipids, glucose and lactate as energy sources. In times of increased stress, lactate becomes the primary energy source of the heart, but persistently elevated lactate levels are linked to poor patient outcomes and increased mortality. Recently, carnosine dipeptidase II (CNDP2) was discovered to catalyse the formation of Lac-Phe, an exercise-induced metabolite derived from lactate, which has been shown to suppress appetite in mice and reduce adipose tissue in humans. This review discusses CNDP2, including its role in lactate clearance, carnosine hydrolysis, oxidative stress regulation, and involvement in metabolite regulation. The association between CNDP2 and cardiometabolic and renal diseases is also explored, and knowledge gaps are highlighted. CNDP2 appears to be a complex participant in human physiological processes and disease, necessitating additional research to unveil its functions and potential therapeutic applications.
Collapse
Affiliation(s)
- Moizle Grace Castro Ocariza
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand.
| | - Louise Nancy Paton
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| | - Evelyn Mary Templeton
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| | - Christopher Joseph Pemberton
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| | - Anna Pauline Pilbrow
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| | - Sarah Appleby
- Department of Medicine, Christchurch Heart Institute, University of Otago (Christchurch), Christchurch, New Zealand
| |
Collapse
|
6
|
Zhang L, Zhao X, Wang Z, Deng H, Zhang X, Wang X, Lao J, Gao M, Hou Y, Han Y. Preadmission metformin use increased the incidence of hyperlactatemia at admission and 30-day in-hospital mortality among T2D patients with heart disease at high risk of hypoxia. Int J Cardiol 2024; 412:132338. [PMID: 38964551 DOI: 10.1016/j.ijcard.2024.132338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Surprisingly, despite the high prevalence of metformin use in type 2 diabetes (T2D) patients with heart disease, limited safety data is available regarding metformin use in patients with acute and critical heart disease. METHODS In this single-center retrospective study, patients admitted to the cardiology department for heart failure (HF) or acute coronary syndrome (ACS) between December 2013 and December 2021 and who underwent arterial blood gas analysis at admission with an estimated glomerular clearance rate of ≥45 ml/min/1.73 m2 were identified. The incidences of hyperlactatemia, acidosis, and 30-day in-hospital mortality were compared between preadmission metformin users and nonusers. RESULTS Of 526 admissions, 193/193 metformin users/nonusers were selected in a propensity score-matched model. Metformin users had greater lactate levels (2.55 ± 2.07 mmol/l vs. 2.00 ± 1.80 mmol/l P < 0.01), a greater incidence of hyperlactatemia [odds ratio (OR) = 2.55; 95% confidence interval (CI), 1.63-3.98; P < 0.01] and acidosis (OR = 1.78; 95% CI, 1.00-3.16; P < 0.05) at admission and a greater incidence of in-hospital mortality (OR = 3.83; 95% CI, 1.05-13.94; P < 0.05), especially those with HF/acute myocardial infarction, elderly age, or without preadmission insulin use. CONCLUSIONS Our results suggest that, compared to metformin nonusers, preadmission use of metformin may be associated with a greater incidence of hyperlactatemia and acidosis at admission and greater 30-day in-hospital mortality among T2D patients with HF or ACS at high risk of hypoxia, particularly those without preadmission insulin use. The safety of metformin in this population needs to be confirmed in prospective controlled trials.
Collapse
Affiliation(s)
- Le Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, PR China
| | - Xia Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, PR China
| | - Zhongsu Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, PR China
| | - Hao Deng
- School of Pharmacy, Shandong University, Jinan, Shandong Province, PR China
| | - Xue Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, PR China
| | - Xuan Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, PR China
| | - Jiahui Lao
- Health and Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, PR China
| | - Mei Gao
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, Shandong Province, PR China
| | - Yinglong Hou
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, Shandong Province, PR China
| | - Yi Han
- Department of Clinical Pharmacy, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, Shandong Province, PR China.
| |
Collapse
|
7
|
Sato K, Heinsar S, Chan J, Farah SM, Wildi K, Obonyo NG, Liu K, Ainola C, Sato N, Abbate G, Wilson ES, Bouquet M, Hyslop K, Passmore MR, Ijuin S, Ro SK, Fior G, Gandini L, Lundon B, Platts DG, Suen JY, Bassi GL, Fraser JF. A novel echocardiographic parameter considering left ventricular afterload during V-A ECMO support. Eur J Clin Invest 2024; 54:e14263. [PMID: 38849326 DOI: 10.1111/eci.14263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/19/2024] [Accepted: 05/25/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Left ventricular stroke work index (LVSWI) and cardiac power index (CPI) account for the haemodynamic load of the left ventricle and are promising prognostic values in cardiogenic shock. However, accurately and non-invasively measuring these parameters during veno-arterial extracorporeal membrane oxygenation (V-A ECMO) is challenging and potentially biased by the extracorporeal circulation. This study aimed to investigate, in an ovine model of cardiogenic shock, whether Pressure-Strain Product (PSP), a novel speckle-tracking echocardiography parameter, (1) can correlate with pressure-volume catheter-based LVSWI and CPI, and (2) can be load-independent during the flow modification of V-A ECMO. METHODS Nine Dorset-cross ewes (51 ± 4 kg) were included. After cardiogenic shock was induced, full support V-A ECMO (X L/min based on 60 mL/kg/min) commenced. At seven time points during 24-h observation, echocardiographic parameters as well as pressure-volume catheter-based LVSWI and CPI were simultaneously measured with X and following X-1 L/min of ECMO flow. PSP was calculated by multiplying global circumferential strain or global radial strain, and mean arterial pressure, for PSPcirc or PSPrad, respectively. RESULTS PSPcirc showed a stronger correlation with LVSWI (correlation coefficient, CC = .360, p < .001) and CPI (CC = .283, p < .001) than other echocardiographic parameters. The predictability of PSPcirc for pressure-volume catheter-based LVSWI (AUC .82) and CPI (AUC .80) was also higher than other echocardiographic parameters. No statistically significant differences were identified between the two ECMO flow variations in PSPcirc (p = .558). CONCLUSIONS A novel echocardiographic parameter, PSP, may non-invasively predict pressure-volume catheter-based LVSWI and CPI in a load-independent manner in a cardiogenic shock supported by V-A ECMO.
Collapse
Affiliation(s)
- Kei Sato
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Silver Heinsar
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Intensive Care Unit, St. Andrews War Memorial Hospital, Brisbane, Queensland, Australia
- Department of Intensive Care, North Estonia Medical Centre, Tallinn, Estonia
| | - Jonathan Chan
- Division of Cardiology, The Prince Charles Hospital, Brisbane, Queensland, Australia
- School of Medicine, Griffith University, Gold Coast, Queensland, Australia
| | - Samia M Farah
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Karin Wildi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Department of Intensive Care Medicine, University Hospital Basel, Basel, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Nchafatso G Obonyo
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Wellcome Trust Centre for Global Health Research, Imperial College London, London, UK
- Initiative to Develop African Research Leaders (IDeAL)/KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Keibun Liu
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Carmen Ainola
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Noriko Sato
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Gabriella Abbate
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Emily S Wilson
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Mahé Bouquet
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Kieran Hyslop
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Shinichi Ijuin
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Department of Emergency and Critical Care Medicine, Hyogo Emergency Medical Center, Kobe, Japan
| | - Sun Kyun Ro
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Department of Thoracic and Cardiovascular Surgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Gabriele Fior
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Lucia Gandini
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Brooke Lundon
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - David G Platts
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Division of Cardiology, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Intensive Care Unit, St. Andrews War Memorial Hospital, Brisbane, Queensland, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Intensive Care Unit, St. Andrews War Memorial Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
8
|
Juyal A, Bisht S, Singh MF. Smart solutions in hypertension diagnosis and management: a deep dive into artificial intelligence and modern wearables for blood pressure monitoring. Blood Press Monit 2024; 29:260-271. [PMID: 38958493 DOI: 10.1097/mbp.0000000000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Hypertension, a widespread cardiovascular issue, presents a major global health challenge. Traditional diagnosis and treatment methods involve periodic blood pressure monitoring and prescribing antihypertensive drugs. Smart technology integration in healthcare offers promising results in optimizing the diagnosis and treatment of various conditions. We investigate its role in improving hypertension diagnosis and treatment effectiveness using machine learning algorithms for early and accurate detection. Intelligent models trained on diverse datasets (encompassing physiological parameters, lifestyle factors, and genetic information) to detect subtle hypertension risk patterns. Adaptive algorithms analyze patient-specific data, optimizing treatment plans based on medication responses and lifestyle habits. This personalized approach ensures effective, minimally invasive interventions tailored to each patient. Wearables and smart sensors provide real-time health insights for proactive treatment adjustments and early complication detection.
Collapse
Affiliation(s)
- Anubhuti Juyal
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Lucknow, Uttar Pradesh
| | - Shradha Bisht
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Lucknow, Uttar Pradesh
| | - Mamta F Singh
- Department of Pharmacology, College of Pharmacy, COER University, Roorkee, Uttarakhand, India
| |
Collapse
|
9
|
Cardiel Nunez K, Hess SY, Arnold CD, Smith TJ, Trehan I, Hiffler L, Sitthideth D, Jones KS, Kounnavong S, Fischer PR. Relationship between lactate and thiamine-responsive disorders in hospitalised infants and children in Lao PDR: secondary analysis of a prospective cohort study. Paediatr Int Child Health 2024; 44:95-104. [PMID: 39511715 PMCID: PMC11581904 DOI: 10.1080/20469047.2024.2421624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Lactate is a by-product of thiamine-deficient cellular metabolism, and hyperlactataemia can indicate severe illness. However, little is known about the clinical significance of hyperlactataemia in thiamine deficiency disorders. AIM To describe the relationship between whole-blood lactate level and thiamine-responsive disorders (TRDs) in children with signs/symptoms of thiamine deficiency in a high-risk region. METHODS This is a secondary analysis of data from the Lao Thiamine study, a prospective cohort study which enrolled hospitalised infants and children (aged 21 days to <18 months) who had at least one sign or symptom suggestive of thiamine deficiency in Lao PDR. Therapeutic thiamine was administered, and clinical evaluations were completed at several time-points over the next 72 h. Three paediatricians reviewed individual case reports to evaluate clinical response to thiamine and assigned TRD status. Data from 402 children were analysed by logistic regression and predictive modelling to examine the relationship between hyperlactataemia and TRDs. RESULTS Baseline hyperlactataemia (lactate >4.0 mmol/L) was associated with an increased odds of clinical improvement after thiamine administration [OR (95% CI) 2.32 (1.28-4.45), p = 0.007]. Baseline hyperlactataemia was a significant predictor of thiamine deficiency (thiamine diphosphate <40 nmol/L) [area under the receiver operating curve (95% CI) 0.76 (0.67-0.84), p < 0.001], and increased odds of mortality [OR (95% CI) 3.51 (1.38-8.94), p = 0.009]. CONCLUSIONS In children with signs/symptoms of thiamine deficiency, hyperlactataemia is associated with a favourable clinical response to thiamine, biochemical thiamine deficiency, and increased odds of mortality. Lactate may be useful in identifying children who might benefit from therapeutic thiamine.
Collapse
Affiliation(s)
- Kristin Cardiel Nunez
- Pediatrics Residency Program, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sonja Y. Hess
- Institute for Global Nutrition and Department of Nutrition, University of California Davis, Davis, California, USA
| | - Charles D. Arnold
- Institute for Global Nutrition and Department of Nutrition, University of California Davis, Davis, California, USA
| | - Taryn J. Smith
- Institute for Global Nutrition and Department of Nutrition, University of California Davis, Davis, California, USA
| | - Indi Trehan
- Departments of Pediatrics, Global Health, and Epidemiology, University of Washington, Seattle, Washington, USA
| | | | - Dalaphone Sitthideth
- Lao Tropical and Public Health Institute, Vientiane, Lao People’s Democratic Republic
| | - Kerry S Jones
- Nutritional Biomarker Laboratory, MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Sengchanh Kounnavong
- Lao Tropical and Public Health Institute, Vientiane, Lao People’s Democratic Republic
| | - Philip R. Fischer
- Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA; Khalifa University College of Medicine and Health Sciences, Abu Dhabi, United Arab Emirates
| |
Collapse
|
10
|
Meinarovich P, Pautova A, Zuev E, Sorokina E, Chernevskaya E, Beloborodova N. An Integrated Approach Based on Clinical Data Combined with Metabolites and Biomarkers for the Assessment of Post-Operative Complications after Cardiac Surgery. J Clin Med 2024; 13:5054. [PMID: 39274267 PMCID: PMC11395730 DOI: 10.3390/jcm13175054] [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: 07/18/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/16/2024] Open
Abstract
Background: Early diagnosis of post-operative complications is an urgent task, allowing timely prescribing of appropriate therapy and reducing the cost of patient treatment. The purpose of this study was to determine whether an integrated approach based on clinical data, along with metabolites and biomarkers, had greater predictive value than the models built on fewer data in the early diagnosis of post-operative complications after cardiac surgery. Methods: The study included patients (n = 62) admitted for planned cardiac surgery (coronary artery bypass grafting with cardiopulmonary bypass) with (n = 26) or without (n = 36) post-operative complications. Clinical and laboratory data on the first day after surgery were analyzed. Additionally, patients' blood samples were collected before and on the first day after surgery to determine biomarkers and metabolites. Results: Multivariate PLS-DA models, predicting the presence or absence of post-operative complications, were built using clinical data, concentrations of metabolites and biomarkers, and the entire data set (ROC-AUC = 0.80, 0.71, and 0.85, respectively). For comparison, we built univariate models using the EuroScore2 and SOFA scales, concentrations of lactate, the dynamic changes of 4-hydroxyphenyllactic acid, and the sum of three sepsis-associated metabolites (ROC-AUC = 0.54, 0.79, 0.62, 0.58, and 0.70, respectively). Conclusions: The proposed complex model using the entire dataset had the best characteristics, which confirms the expediency of searching for new predictive models based on a variety of factors.
