1
|
Hiser SL, Casey K, Nydahl P, Hodgson CL, Needham DM. Intensive care unit acquired weakness and physical rehabilitation in the ICU. BMJ 2025; 388:e077292. [PMID: 39870417 DOI: 10.1136/bmj-2023-077292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Approximately half of critically ill adults experience intensive care unit acquired weakness (ICUAW). Patients who develop ICUAW may have negative outcomes, including longer duration of mechanical ventilation, greater length of stay, and worse mobility, physical functioning, quality of life, and mortality. Early physical rehabilitation interventions have potential for improving ICUAW; however, randomized trials show inconsistent findings on the efficacy of these interventions. This review summarizes the latest evidence on the definition, diagnosis, epidemiology, pathophysiology, risks factors, implications, and management of ICUAW. It specifically highlights research gaps and challenges, with considerations for future research for physical rehabilitation interventions.
Collapse
Affiliation(s)
- Stephanie L Hiser
- Department of Health, Human Function, and Rehabilitation Sciences, George Washington University, Washington, DC, USA
| | - Kelly Casey
- Department of Physical Medicine and Rehabilitation, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Peter Nydahl
- Department for Nursing Research and Development, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Carol L Hodgson
- Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Dale M Needham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Physical Medicine and Rehabilitation. Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Yumoto T, Coopersmith CM. Targeting AMP-activated protein kinase in sepsis. Front Endocrinol (Lausanne) 2024; 15:1452993. [PMID: 39469575 PMCID: PMC11513325 DOI: 10.3389/fendo.2024.1452993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Sepsis is a global health challenge marked by limited clinical options and high mortality rates. AMP-activated protein kinase (AMPK) is a cellular energy sensor that mediates multiple crucial metabolic pathways that may be an attractive therapeutic target in sepsis. Pre-clinical experimental studies have demonstrated that pharmacological activation of AMPK can offer multiple potential benefits during sepsis, including anti-inflammatory effects, induction of autophagy, promotion of mitochondrial biogenesis, enhanced phagocytosis, antimicrobial properties, and regulation of tight junction assembly. This review aims to discuss the existing evidence supporting the therapeutic potential of AMPK activation in sepsis management.
Collapse
Affiliation(s)
- Tetsuya Yumoto
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States
- Department of Emergency, Critical Care and Disaster Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Craig M. Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
3
|
Schaller SJ, Scheffenbichler FT, Bein T, Blobner M, Grunow JJ, Hamsen U, Hermes C, Kaltwasser A, Lewald H, Nydahl P, Reißhauer A, Renzewitz L, Siemon K, Staudinger T, Ullrich R, Weber-Carstens S, Wrigge H, Zergiebel D, Coldewey SM. Guideline on positioning and early mobilisation in the critically ill by an expert panel. Intensive Care Med 2024; 50:1211-1227. [PMID: 39073582 DOI: 10.1007/s00134-024-07532-2] [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: 02/28/2024] [Accepted: 06/15/2024] [Indexed: 07/30/2024]
Abstract
A scientific panel was created consisting of 23 interdisciplinary and interprofessional experts in intensive care medicine, physiotherapy, nursing care, surgery, rehabilitative medicine, and pneumology delegated from scientific societies together with a patient representative and a delegate from the Association of the Scientific Medical Societies who advised methodological implementation. The guideline was created according to the German Association of the Scientific Medical Societies (AWMF), based on The Appraisal of Guidelines for Research and Evaluation (AGREE) II. The topics of (early) mobilisation, neuromuscular electrical stimulation, assist devices for mobilisation, and positioning, including prone positioning, were identified as areas to be addressed and assigned to specialist expert groups, taking conflicts of interest into account. The panel formulated PICO questions (addressing the population, intervention, comparison or control group as well as the resulting outcomes), conducted a systematic literature review with abstract screening and full-text analysis and created summary tables. This was followed by grading the evidence according to the Oxford Centre for Evidence-Based Medicine 2011 Levels of Evidence and a risk of bias assessment. The recommendations were finalized according to GRADE and voted using an online Delphi process followed by a final hybrid consensus conference. The German long version of the guideline was approved by the professional associations. For this English version an update of the systematic review was conducted until April 2024 and recommendation adapted based on new evidence in systematic reviews and randomized controlled trials. In total, 46 recommendations were developed and research gaps addressed.
Collapse
Affiliation(s)
- Stefan J Schaller
- Department of Anaesthesiology and Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany.
| | | | | | - Manfred Blobner
- Department of Anaesthesiology and Intensive Care Medicine, Ulm University, Ulm, Germany
- Department of Anaesthesiology and Intensive Care Medicine, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Julius J Grunow
- Department of Anaesthesiology and Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Uwe Hamsen
- Ruhr University Bochum, Bochum, Germany
- Department of General and Trauma Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Carsten Hermes
- Hochschule für Angewandte Wissenschaften Hamburg (HAW Hamburg), Hamburg, Germany
- Akkon-Hochschule für Humanwissenschaften, Berlin, Germany
| | - Arnold Kaltwasser
- Academy of the District Hospitals Reutlingen, Kreiskliniken Reutlingen, Reutlingen, Germany
| | - Heidrun Lewald
- Department of Anaesthesiology and Intensive Care Medicine, School of Medicine and Health, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Peter Nydahl
- University Hospital of Schleswig-Holstein, Kiel, Germany
- Institute of Nursing Science and Development, Paracelsus Medical University, Salzburg, Austria
| | - Anett Reißhauer
- Department of Rehabilitation Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leonie Renzewitz
- Department of Physiotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Karsten Siemon
- Department of Pneumology, Fachkrankenhaus Kloster Grafschaft, Schmallenberg, Germany
| | - Thomas Staudinger
- Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Roman Ullrich
- Department of Anaesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
- Department of Anaesthesiology and Intensive Care Medicine, AUVA Trauma Center Vienna, Vienna, Austria
| | - Steffen Weber-Carstens
- Department of Anaesthesiology and Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Hermann Wrigge
- Department of Anaesthesiology, Intensive Care and Emergency Medicine, Pain Therapy, Bergmannstrost Hospital, Halle, Germany
- Medical Faculty, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | | | - Sina M Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.
- Septomics Research Center, Jena University Hospital, Jena, Germany.
| |
Collapse
|
4
|
McNelly A, Langan A, Bear DE, Page A, Martin T, Seidu F, Santos F, Rooney K, Liang K, Heales SJ, Baldwin T, Alldritt I, Crossland H, Atherton PJ, Wilkinson D, Montgomery H, Prowle J, Pearse R, Eaton S, Puthucheary ZA. A pilot study of alternative substrates in the critically Ill subject using a ketogenic feed. Nat Commun 2023; 14:8345. [PMID: 38102152 PMCID: PMC10724188 DOI: 10.1038/s41467-023-42659-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 10/18/2023] [Indexed: 12/17/2023] Open
Abstract
Bioenergetic failure caused by impaired utilisation of glucose and fatty acids contributes to organ dysfunction across multiple tissues in critical illness. Ketone bodies may form an alternative substrate source, but the feasibility and safety of inducing a ketogenic state in physiologically unstable patients is not known. Twenty-nine mechanically ventilated adults with multi-organ failure managed on intensive care units were randomised (Ketogenic n = 14, Control n = 15) into a two-centre pilot open-label trial of ketogenic versus standard enteral feeding. The primary endpoints were assessment of feasibility and safety, recruitment and retention rates and achievement of ketosis and glucose control. Ketogenic feeding was feasible, safe, well tolerated and resulted in ketosis in all patients in the intervention group, with a refusal rate of 4.1% and 82.8% retention. Patients who received ketogenic feeding had fewer hypoglycaemic events (0.0% vs. 1.6%), required less exogenous international units of insulin (0 (Interquartile range 0-16) vs.78 (Interquartile range 0-412) but had slightly more daily episodes of diarrhoea (53.5% vs. 42.9%) over the trial period. Ketogenic feeding was feasible and may be an intervention for addressing bioenergetic failure in critically ill patients. Clinical Trials.gov registration: NCT04101071.
Collapse
Affiliation(s)
- Angela McNelly
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Anne Langan
- Department of Dietetics, Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Danielle E Bear
- Department of Nutrition and Dietetics, St Thomas' NHS Foundation Trust, London, UK
- Department of Critical Care, Guy's and St. Thomas' NHS, London, UK
| | | | - Tim Martin
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Fatima Seidu
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Filipa Santos
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Kieron Rooney
- Department of Critical Care, Bristol Royal Infirmary, Bristol, UK
| | - Kaifeng Liang
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Simon J Heales
- Genetic & Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Tomas Baldwin
- Developmental Biology & Cancer, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Isabelle Alldritt
- Centre of Metabolism, Aging & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Aging Research & NIHR Nottingham BRC, University of Nottingham, Nottingham, UK
| | - Hannah Crossland
- Centre of Metabolism, Aging & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Aging Research & NIHR Nottingham BRC, University of Nottingham, Nottingham, UK
| | - Philip J Atherton
- Centre of Metabolism, Aging & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Aging Research & NIHR Nottingham BRC, University of Nottingham, Nottingham, UK
| | - Daniel Wilkinson
- Centre of Metabolism, Aging & Physiology (COMAP), MRC-Versus Arthritis Centre for Musculoskeletal Aging Research & NIHR Nottingham BRC, University of Nottingham, Nottingham, UK
| | - Hugh Montgomery
- University College London (UCL), London, UK
- UCL Hospitals NHS Foundation Trust (UCLH), National Institute for Health Research (NIHR) Biomedical Research Centre (BRC), London, UK
| | - John Prowle
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Rupert Pearse
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK
- Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Simon Eaton
- Developmental Biology & Cancer, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Zudin A Puthucheary
- William Harvey Research Institute, Faculty of Medicine & Dentistry, Queen Mary University of London, London, UK.
- Adult Critical Care Unit, Royal London Hospital, London, UK.
| |
Collapse
|
5
|
Maffiuletti NA, Dirks ML, Stevens-Lapsley J, McNeil CJ. Electrical stimulation for investigating and improving neuromuscular function in vivo: Historical perspective and major advances. J Biomech 2023; 152:111582. [PMID: 37088030 DOI: 10.1016/j.jbiomech.2023.111582] [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: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/25/2023]
Abstract
This historical review summarizes the major advances - particularly from the last 50 years - in transcutaneous motor-level electrical stimulation, which can be used either as a tool to investigate neuromuscular function and its determinants (electrical stimulation for testing; EST) or as a therapeutic/training modality to improve neuromuscular and physical function (neuromuscular electrical stimulation; NMES). We focus on some of the most important applications of electrical stimulation in research and clinical settings, such as the investigation of acute changes, chronic adaptations and pathological alterations of neuromuscular function with EST, as well as the enhancement, preservation and restoration of muscle strength and mass with NMES treatment programs in various populations. For both EST and NMES, several major advances converge around understanding and optimizing motor unit recruitment during electrically-evoked contractions, also taking into account the influence of stimulation site (e.g., muscle belly vs nerve trunk) and type (e.g., pulse duration, frequency, and intensity). This information is equally important both in the context of mechanistic research of neuromuscular function as well as for clinicians who believe that improvements in neuromuscular function are required to provide health-related benefits to their patients.
Collapse
Affiliation(s)
| | - Marlou L Dirks
- Department of Public Health and Sports Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK; Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Jennifer Stevens-Lapsley
- Physical Therapy Program, Department of Physical Medicine and Rehabilitation, University of Colorado, Aurora, CO, USA; VA Eastern Colorado Geriatric Research, Education, and Clinical Center (GRECC), VA Eastern Colorado Health Care System, Aurora, CO, USA
| | - Chris J McNeil
- Integrated Neuromuscular Physiology Laboratory, School of Health and Exercise Sciences, University of British Columbia, Kelowna, Canada
| |
Collapse
|
6
|
Grunow JJ, Gan T, Lewald H, Martyn JAJ, Blobner M, Schaller SJ. Insulin signaling in skeletal muscle during inflammation and/or immobilisation. Intensive Care Med Exp 2023; 11:16. [PMID: 36967414 PMCID: PMC10040391 DOI: 10.1186/s40635-023-00503-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/20/2023] [Indexed: 03/28/2023] Open
Abstract
BACKGROUND The decline in the downstream signal transduction pathway of anabolic hormone, insulin, could play a key role in the muscle atrophy and insulin resistance observed in patients with intensive care unit acquired weakness (ICUAW). This study investigated the impact of immobilisation via surgical knee and ankle fixation and inflammation via Corynebacterium parvum injection, alone and in combination, as risk factors for altering insulin transduction and, therefore, their role in ICUAW. RESULTS Muscle weight was significantly decreased due to immobilisation [estimated effect size (95% CI) - 0.10 g (- 0.12 to - 0.08); p < 0.001] or inflammation [estimated effect size (95% CI) - 0.11 g (- 0.13 to - 0.09); p < 0.001] with an additive effect of both combined (p = 0.024). pAkt was only detectable after insulin stimulation [estimated effect size (95% CI) 85.1-fold (76.2 to 94.0); p < 0.001] irrespective of the group and phosphorylation was not impaired by the different perturbations. Nevertheless, the phosphorylation of GSK3 observed in the control group after insulin stimulation was decreased in the immobilisation [estimated effect size (95% CI) - 40.2 (- 45.6 to - 34.8)] and inflammation [estimated effect size (95% CI) - 55.0 (- 60.4 to - 49.5)] groups. The expression of phosphorylated GS (pGS) was decreased after insulin stimulation in the control group and significantly increased in the immobilisation [estimated effect size (95% CI) 70.6-fold (58.8 to 82.4)] and inflammation [estimated effect size (95% CI) 96.7 (85.0 to 108.5)] groups. CONCLUSIONS Both immobilisation and inflammation significantly induce insulin resistance, i.e., impair the insulin signaling pathway downstream of Akt causing insufficient GSK phosphorylation and, therefore, its activation which caused increased glycogen synthase phosphorylation, which could contribute to muscle atrophy of immobilisation and inflammation.
Collapse
Affiliation(s)
- Julius J Grunow
- Charité - Universitätsmedizin Berlin, corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Operative Intensive Care Medicine (CVK, CCM), Charitéplatz 1, 10117, Berlin, Germany
| | - Thomas Gan
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Anesthesiology and Intensive Care, Ismaninger Straße 22, 81675, Munich, Bavaria, Germany
| | - Heidrun Lewald
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Anesthesiology and Intensive Care, Ismaninger Straße 22, 81675, Munich, Bavaria, Germany
| | - J A Jeevendra Martyn
- Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, and Harvard Medical School, 51 Blossom Street, Room 206, Boston, 02114, MA, USA
| | - Manfred Blobner
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Anesthesiology and Intensive Care, Ismaninger Straße 22, 81675, Munich, Bavaria, Germany
| | - Stefan J Schaller
- Charité - Universitätsmedizin Berlin, corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Operative Intensive Care Medicine (CVK, CCM), Charitéplatz 1, 10117, Berlin, Germany.
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Anesthesiology and Intensive Care, Ismaninger Straße 22, 81675, Munich, Bavaria, Germany.
| |
Collapse
|
7
|
Klawitter F, Ehler J, Bajorat R, Patejdl R. Mitochondrial Dysfunction in Intensive Care Unit-Acquired Weakness and Critical Illness Myopathy: A Narrative Review. Int J Mol Sci 2023; 24:5516. [PMID: 36982590 PMCID: PMC10052131 DOI: 10.3390/ijms24065516] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Mitochondria are key structures providing most of the energy needed to maintain homeostasis. They are the main source of adenosine triphosphate (ATP), participate in glucose, lipid and amino acid metabolism, store calcium and are integral components in various intracellular signaling cascades. However, due to their crucial role in cellular integrity, mitochondrial damage and dysregulation in the context of critical illness can severely impair organ function, leading to energetic crisis and organ failure. Skeletal muscle tissue is rich in mitochondria and, therefore, particularly vulnerable to mitochondrial dysfunction. Intensive care unit-acquired weakness (ICUAW) and critical illness myopathy (CIM) are phenomena of generalized weakness and atrophying skeletal muscle wasting, including preferential myosin breakdown in critical illness, which has also been linked to mitochondrial failure. Hence, imbalanced mitochondrial dynamics, dysregulation of the respiratory chain complexes, alterations in gene expression, disturbed signal transduction as well as impaired nutrient utilization have been proposed as underlying mechanisms. This narrative review aims to highlight the current known molecular mechanisms immanent in mitochondrial dysfunction of patients suffering from ICUAW and CIM, as well as to discuss possible implications for muscle phenotype, function and therapeutic approaches.
Collapse
Affiliation(s)
- Felix Klawitter
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Johannes Ehler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany
| | - Rika Bajorat
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Robert Patejdl
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
8
|
Sipahioglu H, Ilik HKZ, Ozer NT, Onuk S, Koyuncu S, Kuzuguden S, Elay G. Importance of Asprosin for Changes of M. Rectus Femoris Area during the Acute Phase of Medical Critical Illness: A Prospective Observational Study. Healthcare (Basel) 2023; 11:healthcare11050732. [PMID: 36900737 PMCID: PMC10000515 DOI: 10.3390/healthcare11050732] [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: 01/31/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Asprosin, a new adipokine, is secreted by subcutaneous white adipose tissue and causes rapid glucose release. The skeletal muscle mass gradually diminishes with aging. The combination of decreased skeletal muscle mass and critical illness may cause poor clinical outcomes in critically ill older adults. To determine the relationship between the serum asprosin level, fat-free mass, and nutritional status of critically ill older adult patients, critically ill patients over the age of 65 receiving enteral nutrition via feeding tube were included in the study. The patients' cross-sectional area of the rectus femoris (RF) of the lower extremity quadriceps muscle was evaluated by serial measurements. The mean age of the patients was 72 ± 6 years. The median (IQR) serum asprosin level was 31.8 (27.4-38.1) ng/mL on the first study day and 26.1 (23.4-32.3) ng/mL on the fourth study day. Serum asprosin level was high in 96% of the patients on the first day, and it was high in 74% on the fourth day after initiation of enteral feeding. The patients achieved 65.9 ± 34.1% of the daily energy requirement for four study days. A significant moderate correlation between delta serum asprosin level and delta RF was found (Rho = -0.369, p = 0.013). In critically ill older adult patients, a significant negative correlation was determined between serum asprosin level with energy adequacy and lean muscle mass.
Collapse
Affiliation(s)
- Hilal Sipahioglu
- Department of Intensive Care Unit, Kayseri City Training and Research Hospital, Kayseri 38080, Turkey
- Correspondence: ; Tel.: +90-035223-157700 (Ext. 11056)
| | | | - Nurhayat Tugra Ozer
- Department of Clinical Nutrition, Erciyes School of Medicine, Erciyes University, Kayseri 38080, Turkey
| | - Sevda Onuk
- Department of Intensive Care Unit, Kayseri City Training and Research Hospital, Kayseri 38080, Turkey
| | - Sumeyra Koyuncu
- Department of Nephrology, Kayseri City Training and Research Hospital, Kayseri 38080, Turkey
| | - Sibel Kuzuguden
- Department of Biochemistry, Kayseri City Training and Research Hospital, Kayseri 38080, Turkey
| | - Gulseren Elay
- Department of Intensive Care Unit, Gaziantep University, Gaziantep 27470, Turkey
| |
Collapse
|
9
|
Signals for Muscular Protein Turnover and Insulin Resistance in Critically Ill Patients: A Narrative Review. Nutrients 2023; 15:nu15051071. [PMID: 36904071 PMCID: PMC10005516 DOI: 10.3390/nu15051071] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Sarcopenia in critically ill patients is a highly prevalent comorbidity. It is associated with a higher mortality rate, length of mechanical ventilation, and probability of being sent to a nursing home after the Intensive Care Unit (ICU). Despite the number of calories and proteins delivered, there is a complex network of signals of hormones and cytokines that affect muscle metabolism and its protein synthesis and breakdown in critically ill and chronic patients. To date, it is known that a higher number of proteins decreases mortality, but the exact amount needs to be clarified. This complex network of signals affects protein synthesis and breakdown. Some hormones regulate metabolism, such as insulin, insulin growth factor glucocorticoids, and growth hormone, whose secretion is affected by feeding states and inflammation. In addition, cytokines are involved, such as TNF-alpha and HIF-1. These hormones and cytokines have common pathways that activate muscle breakdown effectors, such as the ubiquitin-proteasome system, calpain, and caspase-3. These effectors are responsible for protein breakdown in muscles. Many trials have been conducted with hormones with different results but not with nutritional outcomes. This review examines the effect of hormones and cytokines on muscles. Knowing all the signals and pathways that affect protein synthesis and breakdown can be considered for future therapeutics.
Collapse
|
10
|
Schneider J, Sundaravinayagam D, Blume A, Marg A, Grunwald S, Metzler E, Escobar H, Müthel S, Wang H, Wollersheim T, Weber-Carstens S, Akalin A, Di Virgilio M, Tursun B, Spuler S. Disintegration of the NuRD Complex in Primary Human Muscle Stem Cells in Critical Illness Myopathy. Int J Mol Sci 2023; 24:2772. [PMID: 36769095 PMCID: PMC9916927 DOI: 10.3390/ijms24032772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Critical illness myopathy (CIM) is an acquired, devastating, multifactorial muscle-wasting disease with incomplete recovery. The impact on hospital costs and permanent loss of quality of life is enormous. Incomplete recovery might imply that the function of muscle stem cells (MuSC) is impaired. We tested whether epigenetic alterations could be in part responsible. We characterized human muscle stem cells (MuSC) isolated from early CIM and analyzed epigenetic alterations (CIM n = 15, controls n = 21) by RNA-Seq, immunofluorescence, analysis of DNA repair, and ATAC-Seq. CIM-MuSC were transplanted into immunodeficient NOG mice to assess their regenerative potential. CIM-MuSC exhibited significant growth deficits, reduced ability to differentiate into myotubes, and impaired DNA repair. The chromatin structure was damaged, as characterized by alterations in mRNA of histone 1, depletion or dislocation of core proteins of nucleosome remodeling and deacetylase complex, and loosening of multiple nucleosome-spanning sites. Functionally, CIM-MuSC had a defect in building new muscle fibers. Further, MuSC obtained from the electrically stimulated muscle of CIM patients was very similar to control MuSC, indicating the impact of muscle contraction in the onset of CIM. CIM not only affects working skeletal muscle but has a lasting and severe epigenetic impact on MuSC.