Collapse
Affiliation(s)
- Peter Meinarovich
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25-2 Petrovka Str., 107031 Moscow, Russia
| | - Alisa Pautova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25-2 Petrovka Str., 107031 Moscow, Russia
| | - Evgenii Zuev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25-2 Petrovka Str., 107031 Moscow, Russia
| | - Ekaterina Sorokina
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25-2 Petrovka Str., 107031 Moscow, Russia
| | - Ekaterina Chernevskaya
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25-2 Petrovka Str., 107031 Moscow, Russia
| | - Natalia Beloborodova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 25-2 Petrovka Str., 107031 Moscow, Russia
| |
Collapse
|
11
|
Billig H, Al Zaidi M, Quacken F, Görtzen-Patin J, Goody PR, Gräff I, Nickenig G, Zimmer S, Aksoy A. Blood glucose and lactate levels as early predictive markers in patients presenting with cardiogenic shock: A retrospective cohort study. PLoS One 2024; 19:e0306107. [PMID: 39052641 PMCID: PMC11271948 DOI: 10.1371/journal.pone.0306107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/11/2024] [Indexed: 07/27/2024] Open
Abstract
Lactate and glucose are widely used biochemical parameters in current predictive risk scores for cardiogenic shock. Data regarding the relationship between lactate and glucose levels in cardiogenic shock are limited. Thus, we aimed to analyze glucose and lactate as early markers for in-hospital mortality in cardiogenic shock. In this retrospective cohort study, 312 patients presenting with cardiogenic shock to a tertiary-care hospital between 2016 and 2018 were included. Apparent cardiogenic shock was defined as hypoperfusion with hemodynamic compromise and biochemical marker increase due to diminished tissue perfusion, corresponding to SCAI shock stages. In-hospital mortality was assessed as the primary endpoint. The median age of the study population was 71 (60-79) years and the etiology of cardiogenic shock was acute myocardial infarction in 45.8%. Overall in-hospital mortality was 67.6%. In the receiver operating curve analysis, the area under the receiver-operating curve (AUC) for prediction of in-hospital mortality was higher for lactate (AUC: 0.757) than for glucose (AUC: 0.652). Both values were significantly associated with outcome (groups created with best cutoff values obtained from the Youden index). Correlation analysis showed a significant non-linear association of both values. In a multivariable stepwise Cox regression analysis, lactate remained an independent predictor for in-hospital mortality, whilst glucose, despite being implicated in energy metabolism, was not independently predictive for mortality. Together, these data suggest that lactate at admission is superior for mortality prediction in patients with apparent cardiogenic shock. Glucose was not independently predictive for mortality.
Collapse
Affiliation(s)
- Hannah Billig
- Department of Cardiology—University Hospital Bonn, Bonn, Germany
| | | | - Florian Quacken
- Department of Cardiology—University Hospital Bonn, Bonn, Germany
| | | | | | - Ingo Gräff
- Department of clinical acute- and emergency medicine, University Hospital Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Cardiology—University Hospital Bonn, Bonn, Germany
| | - Sebastian Zimmer
- Department of Cardiology—University Hospital Bonn, Bonn, Germany
| | - Adem Aksoy
- Department of Cardiology—University Hospital Bonn, Bonn, Germany
| |
Collapse
|
12
|
Janssen J, Chirico N, Ainsworth MJ, Cedillo-Servin G, Viola M, Dokter I, Vermonden T, Doevendans PA, Serra M, Voets IK, Malda J, Castilho M, van Laake LW, Sluijter JPG, Sampaio-Pinto V, van Mil A. Hypothermic and cryogenic preservation of cardiac tissue-engineered constructs. Biomater Sci 2024; 12:3866-3881. [PMID: 38910521 PMCID: PMC11265564 DOI: 10.1039/d3bm01908j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/15/2024] [Indexed: 06/25/2024]
Abstract
Cardiac tissue engineering (cTE) has already advanced towards the first clinical trials, investigating safety and feasibility of cTE construct transplantation in failing hearts. However, the lack of well-established preservation methods poses a hindrance to further scalability, commercialization, and transportation, thereby reducing their clinical implementation. In this study, hypothermic preservation (4 °C) and two methods for cryopreservation (i.e., a slow and fast cooling approach to -196 °C and -150 °C, respectively) were investigated as potential solutions to extend the cTE construct implantation window. The cTE model used consisted of human induced pluripotent stem cell-derived cardiomyocytes and human cardiac fibroblasts embedded in a natural-derived hydrogel and supported by a polymeric melt electrowritten hexagonal scaffold. Constructs, composed of cardiomyocytes of different maturity, were preserved for three days, using several commercially available preservation protocols and solutions. Cardiomyocyte viability, function (beat rate and calcium handling), and metabolic activity were investigated after rewarming. Our observations show that cardiomyocytes' age did not influence post-rewarming viability, however, it influenced construct function. Hypothermic preservation with HypoThermosol® ensured cardiomyocyte viability and function. Furthermore, fast freezing outperformed slow freezing, but both viability and function were severely reduced after rewarming. In conclusion, whereas long-term preservation remains a challenge, hypothermic preservation with HypoThermosol® represents a promising solution for cTE construct short-term preservation and potential transportation, aiding in off-the-shelf availability, ultimately increasing their clinical applicability.
Collapse
Affiliation(s)
- Jasmijn Janssen
- Department of Cardiology, Experimental Cardiology Laboratory, Circulatory Health Research Center, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands.
| | - Nino Chirico
- Department of Cardiology, Experimental Cardiology Laboratory, Circulatory Health Research Center, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands.
| | - Madison J Ainsworth
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Gerardo Cedillo-Servin
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Martina Viola
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
- Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands
| | - Inge Dokter
- Department of Cardiology, Experimental Cardiology Laboratory, Circulatory Health Research Center, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands.
| | - Tina Vermonden
- Department of Pharmaceutical Sciences, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3508 TB Utrecht, The Netherlands
| | - Pieter A Doevendans
- Netherlands Heart Institute (NLHI), Utrecht, 3511 EP, The Netherlands
- Centraal Militair Hospitaal (CMH), Utrecht, 3584 EZ, The Netherlands
| | - Margarida Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ilja K Voets
- Laboratory of Self-Organizing Soft Matter, Department of Chemical Engineering and Chemistry & Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven 5600 MB, PO box 513, The Netherlands
| | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
- Department of Equine Sciences, Faculty of Veterinary Sciences, Utrecht University, Yalelaan 1, Utrecht, 3584 CL, The Netherlands
| | - Miguel Castilho
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, 5612 AE, The Netherlands
| | - Linda W van Laake
- Department of Cardiology, Experimental Cardiology Laboratory, Circulatory Health Research Center, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands.
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, Circulatory Health Research Center, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands.
| | - Vasco Sampaio-Pinto
- Department of Cardiology, Experimental Cardiology Laboratory, Circulatory Health Research Center, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands.
| | - Alain van Mil
- Department of Cardiology, Experimental Cardiology Laboratory, Circulatory Health Research Center, Regenerative Medicine Center Utrecht, University Utrecht, University Medical Center Utrecht, Uppsalalaan 8, Utrecht, 3584 CT, The Netherlands.
| |
Collapse
|
13
|
Kwon SB, Weinerman B, Nametz D, Megjhani M, Lee I, Habib A, Barry O, Park S. Non-invasive pulse arrival time is associated with cardiac index in pediatric heart transplant patients with normal ejection fraction. Physiol Meas 2024; 45:07NT01. [PMID: 38986482 PMCID: PMC11262133 DOI: 10.1088/1361-6579/ad61b9] [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: 01/03/2024] [Revised: 06/05/2024] [Accepted: 07/10/2024] [Indexed: 07/12/2024]
Abstract
Objective.Cardiac Index (CI) is a key physiologic parameter to ensure end organ perfusion in the pediatric intensive care unit (PICU). Determination of CI requires invasive cardiac measurements and is not routinely done at the PICU bedside. To date, there is no gold standard non-invasive means to determine CI. This study aims to use a novel non-invasive methodology, based on routine continuous physiologic data, called Pulse Arrival Time (PAT) as a surrogate for CI in patients with normal Ejection Fraction (EF).Approach.Electrocardiogram (ECG) and photoplethysmogram (PPG) signals were collected from beside monitors at a sampling frequency of 250 samples per second. Continuous PAT, derived from the ECG and PPG waveforms was averaged per patient. Pearson's correlation coefficient was calculated between PAT and CI, PAT and heart rate (HR), and PAT and EF.Main Results.Twenty patients underwent right heart cardiac catheterization. The mean age of patients was 11.7 ± 5.4 years old, ranging from 11 months old to 19 years old, the median age was 13.4 years old. HR in this cohort was 93.8 ± 17.0 beats per minute. The average EF was 54.4 ± 9.6%. The average CI was 3.51 ± 0.72 l min-1m-2, with ranging from 2.6 to 4.77 l min-1m-2. The average PAT was 0.31 ± 0.12 s. Pearson correlation analysis showed a positive correlation between PAT and CI (0.57,p< 0.01). Pearson correlation between HR and CI, and correlation between EF and CI was 0.22 (p= 0.35) and 0.03 (p= 0.23) respectively. The correlation between PAT, when indexed by HR (i.e. PAT × HR), and CI minimally improved to 0.58 (p< 0.01).Significance.This pilot study demonstrates that PAT may serve as a valuable surrogate marker for CI at the bedside, as a non-invasive and continuous modality in the PICU. The use of PAT in clinical practice remains to be thoroughly investigated.
Collapse
Affiliation(s)
- Soon Bin Kwon
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
- Program in Hospital and Intensive Care Informatics, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
| | - Bennett Weinerman
- Program in Hospital and Intensive Care Informatics, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
- Columbia University College of Physicians and Surgeons, Division of Pediatric Critical Care and Hospital Medicine, New York, NY, United States of America
| | - Daniel Nametz
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
- Program in Hospital and Intensive Care Informatics, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
| | - Murad Megjhani
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
- Program in Hospital and Intensive Care Informatics, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
| | - Isaac Lee
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
- Program in Hospital and Intensive Care Informatics, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
| | - Anthony Habib
- Columbia University College of Physicians and Surgeons, Division of Pediatric Anesthesiology, New York, NY, United States of America
| | - Oliver Barry
- Columbia University College of Physicians and Surgeons, Division of Pediatric Cardiology, New York, NY, United States of America
| | - Soojin Park
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
- Program in Hospital and Intensive Care Informatics, Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States of America
- NewYork-Presbyterian Hospital, Columbia University Irving Medical Center, New York, NY, United States of America
- Department of Biomedical Informatics, Columbia University, New York, NY, United States of America
| |
Collapse
|
14
|
Wu X, Yuan L, Xu J, Qi J, Zheng K. Normalized lactate load as an independent prognostic indicator in patients with cardiogenic shock. BMC Cardiovasc Disord 2024; 24:348. [PMID: 38987706 PMCID: PMC11234684 DOI: 10.1186/s12872-024-04013-8] [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: 11/29/2023] [Accepted: 06/26/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Early prognosis evaluation is crucial for decision-making in cardiogenic shock (CS) patients. Dynamic lactate assessment, for example, normalized lactate load, has been a better prognosis predictor than single lactate value in septic shock. Our objective was to investigate the correlation between normalized lactate load and in-hospital mortality in patients with CS. METHODS Data were extracted from the Medical Information Mart for Intensive Care (MIMIC)-IV database. The calculation of lactate load involved the determination of the cumulative area under the lactate curve, while normalized lactate load was computed by dividing the lactate load by the corresponding period. Receiver Operating Characteristic (ROC) curves were constructed, and the evaluation of areas under the curves (AUC) for various parameters was performed using the DeLong test. RESULTS Our study involved a cohort of 1932 CS patients, with 687 individuals (36.1%) experiencing mortality during their hospitalization. The AUC for normalized lactate load demonstrated significant superiority compared to the first lactate (0.675 vs. 0.646, P < 0.001), maximum lactate (0.675 vs. 0.651, P < 0.001), and mean lactate (0.675 vs. 0.669, P = 0.003). Notably, the AUC for normalized lactate load showed comparability to that of the Sequential Organ Failure Assessment (SOFA) score (0.675 vs. 0.695, P = 0.175). CONCLUSION The normalized lactate load was an independently associated with the in-hospital mortality among CS patients.
Collapse
Affiliation(s)
- Xia Wu
- Emergency Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Lin Yuan
- Emergency Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing, 100029, China.
| | - Jiarui Xu
- Emergency Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Jing Qi
- Emergency Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, No.2 Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Keyang Zheng
- Centre of Hypertension, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| |
Collapse
|
15
|
Biancari F, Kaserer A, Perrotti A, Ruggieri VG, Cho SM, Kang JK, Dalén M, Welp H, Jónsson K, Ragnarsson S, Hernández Pérez FJ, Gatti G, Alkhamees K, Loforte A, Lechiancole A, Rosato S, Spadaccio C, Pettinari M, Mariscalco G, Mäkikallio T, Sahli SD, L'Acqua C, Arafat AA, Albabtain MA, AlBarak MM, Laimoud M, Djordjevic I, Krasivskyi I, Samalavicius R, Puodziukaite L, Alonso-Fernandez-Gatta M, Spahn DR, Fiore A. Hyperlactatemia and poor outcome After postcardiotomy veno-arterial extracorporeal membrane oxygenation: An individual patient data meta-Analysis. Perfusion 2024; 39:956-965. [PMID: 37066850 DOI: 10.1177/02676591231170978] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
INTRODUCTION Postcardiotomy veno-arterial extracorporeal membrane oxygenation (V-A-ECMO) is associated with significant mortality. Identification of patients at very high risk for death is elusive and the decision to initiate V-A-ECMO is based on clinical judgment. The prognostic impact of pre-V-A-ECMO arterial lactate level in these critically ill patients has been herein evaluated. METHODS A systematic review was conducted to identify studies on postcardiotomy VA-ECMO for the present individual patient data meta-analysis. RESULTS Overall, 1269 patients selected from 10 studies were included in this analysis. Arterial lactate level at V-A-ECMO initiation was increased in patients who died during the index hospitalization compared to those who survived (9.3 vs 6.6 mmol/L, p < 0.0001). Accordingly, in hospital mortality increased along quintiles of pre-V-A-ECMO arterial lactate level (quintiles: 1, 54.9%; 2, 54.9%; 3, 67.3%; 4, 74.2%; 5, 82.2%, p < 0.0001). The best cut-off for arterial lactate was 6.8 mmol/L (in-hospital mortality, 76.7% vs. 55.7%, p < 0.0001). Multivariable multilevel mixed-effect logistic regression model including arterial lactate level significantly increased the area under the receiver operating characteristics curve (0.731, 95% CI 0.702-0.760 vs 0.679, 95% CI 0.648-0.711, DeLong test p < 0.0001). Classification and regression tree analysis showed the in-hospital mortality was 85.2% in patients aged more than 70 years with pre-V-A-ECMO arterial lactate level ≥6.8 mmol/L. CONCLUSIONS Among patients requiring postcardiotomy V-A-ECMO, hyperlactatemia was associated with a marked increase of in-hospital mortality. Arterial lactate may be useful in guiding the decision-making process and the timing of initiation of postcardiotomy V-A-ECMO.