Collapse
Affiliation(s)
- Joanna Schneider
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
- Charité Universitätsmedizin Berlin, Department of Pediatric Neurology, 13353 Berlin, Germany
- Berlin Institute of Health–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Devakumar Sundaravinayagam
- Laboratory of DNA Repair and Maintenance of Genome Stability, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13092 Berlin, Germany
| | - Alexander Blume
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Andreas Marg
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Stefanie Grunwald
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Eric Metzler
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Helena Escobar
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Stefanie Müthel
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Haicui Wang
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Tobias Wollersheim
- Berlin Institute of Health–Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité Universitätsmedizin Berlin, Department of Anesthesiology and Operative Intensive Care Medicine, 13353 Berlin, Germany
| | - Steffen Weber-Carstens
- Charité Universitätsmedizin Berlin, Department of Anesthesiology and Operative Intensive Care Medicine, 13353 Berlin, Germany
| | - Altuna Akalin
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Michela Di Virgilio
- Laboratory of DNA Repair and Maintenance of Genome Stability, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13092 Berlin, Germany
| | - Baris Tursun
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| |
Collapse
|
11
|
McClelland TJ, Fowler AJ, Davies TW, Pearse R, Prowle J, Puthucheary Z. Can pioglitazone be used for optimization of nutrition in critical illness? A systematic review. JPEN J Parenter Enteral Nutr 2023; 47:459-475. [PMID: 36700419 DOI: 10.1002/jpen.2481] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Skeletal muscle wasting is a determinant of physical disability in survivors of critical illness. Intramuscular bioenergetic failure, altered substrate metabolim, and inflammation are likely underpinning mechanisms. We examined the effect of pioglitazone, a peroxisome proliferator-activated receptor γ agonist, on muscle-related outcomes in adults. METHODS We included randomized controlled trials in which pioglitazone was administered (no dose/dosage restrictions) and muscle-related outcomes were reported. We searched MEDLINE, CENTRAL, EMBASE, CINAHL, and trial registries. Risk of bias was assessed using RoB 2. Primary outcomes were physical function and symptoms, muscle mass and function, or body composition and muscular compositional change. Secondary outcomes included muscle insulin sensitivity, mitochondrial effects, and intramuscular inflammation. RESULTS Fourteen studies over 19 publications (n = 474 patients) were included. Lean body mass was unaffected in three studies (n = 126) and increased by 1.8-1.92 kg in two studies (P = 0.02 and 0.003, respectively; n = 48). Pioglitazone was associated with increased peripheral insulin sensitivity (+23%-72%, standardized mean difference of 0.97 from trial start point to end point [95% CI, 0.36-1.58; n = 213]). Treatment reduced intramuscular tumor necrosis factor-α (TNF-α) levels (-30%; P = 0.02; n = 29), with mixed effects on serum TNF-α and intramyocellular lipid concentrations. Treatment increased intramuscular markers of adenosine triphosphate (ATP) biosynthesis (ATP5A [+33%, P ≤ 0.05], ETFA [+60%, P ≤ 0.05], and CX6B1 [+ 33%, P = 0.01] [n = 24]), PGC1α and PGC1β messenger RNA expression (P < 0.05; n = 26), and AMPK phosphorylation (+38%, P < 0.05; n = 26). These data have low-quality evidence profiles owing to risk of bias. CONCLUSIONS Pioglitazone therapy increases skeletal muscle insulin sensitivity and can decrease intramuscular inflammation.
Collapse
Affiliation(s)
- Thomas J McClelland
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Alexander J Fowler
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Thomas W Davies
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Rupert Pearse
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| | - John Prowle
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| | - Zudin Puthucheary
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| |
Collapse
|
12
|
Grunow JJ, Reiher K, Carbon NM, Engelhardt LJ, Mai K, Koch S, Schefold JC, Z’Graggen W, Schaller SJ, Fielitz J, Spranger J, Weber-Carstens S, Wollersheim T. Muscular myostatin gene expression and plasma concentrations are decreased in critically ill patients. Crit Care 2022; 26:237. [PMID: 35922829 PMCID: PMC9347123 DOI: 10.1186/s13054-022-04101-1] [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: 02/14/2022] [Accepted: 07/07/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The objective was to investigate the role of gene expression and plasma levels of the muscular protein myostatin in intensive care unit-acquired weakness (ICUAW). This was performed to evaluate a potential clinical and/or pathophysiological rationale of therapeutic myostatin inhibition.
Methods
A retrospective analysis from pooled data of two prospective studies to assess the dynamics of myostatin plasma concentrations (day 4, 8 and 14) and myostatin gene (MSTN) expression levels in skeletal muscle (day 15) was performed. Associations of myostatin to clinical and electrophysiological outcomes, muscular metabolism and muscular atrophy pathways were investigated.
Results
MSTN gene expression (median [IQR] fold change: 1.00 [0.68–1.54] vs. 0.26 [0.11–0.80]; p = 0.004) and myostatin plasma concentrations were significantly reduced in all critically ill patients when compared to healthy controls. In critically ill patients, myostatin plasma concentrations increased over time (median [IQR] fold change: day 4: 0.13 [0.08/0.21] vs. day 8: 0.23 [0.10/0.43] vs. day 14: 0.40 [0.26/0.61]; p < 0.001). Patients with ICUAW versus without ICUAW showed significantly lower MSTN gene expression levels (median [IQR] fold change: 0.17 [0.10/0.33] and 0.51 [0.20/0.86]; p = 0.047). Myostatin levels were directly correlated with muscle strength (correlation coefficient 0.339; p = 0.020) and insulin sensitivity index (correlation coefficient 0.357; p = 0.015). No association was observed between myostatin plasma concentrations as well as MSTN expression levels and levels of mobilization, electrophysiological variables, or markers of atrophy pathways.
Conclusion
Muscular gene expression and systemic protein levels of myostatin are downregulated during critical illness. The previously proposed therapeutic inhibition of myostatin does therefore not seem to have a pathophysiological rationale to improve muscle quality in critically ill patients.
Trial registration: ISRCTN77569430—13th of February 2008 and ISRCTN19392591 17th of February 2011.
Graphical abstract
Collapse
|
13
|
Engelhardt LJ, Carbon NM, Weber-Carstens S. [54/m-Muscle weakness and prolonged weaning from mechanical ventilation after peritonitis with septic shock : Preparation course anesthesiological intensive care medicine: case 29]. DIE ANAESTHESIOLOGIE 2022; 71:149-153. [PMID: 35941303 DOI: 10.1007/s00101-022-01166-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Lilian Jo Engelhardt
- Klinik für Anästhesiologie m.S. operative Intensivmedizin, Charité - Universitätsmedizin Berlin, Campus CVK&CCM, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Deutschland
- Institut für Medizinische Informatik, Charité - Universitätsmedizin Berlin, Campus CCM, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Deutschland
| | - Niklas M Carbon
- Klinik für Anästhesiologie m.S. operative Intensivmedizin, Charité - Universitätsmedizin Berlin, Campus CVK&CCM, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Deutschland
| | - Steffen Weber-Carstens
- Klinik für Anästhesiologie m.S. operative Intensivmedizin, Charité - Universitätsmedizin Berlin, Campus CVK&CCM, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353, Berlin, Deutschland.
| |
Collapse
|
14
|
Fernandes JA, Silva MLD, Trancho AC, Macedo JRDD, Martins HR, Silva PE. Assessment of neuromuscular electrical stimulation in critically ill patients: physical therapists’ knowledge and barriers to its use. FISIOTERAPIA E PESQUISA 2022. [DOI: 10.1590/1809-2950/21003529032022en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
ABSTRACT Transcutaneous neuromuscular electrical stimulation (NMES) is considered an important tool to prevent muscle mass and strength loss in patients admitted to intensive care units (ICU). This study aimed to evaluate physical therapists’ profile and knowledge of NMES and identify the main barriers to its use in ICUs. This observational cross-sectional study was conducted via a structured questionnaire created by the authors. It consisted of 12 objective questions to analyze physical therapists’ knowledge of NMES use in critically ill patients. Physical therapists were invited to participate in this study during an international symposium on NMES. In total, 56 physical therapists, with a mean age of 33.5±7.2 years and working an average of 9.7±7 years after graduation, completed the survey. Overall, 34 respondents worked in ICUs, of which only four (12%) reported regular NMES use in their ICUs. We found a low average of correct answers to our questionnaire (25%; 3/12). The main barriers reported to using NMES in ICUs were lack of knowledge (28; 50%) and equipment (24; 43%). The number of correct answers expert and non-expert physical therapists was not statistically significant (p=0.68). Thus, we observed participants’ poor knowledge of NMES use in critically ill patients. Respondents showed that NMES has been underused in their ICUs. Lack of knowledge and equipment seems to be the main barriers for the use of NMES in ICUs.
Collapse
|
15
|
Addinsall AB, Cacciani N, Backéus A, Hedström Y, Shevchenko G, Bergquist J, Larsson L. Electrical stimulated GLUT4 signalling attenuates critical illness-associated muscle wasting. J Cachexia Sarcopenia Muscle 2022; 13:2162-2174. [PMID: 35502572 PMCID: PMC9397497 DOI: 10.1002/jcsm.12978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 02/12/2022] [Accepted: 02/21/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Critical illness myopathy (CIM) is a debilitating condition characterized by the preferential loss of the motor protein myosin. CIM is a by-product of critical care, attributed to impaired recovery, long-term complications, and mortality. CIM pathophysiology is complex, heterogeneous and remains incompletely understood; however, loss of mechanical stimuli contributes to critical illness-associated muscle atrophy and weakness. Passive mechanical loading and electrical stimulation (ES) therapies augment muscle mass and function. While having beneficial outcomes, the mechanistic underpinning of these therapies is less known. Therefore, here we aimed to assess the mechanism by which chronic supramaximal ES ameliorates CIM in a unique experimental rat model of critical care. METHODS Rats were subjected to 8 days of critical care conditions entailing deep sedation, controlled mechanical ventilation, and immobilization with and without direct soleus ES. Muscle size and function were assessed at the single cell level. RNAseq and western blotting were employed to understand the mechanisms driving ES muscle outcomes in CIM. RESULTS Following 8 days of controlled mechanical ventilation and immobilization, soleus muscle mass, myosin : actin ratio, and single muscle fibre maximum force normalized to cross-sectional area (CSA; specific force) were reduced by 40-50% (P < 0.0001). ES significantly reduced the loss of soleus muscle fibre CSA and myosin : actin ratio by approximately 30% (P < 0.05) yet failed to effect specific force. RNAseq pathway analysis revealed downregulation of insulin signalling in the soleus muscle following critical care, and GLUT4 trafficking was reduced by 55% leading to an 85% reduction of muscle glycogen content (P < 0.01). ES promoted phosphofructokinase and insulin signalling pathways to control levels (P < 0.05), consistent with the maintenance of GLUT4 translocation and glycogen levels. AMPK, but not AKT, signalling pathway was stimulated following ES, where the downstream target TBC1D4 increased 3 logFC (P = 0.029) and AMPK-specific P-TBC1D4 levels were increased approximately two-fold (P = 0.06). Reduction of muscle protein degradation rather than increased synthesis promoted soleus CSA, as ES reduced E3 ubiquitin proteins, Atrogin-1 (P = 0.006) and MuRF1 (P = 0.08) by approximately 50%, downstream of AMPK-FoxO3. CONCLUSIONS ES maintained GLUT4 translocation through increased AMPK-TBC1D4 signalling leading to improved muscle glucose homeostasis. Soleus CSA and myosin content was promoted through reduced protein degradation via AMPK-FoxO3 E3 ligases, Atrogin-1 and MuRF1. These results demonstrate chronic supramaximal ES reduces critical care associated muscle wasting, preserved glucose signalling, and reduced muscle protein degradation in CIM.