Collapse
Affiliation(s)
- Fausto Biancari
- Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
- Department of Medicine, South-Karelia Central Hospital, University of Helsinki, Lappeenranta, Finland
| | - Alexander Kaserer
- Institute of Anesthesiology, University and University Hospital Zurich, Zurich, Switzerland
| | - Andrea Perrotti
- Department of Thoracic and Cardio-Vascular Surgery, University Hospital Jean Minjoz, Besançon, France
| | - Vito G Ruggieri
- Division of Cardiothoracic and Vascular Surgery, Robert Debré University Hospital, Reims, France
| | - Sung-Min Cho
- Departments of Neurology, Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jin Kook Kang
- Departments of Neurology, Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Magnus Dalén
- Department of Molecular Medicine and Surgery, and Cardiac Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Henryk Welp
- Department of Cardiothoracic Surgery, Münster University Hospital, Münster, Germany
| | - Kristján Jónsson
- Department of Cardiac Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | | - Giuseppe Gatti
- Cardio-Thoracic and Vascular Department, University Hospital of Trieste, Trieste, Italy
| | | | - Antonio Loforte
- Department of Cardiothoracic, Transplantation and Vascular Surgery, S. Orsola Hospital, Bologna, Italy
| | | | - Stefano Rosato
- Center for Global Health, Italian National Institute, Rome, Italy
| | | | - Matteo Pettinari
- Department of Cardiovascular Surgery, Ziekenhuis Oost-Limburg, 3600 Genk, Belgium
| | - Giovanni Mariscalco
- Department of Intensive Care Medicine and Cardiac Surgery, Glenfield Hospital, Leicester, UK
| | - Timo Mäkikallio
- Department of Medicine, South-Karelia Central Hospital, University of Helsinki, Lappeenranta, Finland
| | - Sebastian D Sahli
- Institute of Anesthesiology, University and University Hospital Zurich, Zurich, Switzerland
| | - Camilla L'Acqua
- Anesthesia and Intensive Care Unit, Centro Cardiologico Monzino, Italy
- Anesthesia and Intensive Care Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Italy
| | - Amr A Arafat
- Adult Cardiac Surgery, Prince Sultan Cardiac Center, Riyadh, Saudi Arabia
- Cardiothoracic Surgery Department, Tanta University, Tanta, Egypt
| | - Monirah A Albabtain
- Cardiology Clinical Pharmacy, Prince Sultan Cardiac Center, Riyadh, Saudi Arabia
| | - Mohammed M AlBarak
- Intensive Care Department, Prince Sultan Cardiac Center, Riyadh, Saudi Arabia
| | - Mohamed Laimoud
- Cardiac Surgical Intensive Care Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Critical Care Medicine Department, Cairo University, Cairo, Egypt
| | - Ilija Djordjevic
- Department of Cardiothoracic Surgery, University Hospital Cologne, Cologne, Germany
| | - Ihor Krasivskyi
- Department of Cardiothoracic Surgery, University Hospital Cologne, Cologne, Germany
| | - Robertas Samalavicius
- 2nd Department of Anesthesia, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Clinic of Emergency Medicine, Medical Faculty, Vilnius University, Vilnius, Lithuania
| | - Lina Puodziukaite
- 2nd Department of Anesthesia, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Marta Alonso-Fernandez-Gatta
- Cardiology Department, University Hospital of Salamanca, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
- CIBER-CV Instituto de Salud Carlos III, Madrid, Spain
| | - Donat R Spahn
- Institute of Anesthesiology, University and University Hospital Zurich, Zurich, Switzerland
| | - Antonio Fiore
- Department of Cardiac Surgery, Hôpitaux Universitaires Henri Mondor, Creteil, France
| |
Collapse
|
16
|
Badmus OO, da Silva AA, Li X, Taylor LC, Greer JR, Wasson AR, McGowan KE, Patel PR, Stec DE. Cardiac lipotoxicity and fibrosis underlie impaired contractility in a mouse model of metabolic dysfunction-associated steatotic liver disease. FASEB Bioadv 2024; 6:131-142. [PMID: 38706754 PMCID: PMC11069051 DOI: 10.1096/fba.2023-00139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 05/07/2024] Open
Abstract
The leading cause of death among patients with metabolic dysfunction-associated steatotic liver disease (MASLD) is cardiovascular disease. A significant percentage of MASLD patients develop heart failure driven by functional and structural alterations in the heart. Previously, we observed cardiac dysfunction in hepatocyte-specific peroxisome proliferator-activated receptor alpha knockout (Ppara HepKO), a mouse model that exhibits hepatic steatosis independent of obesity and insulin resistance. The goal of the present study was to determine mechanisms that underlie hepatic steatosis-induced cardiac dysfunction in Ppara HepKO mice. Experiments were performed in 30-week-old Ppara HepKO and littermate control mice fed regular chow. We observed decreased cardiomyocyte contractility (0.17 ± 0.02 vs. 0.24 ± 0.02 μm, p < 0.05), increased cardiac triglyceride content (0.96 ± 0.13 vs. 0.68 ± 0.06 mM, p < 0.05), collagen type 1 (4.65 ± 0.25 vs. 0.31 ± 0.01 AU, p < 0.001), and collagen type 3 deposition (1.32 ± 0.46 vs. 0.05 ± 0.03 AU, p < 0.05). These changes were associated with increased apoptosis as indicated by terminal deoxynucleotidyl transferase dUTP nick end labeling staining (30.9 ± 4.7 vs. 13.1 ± 0.8%, p < 0.006) and western blots showing increased cleaved caspase-3 (0.27 ± 0.006 vs. 0.08 ± 0.01 AU, p < 0.003) and pro-caspase-3 (5.4 ± 1.5 vs. 0.5 ± 0.3 AU, p < 0.02), B-cell lymphoma protein 2-associated X (0.68 ± 0.07 vs. 0.04 ± 0.04 AU, p < 0.001), and reduced B-cell lymphoma protein 2 (0.29 ± 0.01 vs. 1.47 ± 0.54 AU, p < 0.05). We further observed elevated circulating natriuretic peptides and exercise intolerance in Ppara HepKO mice when compared to controls. Our data demonstrated that lipotoxicity, and fibrosis underlie cardiac dysfunction in MASLD.
Collapse
Affiliation(s)
- Olufunto O. Badmus
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Alexandre A. da Silva
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Xuan Li
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Lucy C. Taylor
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Jennifer R. Greer
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Andrew R. Wasson
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Karis E. McGowan
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Parth R. Patel
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - David E. Stec
- Department of Physiology & Biophysics, Cardiorenal, and Metabolic Diseases Research Center, Cardiovascular‐Renal Research CenterUniversity of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
17
|
Yang DE, Jo S, Lee DH, An WS, Jeong MJ, Son M. Dynamics of serum anion gaps with in-hospital mortality: Analysis of the multi-open databases. PLoS One 2024; 19:e0302206. [PMID: 38625899 PMCID: PMC11020621 DOI: 10.1371/journal.pone.0302206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/31/2024] [Indexed: 04/18/2024] Open
Abstract
BACKGROUND Few studies have investigated the relationship between the anion gap, including the corrected anion gap, and patient mortality in intensive care units (ICUs) without restricting the analysis to specific diseases or medical specialties. Our primary objective was to investigate the association between the anion gap and ICU mortality using multiple open-access databases. METHODS We identified 4229 subjects from the Medical Information Mart for Intensive Care IV (MIMIC-IV) database, whose entries were from between 2008 and 2019. For each patient, the anion gap and corrected anion gap were calculated, and the study sample was divided into tertile groups (T) according to these levels. The association between the anion gap and in-hospital mortality was assessed using hazard ratios (HRs) and 95% confidence intervals (CIs) derived from a multivariable-adjusted Cox proportional hazards model. Besides MIMIC-IV, we also incorporated study samples from two other databases (MIMIC-III and electronic ICU) to calculate summary HRs using a random-effects meta-analysis. RESULTS Within MIMIC-IV, 1015 patients (24%) died during an average follow-up period of 15.5 days. The fully adjusted HRs and 95% CIs for T2 and T3, relative to T1, were 1.31 (95% CI 1.08-1.58) and 1.54 (95% CI 1.24-1.90), respectively. When grouped by corrected anion gap, the results remained statistically significant. In the meta-analysis, the summary HRs and 95% CIs for T2 and T3 were 1.24 (95% CI 1.08-1.43) and 1.55 (95% CI 1.33-1.82), respectively. CONCLUSIONS Both the anion gap and corrected anion gap were associated with in-hospital mortality regardless of specific diseases or medical specialties.
Collapse
Affiliation(s)
- Dong Eun Yang
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Sua Jo
- Department of Hospital Medicine, Inha University Hospital, Incheon, Republic of Korea
| | - Dong Hyun Lee
- Department of Pulmonology and Intensive Care Medicine, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Won Suk An
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Min Jae Jeong
- Department of Physiology, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Minkook Son
- Department of Physiology, Dong-A University College of Medicine, Busan, Republic of Korea
- Department of Data Sciences Convergence, Dong-A University Interdisciplinary Program, Busan, Republic of Korea
| |
Collapse
|
18
|
Orlowska MK, Krycer JR, Reid JD, Mills RJ, Doran MR, Hudson JE. A miniaturized culture platform for control of the metabolic environment. BIOMICROFLUIDICS 2024; 18:024101. [PMID: 38434908 PMCID: PMC10908563 DOI: 10.1063/5.0169143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024]
Abstract
The heart is a metabolic "omnivore" and adjusts its energy source depending on the circulating metabolites. Human cardiac organoids, a three-dimensional in vitro model of the heart wall, are a useful tool to study cardiac physiology and pathology. However, cardiac tissue naturally experiences shear stress and nutrient fluctuations via blood flow in vivo, whilst in vitro models are conventionally cultivated in a static medium. This necessitates the regular refreshing of culture media, which creates acute cellular disturbances and large metabolic fluxes. To culture human cardiac organoids in a more physiological manner, we have developed a perfused bioreactor for cultures in a 96-well plate format. The designed bioreactor is easy to fabricate using a common culture plate and a 3D printer. Its open system allows for the use of traditional molecular biology techniques, prevents flow blockage issues, and provides easy access for sampling and cell assays. We hypothesized that a perfused culture would create more stable environment improving cardiac function and maturation. We found that lactate is rapidly produced by human cardiac organoids, resulting in large fluctuations in this metabolite under static culture. Despite this, neither medium perfusion in bioreactor culture nor lactate supplementation improved cardiac function or maturation. In fact, RNA sequencing revealed little change across the transcriptome. This demonstrates that cardiac organoids are robust in response to fluctuating environmental conditions under normal physiological conditions. Together, we provide a framework for establishing an easily accessible perfusion system that can be adapted to a range of miniaturized cell culture systems.
Collapse
|
19
|
Gutgold A, Salameh S, Nashashibi J, Gershinsky Y. Prognosis of patients with extreme acidosis on admission to the emergency department: A retrospective cohort study. Am J Emerg Med 2024; 76:36-40. [PMID: 37980726 DOI: 10.1016/j.ajem.2023.10.054] [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: 04/23/2023] [Revised: 10/03/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023] Open
Abstract
AIM OF THE STUDY The development of acidosis in critically ill patients is considered to be a negative prognostic factor, and when extreme, even incompatible with life. We aimed to test the prognosis of patients with a pH lower than 6.9 on emergency department admission. METHODS A retrospective cohort study in adult patients admitted to two emergency departments with a pH < 6.9 during the first 12 h of admission. Primary outcome was mortality within 24 h from emergency department admission. We performed a regression analysis of clinical and laboratory data in order to identify factors associated with mortality in this population. RESULTS We analyzed data of 206 admissions to the emergency departments between 2008 and 2018 with extreme acidosis. pH Values ranged from 6.898 to 6.35 (mean 6.8 and median 6.83). 60 (29%) of the patients survived the first 24 h. 35 patients (58%) of those also survived to hospital discharge, and of them 80% have returned to their previous functional status. Patient's age, type of acidosis, cardio-pulmonary resuscitation on arrival, and diagnosis on admission were correlated with survival. CONCLUSIONS A small but significant portion of patients with extreme acidosis on emergency department admission survive at least to 24 h and until hospital discharge. The clinical decision making should be based on other prognostic factors rather than pH value by itself.
Collapse
Affiliation(s)
- Amichai Gutgold
- Medical Intensive Care Unit, Rambam Health Care Campus, Haifa, Israel.
| | - Shaden Salameh
- Emergency Department, Hadassah Medical Center, Mount-Scopus campus, Jerusalem, Israel.
| | | | - Yonatan Gershinsky
- Emergency Department, Hadassah Medical Center, Mount-Scopus campus, Jerusalem, Israel.
| |
Collapse
|
20
|
Broskey NT, Pories WJ, DeMaria EJ, Jones TE, Tanner CJ, Zheng D, Krassovskaia PM, Mitchell LA, Matarese LE, O'Brien KF, Cortright RN, Dohm GL, Houmard JA. Fasting plasma lactate as a possible early clinical marker for metabolic disease risk. Diabetes Metab Syndr 2024; 18:102955. [PMID: 38310736 PMCID: PMC10997452 DOI: 10.1016/j.dsx.2024.102955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND AND AIM Elevated fasting plasma lactate concentrations are evident in individuals with metabolic diseases. However, it has yet to be determined if these associations exist in a young, healthy population as a possible early marker for metabolic disease risk. The purpose of this study was to determine if indices of the metabolic syndrome are related to plasma lactate concentrations in this population. METHODS Fifty (29 ± 7 yr) men (n = 19) and women (n = 31) classified as overweight (26.4 ± 1.8 kg/m2) participated in this observational study. Blood pressure and blood metabolites were measured after an overnight fast. Lactate was also measured before and after a three-day eucaloric high-fat (70 %) diet. The homeostatic model assessment for insulin resistance (HOMA-IR) was calculated as a measure of insulin resistance. Visceral adipose tissue mass was determined via dual X-ray absorptiometry. RESULTS Triglycerides (r = 0.55, p=<0.0001), HOMA-IR (r = 0.53, p=<0.0001), and systolic and diastolic (both, r = 0.36, p = 0.01) blood pressures associated with fasting plasma lactate. No differences in visceral adipose tissue existed between the sexes (p = 0.41); however, the relationship between visceral adipose tissue and lactate existed only in females (r = 0.59, p = 0.02) but not in males (p = 0.53). Fasting lactate and HOMA-IR increased in males (p = 0.01 and p = 0.02, respectively), but not females, following a three-day high-fat diet. CONCLUSION Indices of the metabolic syndrome associated with fasting plasma lactates in young relatively healthy individuals. Fasting lactate also increased in a sex-specific manner after a three-day high fat diet. Thus, lactate could become a clinical marker for metabolic disease risk.