Collapse
Affiliation(s)
- Alex B. Addinsall
- Basic and Clinical Muscle Biology Group, Department of Physiology and PharmacologyKarolinska InstituteSolnaSweden
| | - Nicola Cacciani
- Basic and Clinical Muscle Biology Group, Department of Physiology and PharmacologyKarolinska InstituteSolnaSweden
- Department of Clinical NeuroscienceKarolinska InstituteSolnaSweden
| | - Anders Backéus
- Basic and Clinical Muscle Biology Group, Department of Physiology and PharmacologyKarolinska InstituteSolnaSweden
| | - Yvette Hedström
- Basic and Clinical Muscle Biology Group, Department of Physiology and PharmacologyKarolinska InstituteSolnaSweden
| | - Ganna Shevchenko
- Department of Chemistry – BMC, Analytical ChemistryUppsala UniversityUppsalaSweden
| | - Jonas Bergquist
- Department of Chemistry – BMC, Analytical ChemistryUppsala UniversityUppsalaSweden
| | - Lars Larsson
- Basic and Clinical Muscle Biology Group, Department of Physiology and PharmacologyKarolinska InstituteSolnaSweden
- Department of Clinical NeuroscienceKarolinska InstituteSolnaSweden
- Viron Molecular Medicine InstituteBostonUSA
| |
Collapse
|
16
|
Balke M, Teschler M, Schäfer H, Pape P, Mooren FC, Schmitz B. Therapeutic Potential of Electromyostimulation (EMS) in Critically Ill Patients—A Systematic Review. Front Physiol 2022; 13:865437. [PMID: 35615672 PMCID: PMC9124773 DOI: 10.3389/fphys.2022.865437] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/01/2022] [Indexed: 12/15/2022] Open
Abstract
Ample evidence exists that intensive care unit (ICU) treatment and invasive ventilation induce a transient or permanent decline in muscle mass and function. The functional deficit is often called ICU-acquired weakness with critical illness polyneuropathy (CIP) and/or myopathy (CIM) being the major underlying causes. Histopathological studies in ICU patients indicate loss of myosin filaments, muscle fiber necrosis, atrophy of both muscle fiber types as well as axonal degeneration. Besides medical prevention of risk factors such as sepsis, hyperglycemia and pneumonia, treatment is limited to early passive and active mobilization and one third of CIP/CIM patients discharged from ICU never regain their pre-hospitalization constitution. Electromyostimulation [EMS, also termed neuromuscular electrical stimulation (NMES)] is known to improve strength and function of healthy and already atrophied muscle, and may increase muscle blood flow and induce angiogenesis as well as beneficial systemic vascular adaptations. This systematic review aimed to investigate evidence from randomized controlled trails (RCTs) on the efficacy of EMS to improve the condition of critically ill patients treated on ICU. A systematic search of the literature was conducted using PubMed (Medline), CENTRAL (including Embase and CINAHL), and Google Scholar. Out of 1,917 identified records, 26 articles (1,312 patients) fulfilled the eligibility criteria of investigating at least one functional measure including muscle function, functional independence, or weaning outcomes using a RCT design in critically ill ICU patients. A qualitative approach was used, and results were structured by 1) stimulated muscles/muscle area (quadriceps muscle only; two to four leg muscle groups; legs and arms; chest and abdomen) and 2) treatment duration (≤10 days, >10 days). Stimulation parameters (impulse frequency, pulse width, intensity, duty cycle) were also collected and the net EMS treatment time was calculated. A high grade of heterogeneity between studies was detected with major cofactors being the analyzed patient group and selected outcome variable. The overall efficacy of EMS was inconclusive and neither treatment duration, stimulation site or net EMS treatment time had clear effects on study outcomes. Based on our findings, we provide practical recommendations and suggestions for future studies investigating the therapeutic efficacy of EMS in critically ill patients. Systematic Review Registration: [https://www.crd.york.ac.uk/prospero/], identifier [CRD42021262287].
Collapse
Affiliation(s)
- Maryam Balke
- St. Marien Hospital Cologne, Department of Early Rehabilitation, Cologne, Germany
- Department of Rehabilitation Sciences, Faculty of Health, University of Witten/Herdecke, Witten, Germany
- *Correspondence: Maryam Balke,
| | - Marc Teschler
- Department of Rehabilitation Sciences, Faculty of Health, University of Witten/Herdecke, Witten, Germany
- DRV Clinic Königsfeld, Center for Medical Rehabilitation, Ennepetal, Germany
| | - Hendrik Schäfer
- Department of Rehabilitation Sciences, Faculty of Health, University of Witten/Herdecke, Witten, Germany
- DRV Clinic Königsfeld, Center for Medical Rehabilitation, Ennepetal, Germany
| | - Pantea Pape
- St. Marien Hospital Cologne, Department of Early Rehabilitation, Cologne, Germany
| | - Frank C. Mooren
- Department of Rehabilitation Sciences, Faculty of Health, University of Witten/Herdecke, Witten, Germany
- DRV Clinic Königsfeld, Center for Medical Rehabilitation, Ennepetal, Germany
| | - Boris Schmitz
- Department of Rehabilitation Sciences, Faculty of Health, University of Witten/Herdecke, Witten, Germany
- DRV Clinic Königsfeld, Center for Medical Rehabilitation, Ennepetal, Germany
| |
Collapse
|
17
|
Kny M, Fielitz J. Hidden Agenda - The Involvement of Endoplasmic Reticulum Stress and Unfolded Protein Response in Inflammation-Induced Muscle Wasting. Front Immunol 2022; 13:878755. [PMID: 35615361 PMCID: PMC9124858 DOI: 10.3389/fimmu.2022.878755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Critically ill patients at the intensive care unit (ICU) often develop a generalized weakness, called ICU-acquired weakness (ICUAW). A major contributor to ICUAW is muscle atrophy, a loss of skeletal muscle mass and function. Skeletal muscle assures almost all of the vital functions of our body. It adapts rapidly in response to physiological as well as pathological stress, such as inactivity, immobilization, and inflammation. In response to a reduced workload or inflammation muscle atrophy develops. Recent work suggests that adaptive or maladaptive processes in the endoplasmic reticulum (ER), also known as sarcoplasmic reticulum, contributes to this process. In muscle cells, the ER is a highly specialized cellular organelle that assures calcium homeostasis and therefore muscle contraction. The ER also assures correct folding of proteins that are secreted or localized to the cell membrane. Protein folding is a highly error prone process and accumulation of misfolded or unfolded proteins can cause ER stress, which is counteracted by the activation of a signaling network known as the unfolded protein response (UPR). Three ER membrane residing molecules, protein kinase R-like endoplasmic reticulum kinase (PERK), inositol requiring protein 1a (IRE1a), and activating transcription factor 6 (ATF6) initiate the UPR. The UPR aims to restore ER homeostasis by reducing overall protein synthesis and increasing gene expression of various ER chaperone proteins. If ER stress persists or cannot be resolved cell death pathways are activated. Although, ER stress-induced UPR pathways are known to be important for regulation of skeletal muscle mass and function as well as for inflammation and immune response its function in ICUAW is still elusive. Given recent advances in the development of ER stress modifying molecules for neurodegenerative diseases and cancer, it is important to know whether or not therapeutic interventions in ER stress pathways have favorable effects and these compounds can be used to prevent or treat ICUAW. In this review, we focus on the role of ER stress-induced UPR in skeletal muscle during critical illness and in response to predisposing risk factors such as immobilization, starvation and inflammation as well as ICUAW treatment to foster research for this devastating clinical problem.
Collapse
Affiliation(s)
- Melanie Kny
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Jens Fielitz
- Department of Molecular Cardiology, DZHK (German Center for Cardiovascular Research), Partner Site, Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
18
|
Carbon NM, Engelhardt LJ, Wollersheim T, Grunow JJ, Spies CD, Märdian S, Mai K, Spranger J, Weber-Carstens S. Impact of protocol-based physiotherapy on insulin sensitivity and peripheral glucose metabolism in critically ill patients. J Cachexia Sarcopenia Muscle 2022; 13:1045-1053. [PMID: 35075782 PMCID: PMC8978012 DOI: 10.1002/jcsm.12920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 11/22/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The impact of physiotherapy on insulin sensitivity and peripheral glucose metabolism in critically ill patients is not well understood. METHODS This pooled analysis investigates the impact of different physiotherapeutic strategies on insulin sensitivity in critically ill patients. We pooled data from two previous trials in adult patients with sequential organ failure assessment score (SOFA)≥ 9 within 72 h of intensive care unit (ICU) admission, who received hyperinsulinaemic euglycaemic (HE) clamps. Patients were divided into three groups: standard physiotherapy (sPT, n = 22), protocol-based physiotherapy (pPT, n = 8), and pPT with added muscle activating measures (pPT+, n = 20). Insulin sensitivity index (ISI) was determined by HE clamp. Muscle metabolites lactate, pyruvate, and glycerol were measured in the M. vastus lateralis via microdialysis during the HE clamp. Histochemical visualization of glucose transporter-4 (GLUT4) translocation was performed in surgically extracted muscle biopsies. All data are reported as median (25th/75th percentile) (trial registry: ISRCTN77569430 and ISRCTN19392591/ethics approval: Charité-EA2/061/06 and Charité-EA2/041/10). RESULTS Fifty critically ill patients (admission SOFA 13) showed markedly decreased ISIs on Day 17 (interquartile range) 0.029 (0.022/0.048) (mg/min/kg)/(mU/L) compared with healthy controls 0.103 (0.087/0.111), P < 0.001. ISI correlated with muscle strength measured by medical research council (MRC) score at first awakening (r = 0.383, P = 0.026) and at ICU discharge (r = 0.503, P = 0.002). Different physiotherapeutic strategies showed no effect on the ISI [sPT 0.029 (0.019/0.053) (mg/min/kg)/(mU/L) vs. pPT 0.026 (0.023/0.041) (mg/min/kg)/(mU/L) vs. pPT+ 0.029 (0.023/0.042) (mg/min/kg)/(mU/L); P = 0.919]. Regardless of the physiotherapeutic strategy metabolic flexibility was reduced. Relative change of lactate/pyruvate ratio during HE clamp is as follows: sPT 0.09 (-0.13/0.27) vs. pPT 0.07 (-0.16/0.31) vs. pPT+ -0.06 (-0.19/0.16), P = 0.729, and relative change of glycerol concentration: sPT -0.39 (-0.8/-0.12) vs. pPT -0.21 (-0.33/0.07) vs. pPT+ -0.21 (-0.44/-0.03), P = 0.257. The majority of ICU patients showed abnormal localization of GLUT4 with membranous GLUT4 distribution in 37.5% (3 of 8) of ICU patients receiving sPT, in 42.9% (3 of 7) of ICU patients receiving pPT, and in 53.8% (7 of 13) of ICU patients receiving pPT+ (no statistical testing possible). CONCLUSIONS Our data suggest that a higher duration of muscle activating measures had no impact on insulin sensitivity or metabolic flexibility in critically ill patients with sepsis-related multiple organ failure.
Collapse
Affiliation(s)
- Niklas M Carbon
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lilian J Engelhardt
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tobias Wollersheim
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julius J Grunow
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia D Spies
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sven Märdian
- Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Knut Mai
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Steffen Weber-Carstens
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
19
|
Grunwald SA, Haafke S, Grieben U, Kassner U, Steinhagen-Thiessen E, Spuler S. Statins Aggravate the Risk of Insulin Resistance in Human Muscle. Int J Mol Sci 2022; 23:2398. [PMID: 35216514 PMCID: PMC8876152 DOI: 10.3390/ijms23042398] [Citation(s) in RCA: 3] [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: 01/29/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 01/23/2023] Open
Abstract
Beside their beneficial effects on cardiovascular events, statins are thought to contribute to insulin resistance and type-2 diabetes. It is not known whether these effects are long-term events from statin-treatment or already triggered with the first statin-intake. Skeletal muscle is considered the main site for insulin-stimulated glucose uptake and therefore, a primary target for insulin resistance in the human body. We analyzed localization and expression of proteins related to GLUT4 mediated glucose uptake via AMPKα or AKT in human skeletal muscle tissue from patients with statin-intake >6 months and in primary human myotubes after 96 h statin treatment. The ratio for AMPKα activity significantly increased in human skeletal muscle cells treated with statins for long- and short-term. Furthermore, the insulin-stimulated counterpart, AKT, significantly decreased in activity and protein level, while GSK3ß and mTOR protein expression reduced in statin-treated primary human myotubes, only. However, GLUT4 was normally distributed whereas CAV3 was internalized from plasma membrane around the nucleus in statin-treated primary human myotubes. Statin-treatment activates AMPKα-dependent glucose uptake and remains active after long-term statin treatment. Permanent blocking of its insulin-dependent counterpart AKT activation may lead to metabolic inflexibility and insulin resistance in the long run and may be a direct consequence of statin-treatment.