Collapse
Affiliation(s)
- Nicholas T Broskey
- Departments of Kinesiology, East Carolina University, Greenville, NC, USA; Surgery, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA.
| | - Walter J Pories
- Surgery, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | | | - Terry E Jones
- Physical Therapy, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Charles J Tanner
- Departments of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Donghai Zheng
- Departments of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Polina M Krassovskaia
- Departments of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | | | - Laura E Matarese
- Surgery, East Carolina University, Greenville, NC, USA; Internal Medicine, East Carolina University, Greenville, NC, USA
| | - Kevin F O'Brien
- Public Health, East Carolina University, Greenville, NC, USA
| | - Ronald N Cortright
- Departments of Kinesiology, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, East Carolina University, Greenville, NC, USA; Physiology, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - G Lynis Dohm
- Physiology, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Joseph A Houmard
- Departments of Kinesiology, East Carolina University, Greenville, NC, USA; Surgery, East Carolina University, Greenville, NC, USA; Human Performance Laboratory, East Carolina University, Greenville, NC, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| |
Collapse
|
21
|
Omar R, Saliba W, Khatib M, Zheng Y, Pieters C, Oved H, Silberman E, Zohar O, Hu Z, Kloper V, Broza YY, Dvir T, Grinberg Dana A, Wang Y, Haick H. Biodegradable, Biocompatible, and Implantable Multifunctional Sensing Platform for Cardiac Monitoring. ACS Sens 2024; 9:126-138. [PMID: 38170944 PMCID: PMC10825867 DOI: 10.1021/acssensors.3c01755] [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: 08/24/2023] [Revised: 11/17/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024]
Abstract
Cardiac monitoring after heart surgeries is crucial for health maintenance and detecting postoperative complications early. However, current methods like rigid implants have limitations, as they require performing second complex surgeries for removal, increasing infection and inflammation risks, thus prompting research for improved sensing monitoring technologies. Herein, we introduce a nanosensor platform that is biodegradable, biocompatible, and integrated with multifunctions, suitable for use as implants for cardiac monitoring. The device has two electrochemical biosensors for sensing lactic acid and pH as well as a pressure sensor and a chemiresistor array for detecting volatile organic compounds. Its biocompatibility with myocytes has been tested in vitro, and its biodegradability and sensing function have been proven with ex vivo experiments using a three-dimensional (3D)-printed heart model and 3D-printed cardiac tissue patches. Moreover, an artificial intelligence-based predictive model was designed to fuse sensor data for more precise health assessment, making it a suitable candidate for clinical use. This sensing platform promises impactful applications in the realm of cardiac patient care, laying the foundation for advanced life-saving developments.
Collapse
Affiliation(s)
- Rawan Omar
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Walaa Saliba
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Muhammad Khatib
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Youbin Zheng
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Calvin Pieters
- Department
of Chemical Engineering, Technion-Israel
Institute of Technology, Haifa 320003, Israel
| | - Hadas Oved
- Shmunis
School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eric Silberman
- Shmunis
School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Orr Zohar
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Zhipeng Hu
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Viki Kloper
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Yoav Y. Broza
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Tal Dvir
- Shmunis
School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Department
Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- The
Chaoul Center for Nanoscale Systems, Tel
Aviv University Center for Nanoscience and Nanotechnology, Tel Aviv 6997801, Israel
- Sagol Center
for Regenerative Biotechnology, Tel Aviv
University, Tel Aviv 6997801, Israel
| | - Alon Grinberg Dana
- Department
of Chemical Engineering, Technion-Israel
Institute of Technology, Haifa 320003, Israel
| | - Yan Wang
- Department
of Chemical Engineering, Guangdong Technion-Israel
Institute of Technology (GTIIT), Shantou 515063, Guangdong, China
| | - Hossam Haick
- Department
of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
22
|
Biancari F, Nappi F, Gatti G, Perrotti A, Hervé A, Rosato S, D'Errigo P, Pettinari M, Peterss S, Buech J, Juvonen T, Jormalainen M, Mustonen C, Demal T, Conradi L, Pol M, Kacer P, Dell’Aquila AM, Wisniewski K, Vendramin I, Piani D, Ferrante L, Mäkikallio T, Quintana E, Pruna-Guillen R, Fiore A, Folliguet T, Mariscalco G, Acharya M, Field M, Kuduvalli M, Onorati F, Rossetti C, Gerelli S, Di Perna D, Mazzaro E, Pinto AG, Lega JR, Rinaldi M. Preoperative arterial lactate and outcome after surgery for type A aortic dissection: The ERTAAD multicenter study. Heliyon 2023; 9:e20702. [PMID: 37829811 PMCID: PMC10565766 DOI: 10.1016/j.heliyon.2023.e20702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023] Open
Abstract
Background Acute type A aortic dissection (TAAD) is associated with significant mortality and morbidity. In this study we evaluated the prognostic significance of preoperative arterial lactate concentration on the outcome after surgery for TAAD. Methods The ERTAAD registry included consecutive patients who underwent surgery for acute type A aortic dissection (TAAD) at 18 European centers of cardiac surgery. Results Data on arterial lactate concentration immediately before surgery were available in 2798 (71.7 %) patients. Preoperative concentration of arterial lactate was an independent predictor of in-hospital mortality (mean, 3.5 ± 3.2 vs 2.1 ± 1.8 mmol/L, adjusted OR 1.181, 95%CI 1.129-1.235). The best cutoff value preoperative arterial lactate concentration was 1.8 mmol/L (in-hospital mortality, 12.0 %, vs. 26.6 %, p < 0.0001). The rates of in-hospital mortality increased along increasing quintiles of arterial lactate and it was 12.1 % in the lowest quintile and 33.6 % in the highest quintile (p < 0.0001). The difference between multivariable models with and without preoperative arterial lactate was statistically significant (p = 0.0002). The NRI was 0.296 (95%CI 0.200-0.391) (p < 0.0001) with -17 % of events correctly reclassified (p = 0.0002) and 46 % of non-events correctly reclassified (p < 0.0001). The IDI was 0.025 (95%CI 0.016-0.034) (p < 0.0001). Six studies from a systematic review plus the present one provided data for a pooled analysis which showed that the mean difference of preoperative arterial lactate between 30-day/in-hospital deaths and survivors was 1.85 mmol/L (95%CI 1.22-2.47, p < 0.0001, I2 64 %). Conclusions Hyperlactatemia significantly increased the risk of mortality after surgery for acute TAAD and should be considered in the clinical assessment of these critically ill patients.
Collapse
Affiliation(s)
- Fausto Biancari
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Medicine, South-Karelia Central Hospital, University of Helsinki, Lappeenranta, Finland
| | - Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord de Saint-Denis, Paris, France
| | - Giuseppe Gatti
- Division of Cardiac Surgery, Cardio-thoracic and Vascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Andrea Perrotti
- Department of Thoracic and Cardiovascular Surgery, University of Franche-Comte, Besancon, France
| | - Amélie Hervé
- Department of Thoracic and Cardiovascular Surgery, University of Franche-Comte, Besancon, France
| | - Stefano Rosato
- Center for Global Health, National Health Institute, Rome, Italy
| | - Paola D'Errigo
- Center for Global Health, National Health Institute, Rome, Italy
| | - Matteo Pettinari
- Department of Cardiac Surgery, Ziekenhuis Oost Limburg, Genk, Belgium
| | - Sven Peterss
- LMU University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Joscha Buech
- LMU University Hospital, Ludwig Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Tatu Juvonen
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Research Unit of Surgery, Anesthesia and Critical Care, University of Oulu, Oulu, Finland
| | - Mikko Jormalainen
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Caius Mustonen
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Till Demal
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Lenard Conradi
- Department of Cardiovascular Surgery, University Heart & Vascular Center Hamburg, Hamburg, Germany
| | - Marek Pol
- Department of Cardiac Surgery, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Petr Kacer
- Department of Cardiac Surgery, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Angelo M. Dell’Aquila
- Department of Cardiothoracic Surgery, University Hospital Muenster, Muenster, Germany
| | - Konrad Wisniewski
- Department of Cardiothoracic Surgery, University Hospital Muenster, Muenster, Germany
| | - Igor Vendramin
- Cardiothoracic Department, University Hospital, Udine, Italy
| | - Daniela Piani
- Cardiothoracic Department, University Hospital, Udine, Italy
| | - Luisa Ferrante
- Cardiac Surgery, Molinette Hospital, University of Turin, Turin, Italy
| | - Timo Mäkikallio
- Department of Medicine, South-Karelia Central Hospital, University of Helsinki, Lappeenranta, Finland
| | - Eduard Quintana
- Department of Cardiovascular Surgery, Hospital Clínic de Barcelona, University of Barcelona, Spain
| | - Robert Pruna-Guillen
- Department of Cardiovascular Surgery, Hospital Clínic de Barcelona, University of Barcelona, Spain
| | - Antonio Fiore
- Department of Cardiac Surgery, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, Creteil, France
| | - Thierry Folliguet
- Department of Cardiac Surgery, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, Creteil, France
| | - Giovanni Mariscalco
- Department of Cardiac Surgery, Glenfield Hospital, Leicester, United Kingdom
| | - Metesh Acharya
- Department of Cardiac Surgery, Glenfield Hospital, Leicester, United Kingdom
| | - Mark Field
- Liverpool Centre for Cardiovascular Sciences, Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| | - Manoj Kuduvalli
- Liverpool Centre for Cardiovascular Sciences, Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| | - Francesco Onorati
- Division of Cardiac Surgery, University of Verona Medical School, Verona, Italy
| | - Cecilia Rossetti
- Division of Cardiac Surgery, University of Verona Medical School, Verona, Italy
| | | | | | - Enzo Mazzaro
- Division of Cardiac Surgery, Cardio-thoracic and Vascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Angel G. Pinto
- Cardiovascular Surgery Department, University Hospital Gregorio Marañón, Madrid, Spain
| | - Javier Rodriguez Lega
- Cardiovascular Surgery Department, University Hospital Gregorio Marañón, Madrid, Spain
| | - Mauro Rinaldi
- Cardiac Surgery, Molinette Hospital, University of Turin, Turin, Italy
| |
Collapse
|
23
|
Lenzi A, Biagini D, Ghimenti S, Vivaldi FM, Salvo P, Di Francesco F, Lomonaco T. HiSorb sorptive extraction for determining salivary short chain fatty acids and hydroxy acids in heart failure patients. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1228:123826. [PMID: 37481789 DOI: 10.1016/j.jchromb.2023.123826] [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: 04/05/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
Variations in salivary short-chain fatty acids and hydroxy acids (e.g., lactic acid, and 3-hydroxybutyric acid) levels have been suggested to reflect the dysbiosis of human gut microbiota, which represents an additional factor involved in the onset of heart failure (HF) disease. The physical-chemical properties of these metabolites combined with the complex composition of biological matrices mean that sample pre-treatment procedures are almost unavoidable. This work describes a reliable, simple, and organic solvent free protocol for determining short-chain fatty acids and hydroxy acids in stimulated saliva samples collected from heart failure, obese, and hypertensive patients. The procedure is based on in-situ pentafluorobenzyl bromide (PFB-Br) derivatization and HiSorb sorptive extraction coupled to thermal desorption and gas chromatography-tandem mass spectrometry. The HiSorb extraction device is completely compatible with aqueous matrices, thus saving on time and materials associated with organic solvent-extraction methods. A Central Composite Face-Centred experimental design was used for the optimization of the molar ratio between PFB-Br and target analytes, the derivatization temperature, and the reaction time which were 100, 60 °C, and 180 min, respectively. Detection limits in the range 0.1-100 µM were reached using a small amount of saliva (20 µL). The use of sodium acetate-1-13C as an internal standard improved the intra- and inter-day precision of the method which ranged from 10 to 23%. The optimized protocol was successfully applied for what we believe is the first time to evaluate the salivary levels of short chain fatty acids and hydroxy acids in saliva samples of four groups of patients: i) patients admitted to hospital with acute HF symptoms, ii) patients with chronic HF symptoms, iii) patients without HF symptoms but with obesity, and iv) patients without HF symptoms but with hypertension. The first group of patients showed significantly higher levels of salivary acetic acid and lactic acid at hospital admission as well as the lowest values of hexanoic acid and heptanoic acid. Moreover, the significant high levels of acetic acid, propionic acid, and butyric acid observed in HF respect to the other patients suggest the potential link between oral bacteria and gut dysbiosis.