Collapse
Affiliation(s)
- Stefanie A. Grunwald
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Stefanie Haafke
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Ulrike Grieben
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| | - Ursula Kassner
- Interdisciplinary Lipid Metabolic Center, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (U.K.); (E.S.-T.)
| | - Elisabeth Steinhagen-Thiessen
- Interdisciplinary Lipid Metabolic Center, Charité Universitätsmedizin Berlin, 13353 Berlin, Germany; (U.K.); (E.S.-T.)
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; (S.H.); (U.G.)
| |
Collapse
|
20
|
Engelhardt LJ, Grunow JJ, Wollersheim T, Carbon NM, Balzer F, Spranger J, Weber-Carstens S. Sex-Specific Aspects of Skeletal Muscle Metabolism in the Clinical Context of Intensive Care Unit-Acquired Weakness. J Clin Med 2022; 11:jcm11030846. [PMID: 35160299 PMCID: PMC8836746 DOI: 10.3390/jcm11030846] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/22/2022] [Accepted: 02/03/2022] [Indexed: 02/08/2023] Open
Abstract
(1) Background: Female sex is considered a risk factor for Intensive Care Unit-Acquired Weakness (ICUAW). The aim is to investigate sex-specific aspects of skeletal muscle metabolism in the context of ICUAW. (2) Methods: This is a sex-specific sub-analysis from two prospectively conducted trials examining skeletal muscle metabolism and advanced muscle activating measures in critical illness. Muscle strength was assessed by Medical Research Council Score. The insulin sensitivity index was analyzed by hyperinsulinemic-euglycemic (HE) clamp. Muscular metabolites were studied by microdialysis. M. vastus lateralis biopsies were taken. The molecular analysis included protein degradation pathways. Morphology was assessed by myocyte cross-sectional area (MCSA). Multivariable linear regression models for the effect of sex on outcome parameters were performed. (3) Results: n = 83 (♂n = 57, 68.7%; ♀n = 26, 31.3%) ICU patients were included. ICUAW was present in 81.1%♂ and in 82.4%♀ at first awakening (p = 0.911) and in 59.5%♂ and in 70.6%♀ at ICU discharge (p = 0.432). Insulin sensitivity index was reduced more in women than in men (p = 0.026). Sex was significantly associated with insulin sensitivity index and MCSA of Type IIa fibers in the adjusted regression models. (4) Conclusion: This hypothesis-generating analysis suggests that more pronounced impairments in insulin sensitivity and lower MCSA of Type IIa fibers in critically ill women may be relevant for sex differences in ICUAW.
Collapse
Affiliation(s)
- Lilian Jo Engelhardt
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.J.E.); (J.J.G.); (T.W.); (N.M.C.)
- Institute of Medical Informatics, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany;
| | - Julius J. Grunow
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.J.E.); (J.J.G.); (T.W.); (N.M.C.)
| | - Tobias Wollersheim
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.J.E.); (J.J.G.); (T.W.); (N.M.C.)
| | - Niklas M. Carbon
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.J.E.); (J.J.G.); (T.W.); (N.M.C.)
| | - Felix Balzer
- Institute of Medical Informatics, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany;
| | - Joachim Spranger
- Department of Endocrinology and Metabolic Diseases, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany;
| | - Steffen Weber-Carstens
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (L.J.E.); (J.J.G.); (T.W.); (N.M.C.)
- Correspondence:
| |
Collapse
|
21
|
Zanders L, Kny M, Hahn A, Schmidt S, Wundersitz S, Todiras M, Lahmann I, Bandyopadhyay A, Wollersheim T, Kaderali L, Luft FC, Birchmeier C, Weber-Carstens S, Fielitz J. Sepsis induces interleukin 6, gp130/JAK2/STAT3, and muscle wasting. J Cachexia Sarcopenia Muscle 2022; 13:713-727. [PMID: 34821076 PMCID: PMC8818599 DOI: 10.1002/jcsm.12867] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sepsis and inflammation can cause intensive care unit-acquired weakness (ICUAW). Increased interleukin-6 (IL-6) plasma levels are a risk factor for ICUAW. IL-6 signalling involves the glycoprotein 130 (gp130) receptor and the JAK/STAT-pathway, but its role in sepsis-induced muscle wasting is uncertain. In a clinical observational study, we found that the IL-6 target gene, SOCS3, was increased in skeletal muscle of ICUAW patients indicative for JAK/STAT-pathway activation. We tested the hypothesis that the IL-6/gp130-pathway mediates ICUAW muscle atrophy. METHODS We sequenced RNA (RNAseq) from tibialis anterior (TA) muscle of cecal ligation and puncture-operated (CLP) and sham-operated wildtype (WT) mice. The effects of the IL-6/gp130/JAK2/STAT3-pathway were investigated by analysing the atrophy phenotype, gene expression, and protein contents of C2C12 myotubes. Mice lacking Il6st, encoding gp130, in myocytes (cKO) and WT controls, as well as mice treated with the JAK2 inhibitor AG490 or vehicle were exposed to CLP or sham surgery for 24 or 96 h. RESULTS Analyses of differentially expressed genes in RNAseq (≥2-log2-fold change, P < 0.01) revealed an activation of IL-6-signalling and JAK/STAT-signalling pathways in muscle of septic mice, which occurred after 24 h and lasted at least for 96 h during sepsis. IL-6 treatment of C2C12 myotubes induced STAT3 phosphorylation (three-fold, P < 0.01) and Socs3 mRNA expression (3.1-fold, P < 0.01) and caused myotube atrophy. Knockdown of Il6st diminished IL-6-induced STAT3 phosphorylation (-30.0%; P < 0.01), Socs3 mRNA expression, and myotube atrophy. JAK2 (- 29.0%; P < 0.01) or STAT3 inhibition (-38.7%; P < 0.05) decreased IL-6-induced Socs3 mRNA expression. Treatment with either inhibitor attenuated myotube atrophy in response to IL-6. CLP-operated septic mice showed an increased STAT3 phosphorylation and Socs3 mRNA expression in TA muscle, which was reduced in septic Il6st-cKO mice by 67.8% (P < 0.05) and 85.6% (P < 0.001), respectively. CLP caused a loss of TA muscle weight, which was attenuated in Il6st-cKO mice (WT: -22.3%, P < 0.001, cKO: -13.5%, P < 0.001; WT vs. cKO P < 0.001). While loss of Il6st resulted in a reduction of MuRF1 protein contents, Atrogin-1 remained unchanged between septic WT and cKO mice. mRNA expression of Trim63/MuRF1 and Fbxo32/Atrogin-1 were unaltered between CLP-treated WT and cKO mice. AG490 treatment reduced STAT3 phosphorylation (-22.2%, P < 0.05) and attenuated TA muscle atrophy in septic mice (29.6% relative reduction of muscle weight loss, P < 0.05). The reduction in muscle atrophy was accompanied by a reduction in Fbxo32/Atrogin-1-mRNA (-81.3%, P < 0.05) and Trim63/MuRF1-mRNA expression (-77.6%, P < 0.05) and protein content. CONCLUSIONS IL-6 via the gp130/JAK2/STAT3-pathway mediates sepsis-induced muscle atrophy possibly contributing to ICUAW.
Collapse
Affiliation(s)
- Lukas Zanders
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Department of Cardiology, Charité Campus Benjamin Franklin, Berlin, Germany
| | - Melanie Kny
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Alexander Hahn
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sibylle Schmidt
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sebastian Wundersitz
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Mihail Todiras
- Cardiovascular hormones, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Nicolae Testemiţanu State University of Medicine and Pharmacy, Chișinău, Moldova
| | - Ines Lahmann
- Developmental Biology/Signal Transduction, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Arnab Bandyopadhyay
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | - Tobias Wollersheim
- Anesthesiology and operative Intensive Care Medicine, Charité Campus Virchow and Campus Mitte, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Lars Kaderali
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Steffen Weber-Carstens
- Anesthesiology and operative Intensive Care Medicine, Charité Campus Virchow and Campus Mitte, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Jens Fielitz
- Experimental and Clinical Research Center (ECRC), Charité-Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany.,Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
22
|
Metzing UB, von Loeffelholz C, Steidl R, Romeike B, Winkler R, Rauchfuß F, Settmacher U, Stoppe C, Coldewey SM, Weinmann C, Weickert MO, Claus RA, Birkenfeld AL, Kosan C, Horn P. Endoplasmic reticulum stress and the unfolded protein response in skeletal muscle of subjects suffering from peritoneal sepsis. Sci Rep 2022; 12:504. [PMID: 35017615 PMCID: PMC8752775 DOI: 10.1038/s41598-021-04517-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
We provide a descriptive characterization of the unfolded protein response (UPR) in skeletal muscle of human patients with peritoneal sepsis and a sepsis model of C57BL/6J mice. Patients undergoing open surgery were included in a cross-sectional study and blood and skeletal muscle samples were taken. Key markers of the UPR and cluster of differentiation 68 (CD68) as surrogate of inflammatory injury were evaluated by real-time PCR and histochemical staining. CD68 mRNA increased with sepsis in skeletal muscle of patients and animals (p < 0.05). Mainly the inositol-requiring enzyme 1α branch of the UPR was upregulated as shown by elevated X-box binding-protein 1 (XBP1u) and its spliced isoform (XBP1s) mRNA (p < 0.05, respectively). Increased expression of Gadd34 indicated activation of PRKR-Like Endoplasmic Reticulum Kinase (PERK) branch of the UPR, and was only observed in mice (p < 0.001) but not human study subjects. Selected cell death signals were upregulated in human and murine muscle, demonstrated by increased bcl-2 associated X protein mRNA and TUNEL staining (p < 0.05). In conclusion we provide a first characterization of the UPR in skeletal muscle in human sepsis.