Collapse
Affiliation(s)
- Alessio Lenzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Silvia Ghimenti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Federico M Vivaldi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Pietro Salvo
- Institute of Clinical Physiology, CNR, Via Giuseppe Moruzzi 3, Pisa, Italy
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy.
| |
Collapse
|
24
|
Kurniawati ER, Weerwind PW, Maessen JG. Understanding lactate and its clearance during extracorporeal membrane oxygenation for supporting refractory cardiogenic shock patients. BMC Cardiovasc Disord 2023; 23:305. [PMID: 37328823 PMCID: PMC10276462 DOI: 10.1186/s12872-023-03221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/30/2023] [Indexed: 06/18/2023] Open
Affiliation(s)
- E R Kurniawati
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, PO Box 5800, P. Debyelaan 25, 6202 AZ, Maastricht, the Netherlands.
| | - P W Weerwind
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, PO Box 5800, P. Debyelaan 25, 6202 AZ, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - J G Maessen
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, PO Box 5800, P. Debyelaan 25, 6202 AZ, Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
25
|
Bernhardt AM, Copeland H, Deswal A, Gluck J, Givertz MM. The International Society for Heart and Lung Transplantation/Heart Failure Society of America Guideline on Acute Mechanical Circulatory Support. J Heart Lung Transplant 2023; 42:e1-e64. [PMID: 36805198 DOI: 10.1016/j.healun.2022.10.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 02/08/2023] Open
Affiliation(s)
- Alexander M Bernhardt
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany.
| | - Hannah Copeland
- Department of Cardiac Surgery, Lutheran Health Physicians, Fort Wayne, Indiana
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason Gluck
- Heart and Vascular Institute, Hartford Hospital, Hartford, Connecticut
| | - Michael M Givertz
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
26
|
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
|
27
|
Deng X, Jiang N, Huang C, Zhou S, Peng L, Zhang L, Liu J, Wang L, Zhou J, Wang Q, Weng L, Peng J, Zhao J, Li M, Zeng X. Mortality and prognostic factors in connective tissue disease-associated pulmonary arterial hypertension patients complicated with right heart failure. Int J Rheum Dis 2023; 26:862-869. [PMID: 36892249 DOI: 10.1111/1756-185x.14660] [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: 01/09/2023] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 03/10/2023]
Abstract
OBJECTIVE To identify predictive factors associated with mortality in connective tissue disease-associated pulmonary arterial hypertension (CTD-PAH) patients who were complicated with right heart failure (RHF). METHODS In this single-center retrospective study, baseline demographics, clinical features, laboratory results, and hemodynamic assessments were collected. Kaplan-Meier analysis was applied to analyze all-cause mortality. Univariate and forward stepwise multivariate Cox proportional regression analyses were performed to identify independent predictors of mortality. RESULTS A total of 51 right heart catheterization-confirmed CTD-PAH patients complicated with RHF were consecutively enrolled in this study from 2012 to 2022. Forty-eight (94%) enrolled patients were female and the mean age was 36.0 ± 11.8 years. Thirty-two (61.5%) were systemic lupus erythematosus-PAH and 33%/67% showed World Health Organization functional class III/IV, respectively. Twenty-five (49%) of those patients died and Kaplan-Meier analysis showed the overall 1-, 3-, and 5-week survival rates from the time of hospitalization as 86.28%, 60.78%, and 56.86%, respectively. RHF in CTD-PAH patients mainly resulted from progression of PAH (n = 19) and infection (n = 5), which also contributed to the leading causes of death. Statistical analysis between survivors and non-survivors showed that death from RHF was associated with higher levels of urea (9.66 vs 6.34 mmol/L, P = 0.002), lactate (cLac: 2.65 vs 1.9 mmol/L, P = 0.006), total bilirubin (23.1 vs 16.9 μmol/L, P = 0.018) and direct bilirubin (10.5 vs 6.5 μmol/L, P = 0.004), but with lower levels of hematocrit (33.7 vs 39, P = 0.004), cNa+ (131 vs 136 mmol/L, P = 0.003). Univariate and forward stepwise multivariate Cox proportional regression analyses indicated that the level of cLac (hazards ratio:1.297; 95% CI: 1.076-1.564; P = 0.006) was an independent risk factor for mortality. CONCLUSION The short-term prognosis of CTD-PAH complicated with RHF was very poor, and hyperlactic acidemia (cLac > 2.85 mmoL/L) was an independent predicting factor for mortality of CTD-PAH patients complicated with RHF.
Collapse
Affiliation(s)
- Xiaoyue Deng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China.,Medical Science Research Center (MRC), Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Nan Jiang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Can Huang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Shuang Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Linyi Peng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Li Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jinjing Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Li Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Qian Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Li Weng
- Medical Intensive Care Unit, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinmin Peng
- Medical Intensive Care Unit, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
28
|
Early Changes in Acute Myocardial Infarction in Pigs: Achieving Early Detection with Wearable Devices. Diagnostics (Basel) 2023; 13:diagnostics13061006. [PMID: 36980316 PMCID: PMC10046897 DOI: 10.3390/diagnostics13061006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 03/09/2023] Open
Abstract
We examined the changes in variables that could be recorded on wearable devices during the early stages of acute myocardial infarction (AMI) in an animal model. Early diagnosis of AMI is important for prognosis; however, delayed diagnosis is common because of patient hesitation and lack of timely evaluations. Wearable devices are becoming increasingly sophisticated in the ability to track indicators. In this study, we retrospectively reviewed the changes in four variables during AMI in a pig model to assess their ability to help predict AMI onset. AMI was created in 33 pigs by 90-min balloon occlusion of the left anterior descending artery. Blood pressure, EKG, and lactate and cardiac troponin I levels were recorded during the occlusion period. Blood pressure declined significantly within 15 min after balloon inflation (mean arterial pressure, from 61 ± 8 to 50 ± 8 mmHg) and remained at this low level. Within 5 min of balloon inflation, the EKG showed ST-elevation in precordial leads V1–V3. Blood lactate levels increased gradually after occlusion and peaked at 60 min (from 1.48 to 2.53 mmol/L). The continuous transdermal troponin sensor demonstrated a gradual increase in troponin levels over time. Our data suggest that significant changes in key indicators (blood pressure, EKG leads V1–V3, and lactate and troponin levels) occurred at the onset of AMI. Monitoring of these variables could be used to develop an algorithm and alert patients early at the onset of AMI with the help of a wearable device.
Collapse
|
29
|
Bernhardt AM, Copeland H, Deswal A, Gluck J, Givertz MM. The International Society for Heart and Lung Transplantation/Heart Failure Society of America Guideline on Acute Mechanical Circulatory Support. J Card Fail 2023; 29:304-374. [PMID: 36754750 DOI: 10.1016/j.cardfail.2022.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Alexander M Bernhardt
- Department of Cardiovascular Surgery, University Heart and Vascular Center Hamburg, Hamburg, Germany.
| | - Hannah Copeland
- Department of Cardiac Surgery, Lutheran Health Physicians, Fort Wayne, Indiana
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason Gluck
- Heart and Vascular Institute, Hartford Hospital, Hartford, Connecticut
| | - Michael M Givertz
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
30
|
Maraziti G, Marchini L, Barbieri G, Falcone M, Corradi F, Graziani M, Ghiadoni L, Becattini C. Arterial lactate as a risk factor for death in respiratory failure related to coronavirus disease 2019: an observational study. Ther Adv Respir Dis 2023; 17:17534666231186730. [PMID: 37646253 PMCID: PMC10469239 DOI: 10.1177/17534666231186730] [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: 12/13/2022] [Accepted: 06/22/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Arterial lactate is a recognized biomarker associated with death in critically ill patients. The prognostic role of arterial lactate in acute respiratory failure due to the novel coronavirus disease 2019 (COVID-19) is unclear. OBJECTIVES We aimed to investigate the prognostic role of arterial lactate levels at admission in patients with COVID-19-related acute respiratory failure. DESIGN AND METHODS Cohorts of consecutive patients admitted to nonintensive care units (ICU) at study centers for COVID-19-related respiratory failure were merged into a collaborative database. The prognostic role of lactate levels at admission was assessed for continuous values and values ⩾2.0 mmol/l, and lactate clearance at 24 h through delta-lactate (ΔLac). The study outcome was 30-day in-hospital death. Cox proportional regression model was used to assess independent predictors of the study outcome. RESULTS At admission, 14.6% of patients had lactate levels ⩾2 mmol/l. In-hospital death at 30 days occurred in 57 out of 206 patients; 22.3% and 56.7% with normal or ⩾ 2 mmol/l lactate at admission, respectively. The median lactate level was 1.0 [interquartile range (IQR) 0.8-1.3] mmol/l and 1.3 (IQR 1.0-2.1) mmol/l in survivors and nonsurvivors, respectively (p-value < 0.001). After adjusting for age, relevant comorbidities, acidemia, and the severity of respiratory failure, lactate ⩾2.0 mmol/l was associated with in-hospital death (HR 2.53, 95% CI 1.29-4.95, p-value 0.0066), while Δ Lac ⩾0 was not (HR 1.37, 95% CI 0.42-4.49). These results were confirmed in patients with a pO2/FiO2-ratio (P/F ratio) ⩽300 mmHg. CONCLUSIONS In our study, increased arterial lactate at admission was independently associated with in-hospital death at 30 days in non-ICU patients with acute respiratory failure related to COVID-19.
Collapse
Affiliation(s)
- Giorgio Maraziti
- Internal Cardiovascular and Emergency Medicine–Stroke Unit, S. Maria della Misericordia Hospital, University of Perugia, Perugia 06156, Italy
| | - Laura Marchini
- Internal Cardiovascular and Emergency Medicine–Stroke Unit, S. Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Greta Barbieri
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
- Emergency Medicine Department, Pisa University Hospital, Italy
| | - Marco Falcone
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Corradi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
- Anaesthesia and Intensive Care Unit, Ospedali Galliera, Genova, Italy
| | - Mara Graziani
- Internal Cardiovascular and Emergency Medicine–Stroke Unit, S. Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Lorenzo Ghiadoni
- Emergency Medicine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Cecilia Becattini
- Internal Cardiovascular and Emergency Medicine–Stroke Unit, S. Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| |
Collapse
|
31
|
Wang R, Li J, Chen H, Xiao Z, Xu R, Hu Y, Chen S, Wang X, Zheng S. Preoperative albumin corrected anion gap is associated with in-hospital and long-term mortality in patients undergoing coronary artery bypass grafting in a retrospective cohort study. J Thorac Dis 2022; 14:4894-4903. [PMID: 36647463 PMCID: PMC9840039 DOI: 10.21037/jtd-22-1633] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022]
Abstract
Background Coronary artery disease remains a global health concern and the leading cause of death. Till today, coronary artery bypass grafting (CABG) is one of the main treatment strategies for coronary artery disease, especially for Multivessel coronary disease or complex coronary lesions. The present study aimed to explore the relationship of preoperative albumin corrected anion gap (ACAG) with mortality in all those patients who undergoing CABG. Methods All the patients undergoing CABG were included in the study. All clinical data were collected from CareVue and MetaVision system. The predictive value of ACAG for mortality was determined by receiver operating characteristic (ROC) curves survival curves were estimated using the Kaplan-Meier method. Multivariate regression models were constructed to determine the association of ACAG with mortality. Results A total of 2,180 patients were identified and divided into a high ACAG group (ACAG ≥16.0 mmol/L) and low ACAG group (ACAG <16.0 mmol/L) according to the ROC analysis. Patients in the high ACAG group were older and presented with more comorbidities and concomitant valvular surgeries. Further more, in the high ACAG group, we observed a higher length of stay in the intensive care unit [3.88 (2.15, 7.09) vs. 2.29 (1.29, 3.94), P<0.001]. Both the in-hospital mortality [28 (4.5%) vs. 11 (0.7%), P<0.001], and the 4-year mortality [125 (27.1%) vs. 111 (12.7%), P<0.001] were also rised in those patients. And it was also showed in the survival curves, patients with ACAG ≥16.0 mmol/L had a significant lower 4-year survival (P<0.001). While in the multivariate regression model, we found ACAG was act as an independent risk factor for both the in-hospital mortality [odds ratio (OR): 1.248 (1.060, 1.470), P=0.008] and the 4-year mortality [hazard ratio (HR): 1.134 (1.063, 1.210), P<0.001]. An ACAG ≥16.0 mmol/L was significantly associated with a 2.7-fold risk of in-hospital mortality [OR: 2.732 (1.129, 6.610), P=0.026]. Conclusions Preoperative ACAG is an independent risk factor for in-hospital and long-term mortality in CABG patients. A higher ACAG may relate to severe coronary artery stenosis and cardiac dysfunction, which is more likely to lead to a postoperative systemic inflammatory response, microcirculation disorder, and subsequent complications.
Collapse
Affiliation(s)
- Ruiling Wang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiale Li
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huimin Chen
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zezhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rongning Xu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Hu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sikai Chen
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
32
|
Song J, Yu T, Yan Q, Wu L, Li S, Wang L. A simple APACHE IV risk dynamic nomogram that incorporates early admitted lactate for the initial assessment of 28-day mortality in critically ill patients with acute myocardial infarction. BMC Cardiovasc Disord 2022; 22:502. [PMID: 36434509 PMCID: PMC9700900 DOI: 10.1186/s12872-022-02960-8] [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: 05/10/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Early risk stratification is important for patients with acute myocardial infarction (AMI). We aimed to develop a simple APACHE IV dynamic nomogram, combined with easily available clinical parameters within 24 h of admission, thus improving its predictive power to assess the risk of mortality at 28 days. METHODS Clinical information on AMI patients was extracted from the eICU database v2.0. A preliminary XGBoost examination of the degree of association between all variables in the database and 28-day mortality was conducted. Univariate and multivariate logistic regression analysis were used to perform screening of variables. Based on the multifactorial analysis, a dynamic nomogram predicting 28-day mortality in these patients was developed. To cope with missing data in records with missing variables, we applied the multiple imputation method. Predictive models are evaluated in three main areas, namely discrimination, calibration, and clinical validity. The discrimination is mainly represented by the area under the receiver operating characteristic curve (AUC), net reclassification improvement (NRI) and integrated discrimination improvement (IDI). Calibration is represented by the calibration plot. Clinical validity is represented by the decision curve analysis (DCA) curve. RESULTS A total of 504 people were included in the study. All 504 people were used to build the predictive model, and the internal validation model used a 500-bootstrap method. Multivariate analysis showed that four variables, APACHE IV, the first sample of admission lactate, prior atrial fibrillation (AF), and gender, were included in the nomogram as independent predictors of 28-day mortality in AMI. The prediction model had an AUC of 0.819 (95%CI 0.770-0.868) whereas the internal validation model had an AUC of 0.814 (95%CI 0.765-0.860). Calibration and DCA curves indicated that the dynamic nomogram in this study were reflective of real-world conditions and could be applied clinically. The predictive model composed of these four variables outperformed a single APACHE IV in terms of NRI and IDI. The NRI was 16.4% (95% CI: 6.1-26.8%; p = 0.0019) and the IDI was 16.4% (95% CI: 6.0-26.8%; p = 0.0020). Lactate accounted for nearly half of the total NRI, which showed that lactate was the most important of the other three variables. CONCLUSION The prediction model constructed by APACHE IV in combination with the first sample of admission lactate, prior AF, and gender outperformed the APACHE IV scoring system alone in predicting 28-day mortality in AMI. The prediction dynamic nomogram model was published via a website app, allowing clinicians to improve the predictive efficacy of the APACHE IV score by 16.4% in less than 1 min.