Collapse
Affiliation(s)
- Uta Barbara Metzing
- Department of Trauma, Hand and Reconstructive Surgery, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Christian von Loeffelholz
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany.
| | - Ricardo Steidl
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
- Department of Anaesthesiology, Intensive Care, Pain Medicine and Emergency Medicine, Bundeswehrkrankenhaus Berlin, Berlin, Germany
| | - Bernd Romeike
- Section of Neuropathology, Department of Pathology, Jena University Hospital, Jena, Germany
- Dean's Office, Medical Didactics, University Rostock Medical Center, Rostock, Germany
| | - René Winkler
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, Jena, Germany
| | - Falk Rauchfuß
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | - Christian Stoppe
- Department of Anesthesiology and Intensive Care Medicine Wuerzburg, University Hospital, Wuerzburg, Germany
- 3CARE-Cardiovascular Critical Care & Anesthesia Evaluation and Research, Medical Faculty RWTH Aachen, Aachen, Germany
| | - Sina M Coldewey
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
- Septomics Research Centre, Jena University Hospital, Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Claudia Weinmann
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | - Martin O Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
- Translational and Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
- Centre of Applied Biological and Exercise Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, UK
| | - Ralf A Claus
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, 07747, Jena, Germany
| | - Andreas L Birkenfeld
- Department of Diabetology Endocrinology and Nephrology, Internal Medicine IV, University Hospital Tübingen, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
- Division of Translational Diabetology, Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
- Department of Diabetes, School of Life Course Science and Medicine, Kings College London, London, UK
| | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, Jena, Germany
| | - Paul Horn
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Department of Internal Medicine IV, Gastroenterology, Hepatology and Infectious Diseases, Jena University Hospital, Jena, Germany
| |
Collapse
|
23
|
Haberecht-Müller S, Krüger E, Fielitz J. Out of Control: The Role of the Ubiquitin Proteasome System in Skeletal Muscle during Inflammation. Biomolecules 2021; 11:biom11091327. [PMID: 34572540 PMCID: PMC8468834 DOI: 10.3390/biom11091327] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
The majority of critically ill intensive care unit (ICU) patients with severe sepsis develop ICU-acquired weakness (ICUAW) characterized by loss of muscle mass, reduction in myofiber size and decreased muscle strength leading to persisting physical impairment. This phenotype results from a dysregulated protein homeostasis with increased protein degradation and decreased protein synthesis, eventually causing a decrease in muscle structural proteins. The ubiquitin proteasome system (UPS) is the predominant protein-degrading system in muscle that is activated during diverse muscle atrophy conditions, e.g., inflammation. The specificity of UPS-mediated protein degradation is assured by E3 ubiquitin ligases, such as atrogin-1 and MuRF1, which target structural and contractile proteins, proteins involved in energy metabolism and transcription factors for UPS-dependent degradation. Although the regulation of activity and function of E3 ubiquitin ligases in inflammation-induced muscle atrophy is well perceived, the contribution of the proteasome to muscle atrophy during inflammation is still elusive. During inflammation, a shift from standard- to immunoproteasome was described; however, to which extent this contributes to muscle wasting and whether this changes targeting of specific muscular proteins is not well described. This review summarizes the function of the main proinflammatory cytokines and acute phase response proteins and their signaling pathways in inflammation-induced muscle atrophy with a focus on UPS-mediated protein degradation in muscle during sepsis. The regulation and target-specificity of the main E3 ubiquitin ligases in muscle atrophy and their mode of action on myofibrillar proteins will be reported. The function of the standard- and immunoproteasome in inflammation-induced muscle atrophy will be described and the effects of proteasome-inhibitors as treatment strategies will be discussed.
Collapse
Affiliation(s)
- Stefanie Haberecht-Müller
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
- Correspondence: (E.K.); (J.F.)
| | - Jens Fielitz
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
- Correspondence: (E.K.); (J.F.)
| |
Collapse
|
24
|
Langer HT, Mossakowski AA, Avey AM, Wohlgemuth RP, Smith LR, Zbinden-Foncea H, Baar K. A mutation in desmin makes skeletal muscle less vulnerable to acute muscle damage after eccentric loading in rats. FASEB J 2021; 35:e21860. [PMID: 34411340 PMCID: PMC9292853 DOI: 10.1096/fj.202100711rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/15/2021] [Accepted: 08/02/2021] [Indexed: 01/23/2023]
Abstract
Desminopathy is the most common intermediate filament disease in humans. The most frequent mutation causing desminopathy in patients is a R350P DES missense mutation. We have developed a rat model with an analogous mutation in R349P Des. To investigate the role of R349P Des in mechanical loading, we stimulated the sciatic nerve of wild‐type littermates (WT) (n = 6) and animals carrying the mutation (MUT) (n = 6) causing a lengthening contraction of the dorsi flexor muscles. MUT animals showed signs of ongoing regeneration at baseline as indicated by a higher number of central nuclei (genotype: P < .0001). While stimulation did not impact central nuclei, we found an increased number of IgG positive fibers (membrane damage indicator) after eccentric contractions with both genotypes (stimulation: P < .01). Interestingly, WT animals displayed a more pronounced increase in IgG positive fibers with stimulation compared to MUT (interaction: P < .05). In addition to altered histology, molecular signaling on the protein level differed between WT and MUT. The membrane repair protein dysferlin decreased with eccentric loading in WT but increased in MUT (interaction: P < .05). The autophagic substrate p62 was increased in both genotypes with loading (stimulation: P < .05) but tended to be more elevated in WT (interaction: P = .05). Caspase 3 levels, a central regulator of apoptotic cell death, was increased with stimulation in both genotypes (stimulation: P < .01) but more so in WT animals (interaction: P < .0001). Overall, our data indicate that R349P Des rats have a lower susceptibility to structural muscle damage of the cytoskeleton and sarcolemma with acute eccentric loading.
Collapse
Affiliation(s)
- Henning T Langer
- Functional Molecular Biology Laboratory, Department of Physiology and Membrane Biology, University of California, Davis, California, USA
| | - Agata A Mossakowski
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alec M Avey
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Ross P Wohlgemuth
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Lucas R Smith
- Neurobiology, Physiology and Behavior, University of California, Davis, California, USA
| | - Herman Zbinden-Foncea
- Exercise Physiology Laboratory, School of Kinesiology, Universidad Finis Terrae, Santiago, Chile
| | - Keith Baar
- Functional Molecular Biology Laboratory, Department of Physiology and Membrane Biology, University of California, Davis, California, USA.,Neurobiology, Physiology and Behavior, University of California, Davis, California, USA.,VA Northern California Health Care System, Mather, California, USA
| |
Collapse
|
25
|
Preau S, Vodovar D, Jung B, Lancel S, Zafrani L, Flatres A, Oualha M, Voiriot G, Jouan Y, Joffre J, Huel F, De Prost N, Silva S, Azabou E, Radermacher P. Energetic dysfunction in sepsis: a narrative review. Ann Intensive Care 2021; 11:104. [PMID: 34216304 PMCID: PMC8254847 DOI: 10.1186/s13613-021-00893-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Background Growing evidence associates organ dysfunction(s) with impaired metabolism in sepsis. Recent research has increased our understanding of the role of substrate utilization and mitochondrial dysfunction in the pathophysiology of sepsis-related organ dysfunction. The purpose of this review is to present this evidence as a coherent whole and to highlight future research directions. Main text Sepsis is characterized by systemic and organ-specific changes in metabolism. Alterations of oxygen consumption, increased levels of circulating substrates, impaired glucose and lipid oxidation, and mitochondrial dysfunction are all associated with organ dysfunction and poor outcomes in both animal models and patients. The pathophysiological relevance of bioenergetics and metabolism in the specific examples of sepsis-related immunodeficiency, cerebral dysfunction, cardiomyopathy, acute kidney injury and diaphragmatic failure is also described. Conclusions Recent understandings in substrate utilization and mitochondrial dysfunction may pave the way for new diagnostic and therapeutic approaches. These findings could help physicians to identify distinct subgroups of sepsis and to develop personalized treatment strategies. Implications for their use as bioenergetic targets to identify metabolism- and mitochondria-targeted treatments need to be evaluated in future studies. Supplementary Information The online version contains supplementary material available at 10.1186/s13613-021-00893-7.
Collapse
Affiliation(s)
- Sebastien Preau
- U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France.
| | - Dominique Vodovar
- Centre AntiPoison de Paris, Hôpital Fernand Widal, APHP, 75010, Paris, France.,Faculté de pharmacie, UMRS 1144, 75006, Paris, France.,Université de Paris, UFR de Médecine, 75010, Paris, France
| | - Boris Jung
- Medical Intensive Care Unit, Lapeyronie Teaching Hospital, Montpellier University Hospital and PhyMedExp, University of Montpellier, Montpellier, France
| | - Steve Lancel
- U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Lara Zafrani
- Médecine Intensive Réanimation, Hôpital Saint-Louis, AP-HP, Université de Paris, Paris, France.,INSERM UMR 976, Hôpital Saint Louis, Université de Paris, Paris, France
| | | | - Mehdi Oualha
- Pediatric Intensive Care Unit, Necker Hospital, APHP, Centre - Paris University, Paris, France
| | - Guillaume Voiriot
- Service de Médecine Intensive Réanimation, Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Youenn Jouan
- Service de Médecine Intensive Réanimation, CHRU Tours, Tours, France.,Faculté de Médecine de Tours, INSERM U1100 Centre d'Etudes des Pathologies Respiratoires, Tours, France
| | - Jeremie Joffre
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, 94143, USA
| | - Fabrice Huel
- Réanimation médico-chirurgicale, Université de Paris, Assistance Publique - Hôpitaux de Paris, Hôpital Louis Mourier, Paris, France
| | - Nicolas De Prost
- Service de Réanimation Médicale, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Cedex 94010, Créteil, France
| | - Stein Silva
- Réanimation URM CHU Purpan, Cedex 31300, Toulouse, France.,Toulouse NeuroImaging Center INSERM1214, Cedex 31300, Toulouse, France
| | - Eric Azabou
- Clinical Neurophysiology and Neuromodulation Unit, Departments of Physiology and Critical Care Medicine, Raymond Poincaré Hospital, AP-HP, Inserm UMR 1173, Infection and Inflammation (2I), University of Versailles (UVSQ), Paris-Saclay University, Paris, France
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum, Ulm, Germany
| |
Collapse
|
26
|
Flower L, Page A, Puthucheary Z. Should nutritional therapy be modified to account for mitochondrial dysfunction in critical illness? JPEN J Parenter Enteral Nutr 2021; 45:60-65. [PMID: 34115880 DOI: 10.1002/jpen.2190] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/21/2021] [Accepted: 05/10/2021] [Indexed: 11/07/2022]
Abstract
Metabolic dysfunction, and its associated muscle atrophy, remains the most common complication of critical care. At the centre of this is mitochondrial dysfunction, secondary to hypoxia and systemic inflammation. This leads to a bioenergetic crisis, with decreased intramuscular adenosine tri-phosphate content and a reduction in the highly energy dependent process of protein synthesis. Numerous methods have been studied to try and reduce these effects, with only limited success. Trials investigating the use of increased calorie and protein administration have instead found a decrease in relative lean body mass, and a potential increase in morbidity and mortality. Ketone bodies have been proposed as alternative substrates for metabolism in critical illness, with promising results seen in animal models. They are currently being investigated in critical care patients in the Alternative Substrates in the Critically Ill Subjects trial. The evidence to date suggests that individualised feeding regimens may be key in the nutritional approach to critical illness. Consideration of individual patient factors will need to be combined with personalised protein content, total energy load received, and the timings of such feeds. This review covers mitochondrial dysfunction in critical illness, and how it contributes to muscle wasting and the resultant morbidity and mortality and the scientific basis of why current nutritional approaches to date have not been successful in negating this effect. These two factors underpin the need for consideration of alternative nutritional strategies in the critically ill patient. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Luke Flower
- William Harvey Research Institute, Queen Mary University of London, London.,Department of Anaesthesia, University College Hospital, 235 Euston Road, London, UK
| | - Alexandria Page
- William Harvey Research Institute, Queen Mary University of London, London.,Adult Critical Care Unit, The Royal London Hospital, Barts Health NHS Trust, Whitechapel Road, London, UK
| | - Zudin Puthucheary
- William Harvey Research Institute, Queen Mary University of London, London.,Adult Critical Care Unit, The Royal London Hospital, Barts Health NHS Trust, Whitechapel Road, London, UK
| |
Collapse
|
27
|
Mörgeli R, Wollersheim T, Engelhardt LJ, Grunow JJ, Lachmann G, Carbon NM, Koch S, Spies C, Weber-Carstens S. Critical illness myopathy precedes hyperglycaemia and high glucose variability. J Crit Care 2021; 63:32-39. [PMID: 33592497 DOI: 10.1016/j.jcrc.2021.01.012] [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: 10/08/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Critical Illness Myopathy (CIM) is a serious ICU complication, and dysglycaemia is widely regarded as a risk factor. Although glucose variability (GV) has been independently linked to ICU mortality, an association with CIM has not been investigated. This study examines the relationship between CIM and GV. METHODS Retrospective investigation including ICU patients with SOFA ≥8, mechanical ventilation, and CIM diagnostics. Glucose readings were collected every 6 h throughout the first week of treatment, when CIM is thought to develop. GV was measured using standard deviation (SD), coefficient of variability (CV), mean absolute glucose (MAG), mean amplitude of glycaemic excursions (MAGE), and mean of daily difference (MODD). RESULTS 74 patients were included, and 50 (67.6%) developed CIM. Time on glycaemic target (70-179 mg/dL), caloric and insulin intakes, mean, maximum and minimum blood glucose values were similar for all patients until the 5th day, after which CIM patients exhibited higher mean and maximum glucose levels. Significantly higher GV in CIM patients were observed on day 5 (SD, CV, MAG, MAGE), day 6 (MODD), and day 7 (SD, CV, MAG). CONCLUSIONS CIM patients developed transient increases in GV and hyperglycaemia only late in the first week, suggesting that myopathy precedes dysglycaemia.
Collapse
Affiliation(s)
- Rudolf Mörgeli
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | - Tobias Wollersheim
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, D-10178 Berlin, Germany.
| | - Lilian Jo Engelhardt
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | - Julius J Grunow
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, D-10178 Berlin, Germany.
| | - Gunnar Lachmann
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, D-10178 Berlin, Germany.
| | - Niklas M Carbon
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | - Susanne Koch
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | - Claudia Spies
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany.
| | - Steffen Weber-Carstens
- Department of Anaesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, D-13353 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, D-10178 Berlin, Germany.
| |
Collapse
|
28
|
Schönhofer B, Geiseler J, Dellweg D, Fuchs H, Moerer O, Weber-Carstens S, Westhoff M, Windisch W. Prolonged Weaning: S2k Guideline Published by the German Respiratory Society. Respiration 2020; 99:1-102. [PMID: 33302267 DOI: 10.1159/000510085] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 01/28/2023] Open
Abstract
Mechanical ventilation (MV) is an essential part of modern intensive care medicine. MV is performed in patients with severe respiratory failure caused by respiratory muscle insufficiency and/or lung parenchymal disease; that is, when other treatments such as medication, oxygen administration, secretion management, continuous positive airway pressure (CPAP), or nasal high-flow therapy have failed. MV is required for maintaining gas exchange and allows more time to curatively treat the underlying cause of respiratory failure. In the majority of ventilated patients, liberation or "weaning" from MV is routine, without the occurrence of any major problems. However, approximately 20% of patients require ongoing MV, despite amelioration of the conditions that precipitated the need for it in the first place. Approximately 40-50% of the time spent on MV is required to liberate the patient from the ventilator, a process called "weaning". In addition to acute respiratory failure, numerous factors can influence the duration and success rate of the weaning process; these include age, comorbidities, and conditions and complications acquired during the ICU stay. According to international consensus, "prolonged weaning" is defined as the weaning process in patients who have failed at least 3 weaning attempts, or require more than 7 days of weaning after the first spontaneous breathing trial (SBT). Given that prolonged weaning is a complex process, an interdisciplinary approach is essential for it to be successful. In specialised weaning centres, approximately 50% of patients with initial weaning failure can be liberated from MV after prolonged weaning. However, the heterogeneity of patients undergoing prolonged weaning precludes the direct comparison of individual centres. Patients with persistent weaning failure either die during the weaning process, or are discharged back to their home or to a long-term care facility with ongoing MV. Urged by the growing importance of prolonged weaning, this Sk2 Guideline was first published in 2014 as an initiative of the German Respiratory Society (DGP), in conjunction with other scientific societies involved in prolonged weaning. The emergence of new research, clinical study findings and registry data, as well as the accumulation of experience in daily practice, have made the revision of this guideline necessary. The following topics are dealt with in the present guideline: Definitions, epidemiology, weaning categories, underlying pathophysiology, prevention of prolonged weaning, treatment strategies in prolonged weaning, the weaning unit, discharge from hospital on MV, and recommendations for end-of-life decisions. Special emphasis was placed on the following themes: (1) A new classification of patient sub-groups in prolonged weaning. (2) Important aspects of pulmonary rehabilitation and neurorehabilitation in prolonged weaning. (3) Infrastructure and process organisation in the care of patients in prolonged weaning based on a continuous treatment concept. (4) Changes in therapeutic goals and communication with relatives. Aspects of paediatric weaning are addressed separately within individual chapters. The main aim of the revised guideline was to summarize both current evidence and expert-based knowledge on the topic of "prolonged weaning", and to use this information as a foundation for formulating recommendations related to "prolonged weaning", not only in acute medicine but also in the field of chronic intensive care medicine. The following professionals served as important addressees for this guideline: intensivists, pulmonary medicine specialists, anaesthesiologists, internists, cardiologists, surgeons, neurologists, paediatricians, geriatricians, palliative care clinicians, rehabilitation physicians, intensive/chronic care nurses, physiotherapists, respiratory therapists, speech therapists, medical service of health insurance, and associated ventilator manufacturers.
Collapse
Affiliation(s)
- Bernd Schönhofer
- Klinikum Agnes Karll Krankenhaus, Klinikum Region Hannover, Laatzen, Germany,
| | - Jens Geiseler
- Klinikum Vest, Medizinische Klinik IV: Pneumologie, Beatmungs- und Schlafmedizin, Marl, Germany
| | - Dominic Dellweg
- Fachkrankenhaus Kloster Grafschaft GmbH, Abteilung Pneumologie II, Schmallenberg, Germany
| | - Hans Fuchs
- Universitätsklinikum Freiburg, Zentrum für Kinder- und Jugendmedizin, Neonatologie und Pädiatrische Intensivmedizin, Freiburg, Germany
| | - Onnen Moerer
- Universitätsmedizin Göttingen, Klinik für Anästhesiologie, Göttingen, Germany
| | - Steffen Weber-Carstens
- Charité, Universitätsmedizin Berlin, Klinik für Anästhesiologie mit Schwerpunkt operative Intensivmedizin, Campus Virchow-Klinikum und Campus Mitte, Berlin, Germany
| | - Michael Westhoff
- Lungenklinik Hemer, Hemer, Germany
- Universität Witten/Herdecke, Herdecke, Germany
| | - Wolfram Windisch
- Lungenklinik, Kliniken der Stadt Köln gGmbH, Universität Witten/Herdecke, Herdecke, Germany
| |
Collapse
|
29
|
Schefold JC, Wollersheim T, Grunow JJ, Luedi MM, Z'Graggen WJ, Weber-Carstens S. Muscular weakness and muscle wasting in the critically ill. J Cachexia Sarcopenia Muscle 2020; 11:1399-1412. [PMID: 32893974 PMCID: PMC7749542 DOI: 10.1002/jcsm.12620] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/10/2020] [Accepted: 08/23/2020] [Indexed: 12/17/2022] Open
Affiliation(s)
- Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Tobias Wollersheim
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Julius J Grunow
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Markus M Luedi
- Department of Anaesthesiology and Pain Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Werner J Z'Graggen
- Department of Neurology and Neurosurgery, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Steffen Weber-Carstens
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
30
|
Segers J, Vanhorebeek I, Langer D, Charususin N, Wei W, Frickx B, Demeyere I, Clerckx B, Casaer M, Derese I, Derde S, Pauwels L, Van den Berghe G, Hermans G, Gosselink R. Early neuromuscular electrical stimulation reduces the loss of muscle mass in critically ill patients - A within subject randomized controlled trial. J Crit Care 2020; 62:65-71. [PMID: 33285371 DOI: 10.1016/j.jcrc.2020.11.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 01/21/2023]
Abstract
PURPOSE To investigate the effect of Neuromuscular Electrical Stimulation (NMES) on muscle thickness, strength and morphological and molecular markers of the quadriceps. MATERIALS AND METHODS Adult critically ill patients with an expected prolonged stay received unilateral quadriceps NMES sessions for 7 consecutive days. Before and after the intervention period, quadriceps thickness was measured with ultrasound. After the intervention period, strength was assessed in cooperative patients and muscle biopsies were taken. Multivariable regression was performed to identify factors affecting muscle thickness loss. RESULTS Muscle thickness decreased less in the stimulated leg (-6 ± 16% versus -12 ± 15%, p = 0.014, n = 47). Strength was comparable. Opioid administration, minimal muscle contraction and more muscle thickness loss in the non-stimulated muscle were independently associated with better muscle thickness preservation. Stimulated muscles showed a shift towards larger myofibers and higher MyHC-I gene expression. NMES did not affect gene expression of other myofibrillary proteins, MuRF-1 or atrogin-1. Signs of myofiber necrosis and inflammation were comparable for both muscles. CONCLUSIONS NMES attenuated the loss of muscle mass, but not of strength, in critically ill patients. Preservation of muscle mass was more likely in patients receiving opioids, patients with a minimal muscle contraction during NMES and patients more prone to lose muscle mass. TRIAL REGISTRATION clinicaltrials.govNCT02133300.
Collapse
Affiliation(s)
- Johan Segers
- Department of Rehabilitation Sciences, Faculty of Kinesiology and Rehabilitation Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium; Department of Intensive Care, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Ilse Vanhorebeek
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Daniel Langer
- Department of Rehabilitation Sciences, Faculty of Kinesiology and Rehabilitation Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Noppawan Charususin
- Department of Rehabilitation Sciences, Faculty of Kinesiology and Rehabilitation Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium; Department of Physical therapy, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, Thailand
| | - Weili Wei
- Department of Rehabilitation Sciences, Faculty of Kinesiology and Rehabilitation Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium; Department of Respiratory and Critical Care, Tongji Hospital, Shangai, China
| | - Bregje Frickx
- Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Inge Demeyere
- Department of Intensive Care, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Beatrix Clerckx
- Department of Rehabilitation Sciences, Faculty of Kinesiology and Rehabilitation Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium; Department of Intensive Care, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Michael Casaer
- Department of Intensive Care, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Inge Derese
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Sarah Derde
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Lies Pauwels
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Greet Van den Berghe
- Department of Intensive Care, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Greet Hermans
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium; Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Rik Gosselink
- Department of Rehabilitation Sciences, Faculty of Kinesiology and Rehabilitation Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium; Department of Intensive Care, University Hospitals Leuven, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
31
|
Intensive Care Unit-Acquired Weakness: Not just Another Muscle Atrophying Condition. Int J Mol Sci 2020; 21:ijms21217840. [PMID: 33105809 PMCID: PMC7660068 DOI: 10.3390/ijms21217840] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Intensive care unit-acquired weakness (ICUAW) occurs in critically ill patients stemming from the critical illness itself, and results in sustained disability long after the ICU stay. Weakness can be attributed to muscle wasting, impaired contractility, neuropathy, and major pathways associated with muscle protein degradation such as the ubiquitin proteasome system and dysregulated autophagy. Furthermore, it is characterized by the preferential loss of myosin, a distinct feature of the condition. While many risk factors for ICUAW have been identified, effective interventions to offset these changes remain elusive. In addition, our understanding of the mechanisms underlying the long-term, sustained weakness observed in a subset of patients after discharge is minimal. Herein, we discuss the various proposed pathways involved in the pathophysiology of ICUAW, with a focus on the mechanisms underpinning skeletal muscle wasting and impaired contractility, and the animal models used to study them. Furthermore, we will explore the contributions of inflammation, steroid use, and paralysis to the development of ICUAW and how it pertains to those with the corona virus disease of 2019 (COVID-19). We then elaborate on interventions tested as a means to offset these decrements in muscle function that occur as a result of critical illness, and we propose new strategies to explore the molecular mechanisms of ICUAW, including serum-related biomarkers and 3D human skeletal muscle culture models.