Collapse
Affiliation(s)
- Jikai Song
- grid.410645.20000 0001 0455 0905Zhejiang Provincial People’s Hospital, Qingdao University, Hangzhou, Zhejiang Province China
| | - Tianhang Yu
- grid.440734.00000 0001 0707 0296North China University of Science and Technology, Tangshan, Hebei Province China
| | - Qiqi Yan
- grid.410645.20000 0001 0455 0905Zhejiang Provincial People’s Hospital, Qingdao University, Hangzhou, Zhejiang Province China
| | - Liuyang Wu
- grid.410645.20000 0001 0455 0905Zhejiang Provincial People’s Hospital, Qingdao University, Hangzhou, Zhejiang Province China
| | - Sujing Li
- grid.410645.20000 0001 0455 0905Zhejiang Provincial People’s Hospital, Qingdao University, Hangzhou, Zhejiang Province China
| | - Lihong Wang
- grid.410645.20000 0001 0455 0905Zhejiang Provincial People’s Hospital, Qingdao University, Hangzhou, Zhejiang Province China
| |
Collapse
|
33
|
Wang Z, Li K, Xu J, Cheng X, Wang D. Construction of a lactate-related prognostic signature for predicting prognosis after surgical repair for acute type a aortic dissection. Front Physiol 2022; 13:1008869. [PMID: 36467680 PMCID: PMC9709272 DOI: 10.3389/fphys.2022.1008869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/03/2022] [Indexed: 10/14/2023] Open
Abstract
Background: Serum lactate is commonly measured in the perioperative period in patients who have undergone surgery for an acute type A aortic dissection (ATAAD). However, conflicting data has been reported as to whether lactate elevation is associated with short-term prognosis. The aim of the current study was to determine the association between perioperative arterial lactate levels and postoperative 30-day mortality. Methods: Patients who underwent repair of a ATAAD at our institution were retrospectively screened and those with comprehensive measurements of serum lactate before surgery and at 0, 1, 3, 6, 12, and 24 h after surgery in the intensive care unit (ICU) were selected for the analysis. Patients' demographic features and outcomes were reviewed to determine risk factors associated with 30-day mortality using logistic regression modeling. The association between serum lactate levels at different time points and 30-day mortality were analyzed by receiver-operating characteristic curves. Results: 513 patients were identified and retrospectively analyzed for this study including 66 patients (12.9%) who died within 30 days after surgery. Patients who died within 30 days after surgery had elevated lactate levels measured before surgery and at 0, 1, 3, 6, 12, and 24 h after their ICU stay. Lactate measured at 24 h post ICU admission (odds ratio, 2.131; 95% confidence interval, 1.346-3.374; p = 0.001) was a predictor of 30-day mortality. The area under the curve (AUC) for 30-day mortality with lactate levels at 12 h and 24 h post ICU stay were 0.820 and 0.805, respectively. Conclusion: Early elevation of lactate level is correlated with increased 30-day mortality in patients who received ATAAD surgical repair.
Collapse
Affiliation(s)
- Zhigang Wang
- Department of Cardio-thoracic Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Kai Li
- Department of Cardio-thoracic Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jingfang Xu
- Department of Nephrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaofeng Cheng
- Department of Cardio-thoracic Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dongjin Wang
- Department of Cardio-thoracic Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Cardio-thoracic Surgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Szabo D, Szabo A, Magyar L, Banhegyi G, Kugler S, Pinter A, Juhasz V, Ruppert M, Olah A, Ruzsa Z, Edes IF, Szekely A, Becker D, Merkely B, Hizoh I. Admission lactate level and the GRACE 2.0 score are independent and additive predictors of 30-day mortality of STEMI patients treated with primary PCI-Results of a real-world registry. PLoS One 2022; 17:e0277785. [PMID: 36383629 PMCID: PMC9668119 DOI: 10.1371/journal.pone.0277785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In many of the risk estimation algorithms for patients with ST-elevation myocardial infarction (STEMI), heart rate and systolic blood pressure are key predictors. Yet, these parameters may also be altered by the applied medical treatment / circulatory support without concomitant improvement in microcirculation. Therefore, we aimed to investigate whether venous lactate level, a well-known marker of microcirculatory failure, may have an added prognostic value on top of the conventional variables of the "Global Registry of Acute Coronary Events" (GRACE) 2.0 model for predicting 30-day all-cause mortality of STEMI patients treated with primary percutaneous coronary intervention (PCI). METHODS In a prospective single-center registry study conducted from May 2020 through April 2021, we analyzed data of 323 cases. Venous blood gas analysis was performed in all patients at admission. Nested logistic regression models were built using the GRACE 2.0 score alone (base model) and with the addition of venous lactate level (expanded model) with 30-day all-cause mortality as primary outcome measure. Difference in model performance was analyzed by the likelihood ratio (LR) test and the integrated discrimination improvement (IDI). Independence of the predictors was evaluated by the variance inflation factor (VIF). Discrimination and calibration was characterized by the c-statistic and calibration intercept / slope, respectively. RESULTS Addition of lactate level to the GRACE 2.0 score improved the predictions of 30-day mortality significantly as assessed by both LR test (LR Chi-square = 8.7967, p = 0.0030) and IDI (IDI = 0.0685, p = 0.0402), suggesting that the expanded model may have better predictive ability than the GRACE 2.0 score. Furthermore, the VIF was 1.1203, indicating that the measured lactate values were independent of the calculated GRACE 2.0 scores. CONCLUSIONS Our results suggest that admission venous lactate level and the GRACE 2.0 score may be independent and additive predictors of 30-day all-cause mortality of STEMI patients treated with primary PCI.
Collapse
Affiliation(s)
- Dominika Szabo
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Andras Szabo
- School of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Levente Magyar
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | | | - Szilvia Kugler
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Anita Pinter
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Vencel Juhasz
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Mihaly Ruppert
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Attila Olah
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Zoltan Ruzsa
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Division of Invasive Cardiology, 2 Department of Internal Medicine, University of Szeged, Szeged, Hungary
| | | | - Andrea Szekely
- Department of Oxiology and Emergency Care, Semmelweis University, Budapest, Hungary
| | - David Becker
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Istvan Hizoh
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
35
|
Kano R, Sato K. The Relationship between Training Cycle-Dependent Fluctuations in Resting Blood Lactate Levels and Exercise Performance in College-Aged Rugby Players. J Funct Morphol Kinesiol 2022; 7:jfmk7040093. [PMID: 36278754 PMCID: PMC9589951 DOI: 10.3390/jfmk7040093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
An increase in resting blood lactate (La-) concentration due to metabolic conditions has been reported. However, it is not clear whether resting La- changes with training cycles in athletes. The purpose of this study was to test the hypotheses that (1) the morning resting La- levels are lower in periods of high training compared to periods of low training and (2) these changes in La- concentration are related to athletes' metabolic capacity during exercise in male college-aged rugby players. Resting La- and blood glucose concentrations were measured in the morning in eight league rugby players during the summer pre-season period (Pre-period), the training and competition season period (TC-period), and the winter post-season period (Post-period). In each period, anaerobic power, La- concentration, and respiratory responses were measured during the 40 s maximal Wingate anaerobic test (WT). The resting La- concentration in the morning was significantly lower in the TC-Period (1.9 ± 0.6 mmol/L) than in the Post-Period (2.3 ± 0.9 mmol/L). The rate of decrease in La- level immediately after the 40 s WT was significantly higher in the TC-Period than in the Post-Period. The resting La- concentration was significantly correlated with the peak oxygen uptake and the carbon dioxide output during the WT. These results support the hypothesis that an athlete's training cycle (i.e., in season and off season) influences the resting La- levels as well as the metabolic capacity during high-intensity exercise. The monitoring of resting La- fluctuations may provide a convenient indication of the training cycle-dependent metabolic capacity in athletes.
Collapse
Affiliation(s)
- Ryotaro Kano
- Graduate School of Education, Tokyo Gakugei University, Tokyo 184-8501, Japan
| | - Kohei Sato
- Department of Health and Physical Education, Tokyo Gakugei University, Tokyo 184-8501, Japan
- Correspondence:
| |
Collapse
|
36
|
Ganushchak YM, Kurniawati ER, van der Horst IC, van Kuijk SM, Weerwind PW, Lorusso R, Maessen JG. Patterns of oxygen debt repayment in cardiogenic shock patients sustained with extracorporeal life support: A retrospective study. J Crit Care 2022; 71:154044. [DOI: 10.1016/j.jcrc.2022.154044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 03/04/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
|
37
|
Zhang H, Tian W, Sun Y. The value of anion gap for predicting the short-term all-cause mortality of critically ill patients with cardiac diseases, based on MIMIC-III database. Heart Lung 2022; 55:59-67. [PMID: 35477032 DOI: 10.1016/j.hrtlng.2022.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND The association of anion gap (AG) with short-term mortality in the critically ill patients with cardiac diseases is still not well understood. OBJECTIVE To evaluate the association of AG with short-term mortality, and the predictive ability of AG for short-term mortality in critically ill patients with cardiac diseases. METHODS This retrospective cohort study enrolled 9104 critically ill patients with cardiac diseases from the Medical Information Mart for Intensive Care III (MIMIC III) database. The restricted cubic spline models were used to evaluate the nonlinear relationship between AG and short-term mortality. Cox proportional hazards regression models and subgroup analysis were applied to assess the association of AG with short-term mortality. RESULTS The data were divided into three groups by AG tertiles: tertile I (AG <12, n = 2095), tertile II (12 ≤ AG < 15, n = 3195), and tertile III (15 ≤ AG, n = 3814). The restricted cubic spline models revealed continuous AG was non-linearly related to short-term mortality. The elevated AG tertiles were strongly associated with higher in-hospital, 30-day and 90-day mortality (all P for trend < 0.001). After adding AG to traditional severity scores, the area under curves (AUCs) elevated significantly compared to severity scores alone (all DeLong's test: P < 0.001). Subgroup analysis did not indicate significant interaction in most diverse subgroups. CONCLUSION AG was an independent risk factor for short-term all-cause mortality in critically ill patients with cardiac diseases. AG improved significantly the mortality predictive abilities of traditional severity scores when AG was added to these scores.
Collapse
Affiliation(s)
- HuanRui Zhang
- Department of Geriatric Cardiology, The First Affiliated Hospital of China Medical University, NO.155 Nanjing North Street, Heping Ward, Shenyang 110001, China
| | - Wen Tian
- Department of Geriatric Cardiology, The First Affiliated Hospital of China Medical University, NO.155 Nanjing North Street, Heping Ward, Shenyang 110001, China
| | - YuJiao Sun
- Department of Geriatric Cardiology, The First Affiliated Hospital of China Medical University, NO.155 Nanjing North Street, Heping Ward, Shenyang 110001, China.
| |
Collapse
|
38
|
Wang Z, Xu J, Kang Y, Liu L, Zhang L, Wang D. Early dynamic behavior of lactate in predicting continuous renal replacement therapy after surgery for acute type A aortic dissection. Front Cardiovasc Med 2022; 9:948672. [PMID: 35958404 PMCID: PMC9360317 DOI: 10.3389/fcvm.2022.948672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIt has been well known that hyperlactatemia is an independent risk factor for postoperative mortality in patients who received acute type A aortic dissection (ATAAD) surgery. Some patients may require the assistance of continuous renal replacement therapy (CRRT) for acute postoperative renal deficiency and often associate with increased mortality rate. This study aimed to examine the association between the early dynamic change of lactate levels and postoperative CRRT in ATAAD patients who received surgical repairment.MethodsThis retrospective study included 503 patients who received ATAAD surgeries. Serum lactate levels were measured before operation and at 0, 1, 3, 6, 12, 24 h post intensive care unit (ICU) admission. We examined the association between dynamic changes of lactate and CRRT.ResultsAmong all patients, 19.9% (100 patients) required CRRT. Our data showed that the lactate levels were higher in the CRRT group at all timepoints compared to the non-CRRT group. In a multivariate model, lactate levels at 12 h post ICU admission [odds ratio (OR), 1.362; p = 0.007] was identified as an independent predictor for requiring CRRT. Unsurprisingly, 30-day mortality in the CRRT group (41%) was 8.2 times higher than in the non-CRRT group (5%). To better understand the associations between CRRT and lactate levels, patients in the CRRT group were further stratified into the non-survivor group (n = 41) and survivor group (n = 59) based on the 30-day mortality. Elevated lactate levels measured upon ICU admission (OR, 1.284; p = 0.001) and decreased 24 h lactate clearance (OR, 0.237; p = 0.039) were independent risk factors for 30-day mortality in patients who received CRRT. The area under the curve to predict requirement for CRRT at 6 and 12 h post CICU admission were 0.714 and 0.722, respectively, corresponding to lactate cut-off levels of 4.15 and 2.45 mmol/L.ConclusionThe CRRT is commonly required in patients who received ATAAD surgery and often associated with worse mortality. Early dynamic changes of lactate levels can be used to predict the requirement of postoperative CRRT.
Collapse
Affiliation(s)
- Zhigang Wang
- Department of Cardio-Thoracic Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jingfang Xu
- Department of Nephrology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yubei Kang
- Department of Cardio-Thoracic Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ling Liu
- Department of Cardio-Thoracic Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lifang Zhang
- Department of Psychiatry, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Dongjin Wang
- Department of Cardio-Thoracic Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Dongjin Wang,
| |
Collapse
|
39
|
Yang K, Holt M, Fan M, Lam V, Yang Y, Ha T, Williams DL, Li C, Wang X. Cardiovascular Dysfunction in COVID-19: Association Between Endothelial Cell Injury and Lactate. Front Immunol 2022; 13:868679. [PMID: 35401579 PMCID: PMC8984030 DOI: 10.3389/fimmu.2022.868679] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/01/2022] [Indexed: 12/27/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), an infectious respiratory disease propagated by a new virus known as Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), has resulted in global healthcare crises. Emerging evidence from patients with COVID-19 suggests that endothelial cell damage plays a central role in COVID-19 pathogenesis and could be a major contributor to the severity and mortality of COVID-19. Like other infectious diseases, the pathogenesis of COVID-19 is closely associated with metabolic processes. Lactate, a potential biomarker in COVID-19, has recently been shown to mediate endothelial barrier dysfunction. In this review, we provide an overview of cardiovascular injuries and metabolic alterations caused by SARS-CoV-2 infection. We also propose that lactate plays a potential role in COVID-19-driven endothelial cell injury.
Collapse
Affiliation(s)
- Kun Yang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Matthew Holt
- James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Min Fan
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Victor Lam
- College of Arts and Science, New York University, New York City, NY, United States
| | - Yong Yang
- James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - David L. Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Chuanfu Li
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
40
|
Boeken U, Assmann A, Beckmann A, Schmid C, Werdan K, Michels G, Miera O, Schmidt F, Klotz S, Starck C, Pilarczyk K, Rastan A, Burckhardt M, Nothacker M, Muellenbach R, Zausig Y, Haake N, Groesdonk H, Ferrari M, Buerke M, Hennersdorf M, Rosenberg M, Schaible T, Köditz H, Kluge S, Janssens U, Lubnow M, Flemmer A, Herber-Jonat S, Wessel L, Buchwald D, Maier S, Krüger L, Fründ A, Jaksties R, Fischer S, Wiebe K, Hartog CS, Dzemali O, Zimpfer D, Ruttmann-Ulmer E, Schlensak C, Kelm M, Ensminger S. S3 Guideline of Extracorporeal Circulation (ECLS/ECMO) for Cardiocirculatory Failure. Thorac Cardiovasc Surg 2021; 69:S121-S212. [PMID: 34655070 DOI: 10.1055/s-0041-1735490] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Udo Boeken
- Department of Cardiac Surgery, Heinrich-Heine-University Medical School, Duesseldorf, Germany
| | - Alexander Assmann
- Department of Cardiac Surgery, Heinrich-Heine-University Medical School, Duesseldorf, Germany
| | - Andreas Beckmann
- German Society for Thoracic and Cardiovascular Surgery, Langenbeck-Virchow-Haus, Berlin, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Karl Werdan
- Clinic for Internal Medicine III, University Hospital Halle (Saale), Martin-Luther University Halle-Wittenberg, Germany
| | - Guido Michels
- Department of Acute and Emergency Care, St Antonius Hospital Eschweiler, Eschweiler, Germany
| | - Oliver Miera
- Department of Congenital Heart Disease-Pediatric Cardiology, German Heart Center Berlin, Berlin, Germany
| | - Florian Schmidt
- Department of Pediatric Cardiology and Intensive Care Medicine, Medical School Hannover, Hannover, Germany
| | - Stefan Klotz
- Department of Cardiac Surgery, Segeberger Kliniken, Bad Segeberg, Germany
| | - Christoph Starck
- Department of Cardiothoracic and Vascular Surgery, German Heart Centre, Berlin, German
| | - Kevin Pilarczyk
- Department for Intensive Care Medicine, Imland Hospital Rendsburg, Rendsburg, Schleswig-Holstein, Germany
| | - Ardawan Rastan
- Department of Cardiac and Vascular Thoracic Surgery, Philipps-University Hospital Marburg, Marburg, Germany
| | - Marion Burckhardt
- Department of Health Sciences and Management; Baden-Wuerttemberg Cooperative State University (DHBW), Stuttgart, Germany
| | - Monika Nothacker
- Institute for Medical Knowledge Management, Association of the Scientific Medical Societies (AWMF), Universität Marburg, Marburg, Germany
| | - Ralf Muellenbach
- Department of Anaesthesiology and Critical Care Medicine, Campus Kassel of the University of Southampton, Kassel, Germany
| | - York Zausig
- Department of Anesthesiology and Operative Intensive Care Medicine, Aschaffenburg-Alzenau Hospital, Aschaffenburg, Bavaria, Germany
| | - Nils Haake
- Department for Intensive Care Medicine, Imland Hospital Rendsburg, Rendsburg, Schleswig-Holstein, Germany
| | - Heinrich Groesdonk
- Department of Intensive Care Medicine, Helios Clinic Erfurt, Erfurt, Germany
| | - Markus Ferrari
- HSK, Clinic of Internal Medicine I, Helios-Kliniken, Wiesbaden, Germany
| | - Michael Buerke
- Department of Cardiology, Angiology and Intensive Care Medicine, St. Marienkrankenhaus Siegen, Siegen, Germany
| | - Marcus Hennersdorf
- Department of Cardiology, Pneumology, Angiology and Internal Intensive Care Medicine, SLK-Kliniken Heilbronn, Heilbronn, Germany
| | - Mark Rosenberg
- Klinikum Aschaffenburg-Alzenau, Medizinische Klinik 1, Aschaffenburg, Germany
| | - Thomas Schaible
- Department of Neonatology, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Harald Köditz
- Medical University Children's Hospital, Hannover, Germany
| | - Stefan Kluge
- Klinik für Intensivmedizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland
| | - Uwe Janssens
- Medical Clinic and Medical Intensive Care Medicine, St Antonius Hospital, Eschweiler, Germany
| | - Matthias Lubnow
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Flemmer
- Division of Neonatology, Dr. v. Hauner Children's Hospital and Perinatal Center Munich - Grosshadern, LMU Munich, Munich, Germany
| | - Susanne Herber-Jonat
- Division of Neonatology, Dr. v. Hauner Children's Hospital and Perinatal Center Munich - Grosshadern, LMU Munich, Germany
| | - Lucas Wessel
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Dirk Buchwald
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Germany
| | - Sven Maier
- Department of Cardiovascular Surgery, Heart Center Freiburg University, Freiburg, Germany
| | - Lars Krüger
- Division of Thoracic and Cardiovascular Surgery, Heart- and Diabetescentre NRW, Ruhr-University, Bochum, Germany
| | - Andreas Fründ
- Department of Physiotherapy, Heart- and Diabetescentre NRW, Ruhr-University, Bochum, Germany
| | | | - Stefan Fischer
- Department of Thoracic Surgery and Lung Support, Ibbenbueren General Hospital, Ibbenbueren, Germany
| | - Karsten Wiebe
- Department of Cardiothoracic Surgery, Münster University Hospital, Münster, Germany
| | - Christiane S Hartog
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité Universitätsmedizin Berlin, and Klinik Bavaria, Kreischa
| | - Omer Dzemali
- Department of Cardiac Surgery, Triemli City hospital Zurich, Zurich, Switzerland
| | - Daniel Zimpfer
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Christian Schlensak
- Department of Cardio-Thoracic and Vascular Surgery, University of Tübingen, Tübingen, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Heinrich-Heine-University Medical School, Duesseldorf, Germany
| | - Stephan Ensminger
- Department of Cardiac and Thoracic Vascular Surgery, University Hospital of Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
41
|
Zhang T, Wang J, Li X. Association Between Anion Gap and Mortality in Critically Ill Patients with Cardiogenic Shock. Int J Gen Med 2021; 14:4765-4773. [PMID: 34466021 PMCID: PMC8403005 DOI: 10.2147/ijgm.s329150] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/12/2021] [Indexed: 11/23/2022] Open
Abstract
Background No epidemiological study has determined the association between the anion gap (AG) and all-cause mortality in critically ill patients with cardiogenic shock (CS). This study was conducted to clarify the relationship between the AG and mortality in CS. Methods We extracted clinical data from the public database, MIMIC-III V1.4, by using a generalized additive model to identify the nonlinear relationship between the AG and the 30-day mortality in 1248 intensive care unit patients. Cox proportional hazard models were used to assess the association between the AG and the 30-day, 90-day, and 365-day mortality in CS. Results The AG and 30-day all-cause mortality showed a nonlinear relationship, indicated by a J-shaped curve. In the multivariate analysis, after adjusting for potential confounders, a high AG was associated with an increased risk of 30-day, 90-day, and 365-day all-cause mortality in patients with CS compared with patients who had low AG (hazard ratio [95% confidence interval] 1.62 [1.14-2.30]; 1.35 [1.04-1.84]; and 1.38 [1.03-1.84], respectively). Similar results were shown in Model I (adjusted for age, sex and ethnicity) and in Model II (fully adjusting for age, ethnicity, sex, acute kidney injury stage, CHF, renal disease, stroke, malignancy, respiratory failure, pneumonia, sodium, potassium, chloride, BUN, PT, WBC, pH, creatinine, albumin, glucose, bicarbonate, vasopressor use, diastolic blood pressure, respiration rate, temperature, the Elixhauser Comorbidity Index, SOFA score and SAPSII score). Conclusion The relationship between the AG and 30-day all-cause mortality followed a J-shaped curve. Higher AG was associated with an increased risk of 30-day, 90-day, and 365-day all-cause mortality in critically ill patients with CS.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Jie Wang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Xiangyang Li
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| |
Collapse
|
42
|
Fuernau G, Desch S, de Waha-Thiele S, Eitel I, Neumann FJ, Hennersdorf M, Felix SB, Fach A, Böhm M, Pöss J, Jung C, Ouarrak T, Schneider S, Werdan K, Zeymer U, Thiele H. Arterial Lactate in Cardiogenic Shock: Prognostic Value of Clearance Versus Single Values. JACC Cardiovasc Interv 2021; 13:2208-2216. [PMID: 33032708 DOI: 10.1016/j.jcin.2020.06.037] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES This study sought to compare single lactate values at admission (L1) and after 8 h (L2) with lactate clearance (LC) for mortality prediction in cardiogenic shock (CS). BACKGROUND Early estimation of prognosis in CS complicating acute myocardial infarction is crucial for tailored treatment selection. Arterial lactate is the most widely used point-of-care parameter in CS. In septic shock, lactate reduction over time-LC-has been extensively investigated. However, in CS, only limited data exist, and the prognostic value of LC is unknown. METHODS This study is a subanalysis of the IABP-SHOCK II (Intraaortic Balloon Pump in Cardiogenic Shock II) trial and the corresponding registry. Lactate levels were prospectively collected. All-cause mortality at 30 days was assessed as primary endpoint. RESULTS For 671 of 783 (85.7%) patients, L1 and L2 values were available. The area under the receiver-operating characteristic curve (L1: 0.69; L2: 0.76; LC: 0.59) showed no difference between L1 and LC (p = 0.20). In contrast, L2 was a significantly better predictive parameter than L1 or LC (p < 0.001 for both). In multivariable stepwise Cox regression analysis, L2 ≥3.1 mmol/l (best cutoff value by Youden index) and LC <-3.45%/h remained independently predictive for time to death (p < 0.001 for both), with L2 showing the highest chi-square test score (42.1) and hazard ratio (2.89; 95% confidence interval: 2.10 to 3.97). CONCLUSIONS Arterial lactate after 8 h is superior in mortality prediction in comparison with baseline lactate and LC. A cutoff value of 3.1 mmol/l for lactate after 8 h showed the best discrimination for assessing early prognosis in CS and may serve as new treatment goal. (Intraaortic Balloon Pump in Cardiogenic Shock II [IABP-SHOCK II]; NCT00491036).
Collapse
Affiliation(s)
- Georg Fuernau
- Medical Clinic II (Cardiology, Angiology, Intensive Care Medicine), University Heart Center Lübeck, Lübeck, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany.
| | - Steffen Desch
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany; Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig and Leipzig Heart Institute, Leipzig, Germany
| | - Suzanne de Waha-Thiele
- Medical Clinic II (Cardiology, Angiology, Intensive Care Medicine), University Heart Center Lübeck, Lübeck, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Ingo Eitel
- Medical Clinic II (Cardiology, Angiology, Intensive Care Medicine), University Heart Center Lübeck, Lübeck, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Franz-Josef Neumann
- Department of Cardiology, Bad Krozingen Heart Center, University of Freiburg, Bad Krozingen, Germany
| | - Marcus Hennersdorf
- Department of Internal Medicine I, SLK Kiniken Heilbronn, Heilbronn, Germany
| | - Stephan B Felix
- Department of Internal Medicine B, University of Greifswald, Greifswald, Germany
| | - Andreas Fach
- Department of Cardiology/Angiology, Klinikum Links der Weser, Bremen, Germany
| | - Michael Böhm
- Department of Internal Medicine III, University of Homburg, Homburg/Saar, Germany
| | - Janine Pöss
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig and Leipzig Heart Institute, Leipzig, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Düsseldorf, Düsseldorf, Germany
| | | | | | - Karl Werdan
- Department of Internal Medicine III, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany
| | - Uwe Zeymer
- Institut für Herzinfarktforschung, Ludwigshafen, Germany; Medizinische Klinik B, Klinikum Ludwigshafen, Ludwigshafen, Germany
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Leipzig and Leipzig Heart Institute, Leipzig, Germany
| |
Collapse
|
43
|
Palmer JA, Smith AM, Gryshkova V, Donley ELR, Valentin JP, Burrier RE. A Targeted Metabolomics-Based Assay Using Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Identifies Structural and Functional Cardiotoxicity Potential. Toxicol Sci 2021; 174:218-240. [PMID: 32040181 DOI: 10.1093/toxsci/kfaa015] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Implementing screening assays that identify functional and structural cardiotoxicity earlier in the drug development pipeline has the potential to improve safety and decrease the cost and time required to bring new drugs to market. In this study, a metabolic biomarker-based assay was developed that predicts the cardiotoxicity potential of a drug based on changes in the metabolism and viability of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). Assay development and testing was conducted in 2 phases: (1) biomarker identification and (2) targeted assay development. In the first phase, metabolomic data from hiPSC-CM spent media following exposure to 66 drugs were used to identify biomarkers that identified both functional and structural cardiotoxicants. Four metabolites that represent different metabolic pathways (arachidonic acid, lactic acid, 2'-deoxycytidine, and thymidine) were identified as indicators of cardiotoxicity. In phase 2, a targeted, exposure-based biomarker assay was developed that measured these metabolites and hiPSC-CM viability across an 8-point concentration curve. Metabolite-specific predictive thresholds for identifying the cardiotoxicity potential of a drug were established and optimized for balanced accuracy or sensitivity. When predictive thresholds were optimized for balanced accuracy, the assay predicted the cardiotoxicity potential of 81 drugs with 86% balanced accuracy, 83% sensitivity, and 90% specificity. Alternatively, optimizing the thresholds for sensitivity yields a balanced accuracy of 85%, 90% sensitivity, and 79% specificity. This new hiPSC-CM-based assay provides a paradigm that can identify structural and functional cardiotoxic drugs that could be used in conjunction with other endpoints to provide a more comprehensive evaluation of a drug's cardiotoxicity potential.
Collapse
Affiliation(s)
| | - Alan M Smith
- Stemina Biomarker Discovery, Inc, Madison, Wisconsin
| | - Vitalina Gryshkova
- UCB Biopharma SPRL, Investigative Toxicology, Development Science, B-1420 Braine L'Alleud, Belgium
| | | | - Jean-Pierre Valentin
- UCB Biopharma SPRL, Investigative Toxicology, Development Science, B-1420 Braine L'Alleud, Belgium
| | | |
Collapse
|
44
|
Kong S, Chen C, Zheng G, Yao H, Li J, Ye H, Wang X, Qu X, Zhou X, Lu Y, Zhou H. A prognostic nomogram for long-term major adverse cardiovascular events in patients with acute coronary syndrome after percutaneous coronary intervention. BMC Cardiovasc Disord 2021; 21:253. [PMID: 34022791 PMCID: PMC8141252 DOI: 10.1186/s12872-021-02051-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/05/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Accurate prediction of major adverse cardiovascular events (MACEs) is very important for the management of acute coronary syndrome (ACS) patients. We aimed to construct an effective prognostic nomogram for individualized risk estimates of MACEs for patients with ACS after percutaneous coronary intervention (PCI). METHODS This was a prospective study of patients with ACS after PCI from January 2013 to July 2019 (n = 2465). After removing patients with incomplete clinical information, a total of 1986 patients were randomly divided into evaluation (n = 1324) and validation (n = 662) groups. Predictors included in the nomogram were determined by a multivariate Cox proportional hazards regression model based on the training set. Receiver operating characteristic (ROC) curves and calibration curves were used to assess the discrimination and predictive accuracy of the nomogram, which were then compared with those of the classic models. The clinical utility of the nomogram was assessed by X-tile analysis and Kaplan-Meier curve analysis. RESULTS Independent prognostic factors, including lactate level, age, left anterior descending branch stenosis, right coronary artery stenosis, brain natriuretic peptide level, and left ventricular ejection fraction, were determined and contained in the nomogram. The nomogram achieved good areas under the ROC curve of 0.712-0.762 in the training set and 0.724-0.818 in the validation set and well-fitted calibration curves. In addition, participants could be divided into two risk groups (low and high) according to this model. CONCLUSIONS A simple-to-use nomogram incorporating lactate level effectively predicted 6-month, 1-year, and 4-year MACE incidence among patients with ACS after PCI.
Collapse
Affiliation(s)
- Shuting Kong
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Changxi Chen
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Gaoshu Zheng
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hui Yao
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Junfeng Li
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hong Ye
- Cardiac Interventional Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaobo Wang
- Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinghua, 321000, Zhejiang, China
| | - Xiang Qu
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaodong Zhou
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yucheng Lu
- The First Clinical Medical College of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hao Zhou
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
45
|
Pories WJ, Jones TE, Houmard JA, DeMaria E, Dohm GL. Ockham's razor and the metabolic syndrome. Surg Obes Relat Dis 2021; 17:1236-1243. [PMID: 33965350 DOI: 10.1016/j.soard.2021.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/21/2021] [Accepted: 04/02/2021] [Indexed: 12/11/2022]
Abstract
The broad effects of bariatric/metabolic surgery on virtually every tissue and organ system remain unexplained. Weight loss, although a major factor, does not fully account for the rapid, full, and durable remission of type 2 diabetes, return of islet function, reduction of the prevalence of cancers, increase in gray matter of the brain, and decrease in all-cause mortality. This review supports the thesis that the metabolic syndrome is not a group of separate diseases but rather multiple expressions of a shared defect in the utilization of carbohydrates and lipids. That error is probably caused by a dysmetabolic signal from the foregut, stimulated by food, that limits entry of 2-carbon fragments into the tricarboxylic acid cycle, the accumulation of lactate and, in turn, increases in glucose and insulin. Surgery limits that signal by reducing contact between food and foregut mucosa. Speciation of that signal(s) may offer a new pathway for drug development.
Collapse
Affiliation(s)
- Walter J Pories
- Department of Surgery, Brody School of Medicine, East Carolina University, Greenville, North Carolina.
| | - Terry E Jones
- College of Allied Health Sciences, East Carolina University, Greenville, North Carolina
| | - Joseph A Houmard
- Department of Kinesiology, East Carolina University, Greenville, North Carolina
| | - Eric DeMaria
- Department of Surgery, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - G Lynis Dohm
- Department of Physiology, East Carolina University, Greenville, North Carolina
| |
Collapse
|
46
|
Kurniawati ER, Rutjens VGH, Vranken NPA, Delnoij TSR, Lorusso R, van der Horst ICC, Maessen JG, Weerwind PW. Quality of life following adult veno-venous extracorporeal membrane oxygenation for acute respiratory distress syndrome: a systematic review. Qual Life Res 2021; 30:2123-2135. [PMID: 33826058 PMCID: PMC8024673 DOI: 10.1007/s11136-021-02834-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Veno-venous extracorporeal membrane oxygenation (VV-ECMO) has been used successfully for the past decade in adult patients with acute respiratory distress syndrome (ARDS) refractory to conventional ventilatory support. However, knowledge of the health-related quality of life (HRQoL) in VV-ECMO patients is still limited. Thus, this study aimed to provide a comprehensive overview of the HRQoL following VV-ECMO support in ARDS patients. METHODS A systematic search was performed on PubMed and Web of Science databases from January 1st, 2009 to October 19th, 2020. Studies reporting on HRQoL following VV-ECMO for ARDS in adults were included. Two authors independently selected studies, extracted data, and assessed methodological quality. RESULTS Eight studies were eligible for inclusion, consisting of seven observational studies and one randomized controlled trial (total N = 441). All eight studies had a quantitative design and reported 265 VV-ECMO survivors to have a reduced HRQoL compared to a generally healthy population. Follow-up time varied between six months to three years. Additionally, only four studies (total N = 335) compared the HRQoL of VV-ECMO (N = 159) to conventionally treated survivors (N = 176), with one study showing a significantly better HRQoL in VV-ECMO survivors, while three studies were stating comparable HRQoL across groups. Notably, most survivors in these studies appeared to experience varying degrees of anxiety, depression, and post-traumatic stress disorder (PTSD). CONCLUSIONS ARDS survivors supported by VV-ECMO have a decline in HRQoL and suffered from physical and psychological impairments. This HRQoL reduction is comparable or even better to the HRQoL in conventionally treated ARDS survivors.
Collapse
Affiliation(s)
- E R Kurniawati
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, P. Debyelaan 25, PO Box 5800, 6202 AZ, Maastricht, the Netherlands.
| | - V G H Rutjens
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, P. Debyelaan 25, PO Box 5800, 6202 AZ, Maastricht, the Netherlands
| | - N P A Vranken
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, P. Debyelaan 25, PO Box 5800, 6202 AZ, Maastricht, the Netherlands
| | - T S R Delnoij
- Department of Intensive Care Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands.,Department of Cardiology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - R Lorusso
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, P. Debyelaan 25, PO Box 5800, 6202 AZ, Maastricht, the Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - I C C van der Horst
- Department of Intensive Care Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - J G Maessen
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, P. Debyelaan 25, PO Box 5800, 6202 AZ, Maastricht, the Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - P W Weerwind
- Department of Cardiothoracic Surgery, Maastricht University Medical Center+, P. Debyelaan 25, PO Box 5800, 6202 AZ, Maastricht, the Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
47
|
Hauck L, Dadson K, Chauhan S, Grothe D, Billia F. Inhibiting the Pkm2/b-catenin axis drives in vivo replication of adult cardiomyocytes following experimental MI. Cell Death Differ 2021; 28:1398-1417. [PMID: 33288902 PMCID: PMC8027412 DOI: 10.1038/s41418-020-00669-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 11/01/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Adult mammalian cardiomyocytes (CM) are postmitotic, differentiated cells that cannot re-enter the cell cycle after any appreciable injury. Therefore, understanding the factors required to induce CM proliferation for repair is of great clinical importance. While expression of muscle pyruvate kinase 2 (Pkm2), a cytosolic enzyme catalyzing the final step in glycolysis, is high in end-stage heart failure (HF), the loss of Pkm2 promotes proliferation in some cellular systems, in vivo. We hypothesized that in the adult heart CM proliferation may require low Pkm2 activity. Thus, we investigated the potential for Pkm2 to regulate CM proliferation in a mouse model of myocardial infarction (MI) employing inducible, cardiac-specific Pkm2 gene knockout (Pkm2KOi) mice. We found a lack of cardiac hypertrophy or expression of the fetal gene program in Pkm2KOi mice post MI, as compared to vehicle control animals (P < 0.01), correlating with smaller infarct size, improved mitochondrial (mt) function, enhanced angiogenesis, reduced degree of CM apoptosis, and reduced oxidative stress post MI. There was significantly higher numbers of dividing CM in the infarct zone between 3-9 days post MI (P < 0.001). Mechanistically, we determined that Pkm2 interacts with β-catenin (Ctnnb1) in the cytoplasm of CM, inhibiting Ctnnb1 phosphorylation at serine 552 and tyrosine 333, by Akt. In the absence of Pkm2, Ctnnb1 translocates to the nucleus leading to transcriptional activation of proliferation-associated target genes. All these effects are abrogated by genetic co-deletion of Pkm2 and Ctnnb1. Collectively, this work supports a novel antiproliferative function for Pkm2 in CM through the sequestration of Ctnnb1 in the cytoplasm of CM whereas loss of Pkm2 is essential for CM proliferation. Reducing cardiac Pkm2 expression may provide a useful strategy for cardiac repair after MI in patients.
Collapse
Affiliation(s)
- Ludger Hauck
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada
| | - Keith Dadson
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada
| | - Shelly Chauhan
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada
| | - Daniela Grothe
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada
| | - Filio Billia
- Toronto General Research Institute, 100 College St., M5G 1L7, Toronto, ON, Canada.
- Division of Cardiology, University Health Network (UHN), 200 Elizabeth St., Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
48
|
Akhavan AR, Johnson NJ, Friedman B, Hall J, Jablonowski K, Hall MK, Henning DJ. Assessing lactate concentration as a predictor of 28-day in-hospital mortality in the presence of ethanol: A retrospective study of emergency department patients. J Am Coll Emerg Physicians Open 2021; 2:e12397. [PMID: 33718929 PMCID: PMC7926009 DOI: 10.1002/emp2.12397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/12/2021] [Accepted: 02/11/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Presence of ethanol (EtOH) may alter the relationship between blood lactate concentrations and mortality. This study compares lactate-associated mortality risk in the presence and absence of EtOH. METHODS We performed a retrospective cohort study including all patients, age >17 years, presenting from January 2012-December 2018, to an urban, academic emergency department, with a clinically measured lactate. Data were electronically abstracted from the medical record. The primary outcome was 28-day in-hospital mortality. Patients were grouped by EtOH test results as follows: 1) present (any EtOH detected), 2) absent (EtOH concentration measured and not detected), or 3) not ordered. Marginal analysis was used to calculated probability of mortality for fixed values of lactate and model covariates. RESULTS Of 40,956 adult emergency department patients with measured lactate, we excluded 768 (1.89%) for lactate >10.0 mmol/L, leaving 40,240 for analysis: 4,066 (10.1%) EtOH present, 10,819 (26.9%) EtOH absent, 25,355 (63%) EtOH not ordered. Of these, 1790 (4.4%) had 28-day in-hospital mortality. Marginal probability of mortality calculated for specific lactate values found less risk for EtOH Present patients versus EtOH absent patients at lactate 0.0 mmol/L (0.8% [95%CI: 0.5-1.2%] vs 3.2% [2.8-3.6%]), 2.0 mmol/L (1.5% [1.1-1.9%] vs 4.0% [3.7-4.3%]), 4.0 mmol/L (2.6% [2.2-3.1%] vs 5.0% [4.6-5.4%]), until 6.0 mmol/L (4.5% [3.7-5.4%] vs 6.2% [5.4-7.0%]). CONCLUSION EtOH presence significantly alters lactate-associated mortality risk when lactate <6.0 mmol/L. Emergency department clinicians should interpret these lactate values with caution and consider other data for risk stratification when EtOH is present.
Collapse
Affiliation(s)
| | - Nicholas J. Johnson
- Department of Emergency MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Benjamin Friedman
- Department of Emergency MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Jane Hall
- Department of Emergency MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Karl Jablonowski
- Department of Emergency MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - M. Kennedy Hall
- Department of Emergency MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Daniel J. Henning
- Department of Emergency MedicineUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
49
|
Toma D, Toma TE, Bologa C, Lionte C. Unusual aetiology of a type 2 myocardial infarction: a case-based review. Arh Hig Rada Toksikol 2021; 72:80-87. [PMID: 33787179 PMCID: PMC8191432 DOI: 10.2478/aiht-2021-72-3502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/01/2020] [Accepted: 03/01/2021] [Indexed: 11/20/2022] Open
Abstract
Organophosphate pesticide (OP) poisoning is quite common and can cause cardiovascular complications and even direct myocardial injury. However, no guideline has included an acute poisoning as a potential cause for a type 2 myocardial infarction (MI) so far. Here we present a case of a 61-year-old woman brought by ambulance to emergency department one hour after accidental ingestion of an unknown quantity of a solution she used against flea infestation. The patient presented with dizziness, myosis, excessive sweating, hypersalivation, sphincteric incontinence, muscle fasciculation, tremor of the extremities, pale skin, alcoholic and pesticide breath odour. Even though we had no guidelines to fall back on, we successfully treated the patient with low-molecular-weight heparin, antiplatelets, statin, diltiazem, antidote therapy, and supportive care. Physicians should be aware that OP poisoning can induce type 2 MI as a complication within a few hours since exposure, and emergency management should always include close cardiac monitoring.
Collapse
Affiliation(s)
- David Toma
- Sf. Spiridon Emergency County Hospital, Emergency Department, Iași, Romania
| | - Tania-Emima Toma
- Sf. Maria Emergency Children’s Hospital, General Paediatrics, Iași, Romania
| | - Cristina Bologa
- “Grigore T. Popa” University of Medicine and Pharmacy, School of Medicine, Internal Medicine and Clinical Toxicology Department, Iași, Romania
- Sf. Spiridon Emergency County Hospital, 2 Internal Medicine Clinic, Iași, Romania
| | - Cătălina Lionte
- “Grigore T. Popa” University of Medicine and Pharmacy, School of Medicine, Internal Medicine and Clinical Toxicology Department, Iași, Romania
- Sf. Spiridon Emergency County Hospital, 2 Internal Medicine Clinic, Iași, Romania
| |
Collapse
|
50
|
Serum Lactate and A Relative Change in Lactate as Predictors of Mortality in Patients With Cardiogenic Shock - Results from the Cardshock Study. Shock 2021; 53:43-49. [PMID: 30973460 DOI: 10.1097/shk.0000000000001353] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Cardiogenic shock complicating acute myocardial infarction has a very high mortality. Our present study focuses on serial measurement of lactate during admission due to cardiogenic shock and the prognostic effect of lactate and a relative change in lactate in patients after admission and the institution of intensive care treatment. METHODS AND RESULTS This is a secondary analysis of the CardShock study. Data on lactate at baseline were available on 217 of 219 patients.In the study population, the median baseline lactate was 2.8 mmol/L (min-max range, 0.5-23.1 mmol/L).At admission, lactate was predictive of 30-day mortality with an adjusted Hazard ratio (HR) of 1.20 mmol/L (95% confidence interval, CI 1.14-1.27). Within the first 24 h of admission, baseline lactate remained predictive of 30-day mortality. Lactate at 6 h had a HR of 1.14 (95% CI 1.06-1.24) and corresponding values at 12 and 24 h had a HR of 1.10 (1.04-1.17), and of HR 1.19 (95% CI 1.07-1.32), respectively. A 50% reduction in lactate within 6 h resulted in a HR of 0.82 (95% CI 0.72-0.94). Corresponding hazard ratios at 12 and 24 h, were 0.87 (95% CI 0.76-0.98) and 0.74 (95% CI 0.60-0.91), respectively. CONCLUSION The main findings of the present study are that baseline lactate is a powerful predictor of 30-day mortality, lactate at 6, 12, and 24 h after admission are predictors of 30-day mortality, and a relative change in lactate is a significant predictor of survival within the first 24 h after instituting intensive care treatment adding information beyond the information from baseline values.
Collapse
|