Collapse
|
32
|
Lachmann G, Mörgeli R, Kuenz S, Piper SK, Spies C, Kurpanik M, Weber-Carstens S, Wollersheim T. Perioperatively Acquired Weakness. Anesth Analg 2020; 130:341-351. [PMID: 30855340 DOI: 10.1213/ane.0000000000004068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Skeletal muscle failure in critical illness (intensive care unit-acquired weakness) is a well-known complication developing early during intensive care unit stay. However, muscle weakness during the perioperative setting has not yet been investigated. METHODS We performed a subgroup investigation of a prospective observational trial to investigate perioperative muscle weakness. Eighty-nine patients aged 65 years or older were assessed for handgrip strength preoperatively, on the first postoperative day, at intensive care unit discharge, at hospital discharge, and at 3-month follow-up. Functional status was evaluated perioperatively via Barthel index, instrumental activities of daily living, Timed Up and Go test, and functional independence measure. After exclusion of patients with intensive care unit-acquired weakness or intensive care unit stay of ≥72 hours, 59 patients were included into our analyses. Of these, 14 patients had additional pulmonary function tests preoperatively and on postoperative day 1. Blood glucose was measured intraoperatively every 20 minutes. RESULTS Handgrip strength significantly decreased after surgery on postoperative day 1 by 16.4% (P < .001). Postoperative pulmonary function significantly decreased by 13.1% for vital capacity (P = .022) and 12.6% for forced expiratory volume in 1 second (P = .001) on postoperative day 1. Handgrip strength remained significantly reduced at hospital discharge (P = .016) and at the 3-month follow-up (P = .012). Perioperative glucose levels showed no statistically significant impact on muscle weakness. Instrumental activities of daily living (P < .001) and functional independence measure (P < .001) were decreased at hospital discharge, while instrumental activities of daily living remained decreased at the 3-month follow-up (P = .026) compared to preoperative assessments. CONCLUSIONS Perioperatively acquired weakness occurred, indicated by a postoperatively decreased handgrip strength, decreased respiratory muscle function, and impaired functional status, which partly remained up to 3 months.
Collapse
Affiliation(s)
- Gunnar Lachmann
- From the Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Rudolf Mörgeli
- From the Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sophia Kuenz
- From the Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie K Piper
- Berlin Institute of Health (BIH), Berlin, Germany.,Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Spies
- From the Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maryam Kurpanik
- From the Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Steffen Weber-Carstens
- From the Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Tobias Wollersheim
- From the Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | | |
Collapse
|
33
|
Flower L, Puthucheary Z. Muscle wasting in the critically ill patient: how to minimise subsequent disability. Br J Hosp Med (Lond) 2020; 81:1-9. [PMID: 32339009 DOI: 10.12968/hmed.2020.0045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Muscle wasting in critically ill patients is the most common complication associated with critical care. It has significant effects on physical and psychological health, mortality and quality of life. It is most severe in the first few days of illness and in the most critically unwell patients, with muscle loss estimated to occur at 2-3% per day. This muscle loss is likely a result of a reduction in protein synthesis relative to muscle breakdown, resulting in altered protein homeostasis. The associated weakness is associated with in an increase in both short- and long-term mortality and morbidity, with these detrimental effects demonstrated up to 5 years post discharge. This article highlights the significant impact that muscle wasting has on critically ill patients' outcomes, how this can be reduced, and how this might change in the future.
Collapse
Affiliation(s)
- Luke Flower
- Department of Anaesthetics, University College Hospital, London, UK
| | - Zudin Puthucheary
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK.,Adult Critical Care Unit, Royal London Hospital, London, UK
| |
Collapse
|
34
|
Chao WC, Tseng CH, Wu CL, Shih SJ, Yi CY, Chan MC. Higher glycemic variability within the first day of ICU admission is associated with increased 30-day mortality in ICU patients with sepsis. Ann Intensive Care 2020; 10:17. [PMID: 32034567 PMCID: PMC7007493 DOI: 10.1186/s13613-020-0635-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 01/30/2020] [Indexed: 12/29/2022] Open
Abstract
Background High glycemic variability (GV) is common in critically ill patients; however, the prevalence and mortality association with early GV in patients with sepsis remains unclear. Methods This retrospective cohort study was conducted in a medical intensive care unit (ICU) in central Taiwan. Patients in the ICU with sepsis between January 2014 and December 2015 were included for analysis. All of these patients received protocol-based management, including blood sugar monitoring every 2 h for the first 24 h of ICU admission. Mean amplitude of glycemic excursions (MAGE) and coefficient of variation (CoV) were used to assess GV. Results A total of 452 patients (mean age 71.4 ± 14.7 years; 76.7% men) were enrolled for analysis. They were divided into high GV (43.4%, 196/452) and low GV (56.6%, 256/512) groups using MAGE 65 mg/dL as the cut-off point. Patients with high GV tended to have higher HbA1c (6.7 ± 1.8% vs. 5.9 ± 0.9%, p < 0.01) and were more likely to have diabetes mellitus (DM) (50.0% vs. 23.4%, p < 0.01) compared with those in the low GV group. Kaplan–Meier analysis showed that a high GV was associated with increased 30-day mortality (log-rank test, p = 0.018). The association remained strong in the non-DM (log-rank test, p = 0.035), but not in the DM (log-rank test, p = 0.254) group. Multivariate Cox proportional hazard regression analysis identified that high APACHE II score (adjusted hazard ratio (aHR) 1.045, 95% confidence interval (CI) 1.013–1.078), high serum lactate level at 0 h (aHR 1.009, 95% CI 1.003–1.014), having chronic airway disease (aHR 0.478, 95% CI 0.302–0.756), high mean day 1 glucose (aHR 1.008, 95% CI 1.000–1.016), and high MAGE (aHR 1.607, 95% CI 1.008–2.563) were independently associated with increased 30-day mortality. The association with 30-day mortality remained consistent when using CoV to assess GV. Conclusions We found that approximately 40% of the septic patients had a high early GV, defined as MAGE > 65 mg/dL. Higher GV within 24 h of ICU admission was independently associated with increased 30-day mortality. These findings highlight the need to monitor GV in septic patients early during an ICU admission.
Collapse
Affiliation(s)
- Wen-Cheng Chao
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan.,Department of Critical Care Medicine, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan
| | - Chien-Hua Tseng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chieh-Liang Wu
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan.,Center of Quality Management, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan.,Department of Automatic Control Engineering, Feng Chia University, Taichung, Taiwan
| | - Sou-Jen Shih
- Department of Nursing, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan
| | - Chi-Yuan Yi
- Department of Nursing, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan
| | - Ming-Cheng Chan
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan. .,Division of Critical Care and Respiratory Therapy, Department of Internal Medicine, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sect. 4, Taichung, 40705, Taiwan. .,Central Taiwan University of Science and Technology, Taichung, Taiwan. .,The College of Science, Tunghai University, Taichung, 40704, Taiwan.
| |
Collapse
|
35
|
Nerve damage induced skeletal muscle atrophy is associated with increased accumulation of intramuscular glucose and polyol pathway intermediates. Sci Rep 2020; 10:1908. [PMID: 32024865 PMCID: PMC7002415 DOI: 10.1038/s41598-020-58213-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/23/2019] [Indexed: 01/21/2023] Open
Abstract
Perturbations in skeletal muscle metabolism have been reported for a variety of neuromuscular diseases. However, the role of metabolism after constriction injury to a nerve and the associated muscle atrophy is unclear. We have analyzed rat tibialis anterior (TA) four weeks after unilateral constriction injury to the sciatic nerve (DMG) and in the contralateral control leg (CTRL) (n = 7) to investigate changes of the metabolome, immunohistochemistry and protein levels. Untargeted metabolomics identified 79 polar metabolites, 27 of which were significantly altered in DMG compared to CTRL. Glucose concentrations were increased 2.6-fold in DMG, while glucose 6-phosphate (G6-P) was unchanged. Intermediates of the polyol pathway were increased in DMG, particularly fructose (1.7-fold). GLUT4 localization was scattered as opposed to clearly at the sarcolemma. Despite the altered localization, we found GLUT4 protein levels to be increased 7.8-fold while GLUT1 was decreased 1.7-fold in nerve damaged TA. PFK1 and GS levels were both decreased 2.1-fold, indicating an inability of glycolysis and glycogen synthesis to process glucose at sufficient rates. In conclusion, chronic nerve constriction causes increased GLUT4 levels in conjunction with decreased glycolytic activity and glycogen storage in skeletal muscle, resulting in accumulation of intramuscular glucose and polyol pathway intermediates.
Collapse
|
36
|
Waldauf P, Gojda J, Urban T, Hrušková N, Blahutová B, Hejnová M, Jiroutková K, Fric M, Jánský P, Kukulová J, Stephens F, Řasová K, Duška F. Functional electrical stimulation-assisted cycle ergometry in the critically ill: protocol for a randomized controlled trial. Trials 2019; 20:724. [PMID: 31842936 PMCID: PMC6915865 DOI: 10.1186/s13063-019-3745-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/23/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Intensive care unit (ICU)-acquired weakness is the most important cause of failed functional outcome in survivors of critical care. Most damage occurs during the first week when patients are not cooperative enough with conventional rehabilitation. Functional electrical stimulation-assisted cycle ergometry (FES-CE) applied within 48 h of ICU admission may improve muscle function and long-term outcome. METHODS An assessor-blinded, pragmatic, single-centre randomized controlled trial will be performed. Adults (n = 150) mechanically ventilated for < 48 h from four ICUs who are estimated to need > 7 days of critical care will be randomized (1:1) to receive either standard of care or FES-CE-based intensified rehabilitation, which will continue until ICU discharge. PRIMARY OUTCOME quality of life measured by 36-Item Short Form Health Survey score at 6 months. SECONDARY OUTCOMES functional performance at ICU discharge, muscle mass (vastus ultrasound, N-balance) and function (Medical Research Council score, insulin sensitivity). In a subgroup (n = 30) we will assess insulin sensitivity and perform skeletal muscle biopsies to look at mitochondrial function, fibre typing and regulatory protein expression. TRIAL REGISTRATION ClinicalTrials.gov, NCT02864745. Registered on 12 August 2016.
Collapse
Affiliation(s)
- Petr Waldauf
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic
| | - Jan Gojda
- Department of Internal Medicine II, Charles University, 3rd Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Tomáš Urban
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic
| | - Natália Hrušková
- Department of Rehabilitation, Charles University, 3rd Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Barbora Blahutová
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic
- Department of Rehabilitation, Charles University, 3rd Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Marie Hejnová
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic
- Department of Rehabilitation, Charles University, 3rd Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - Kateřina Jiroutková
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic
| | - Michal Fric
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic
| | - Pavel Jánský
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic
| | - Jana Kukulová
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic
| | - Francis Stephens
- College of Life and Environmental Sciences, Sport and Health Sciences, University of Exeter, Exeter, UK
| | - Kamila Řasová
- Department of Rehabilitation, Charles University, 3rd Faculty of Medicine and FNKV University Hospital, Prague, Czech Republic
| | - František Duška
- Department of Anaesthesiology and Intensive Care Medicine, Charles University, 3rd Faculty of Medicine and KAR FNKV University Hospital, Fac Med 3, Srobarova 50, 10034, Prague, Czech Republic.
| |
Collapse
|
37
|
Neuromuscular electrical stimulation in critically ill traumatic brain injury patients attenuates muscle atrophy, neurophysiological disorders, and weakness: a randomized controlled trial. J Intensive Care 2019; 7:59. [PMID: 31890221 PMCID: PMC6909464 DOI: 10.1186/s40560-019-0417-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022] Open
Abstract
Background Critically ill traumatic brain injury (TBI) patients experience extensive muscle damage during their stay in the intensive care unit. Neuromuscular electrical stimulation (NMES) has been considered a promising treatment to reduce the functional and clinical impacts of this. However, the time needed for NMES to produce effects over the muscles is still unclear. This study primarily aimed to assess the time needed and effects of an NMES protocol on muscle architecture, neuromuscular electrophysiological disorder (NED), and muscle strength, and secondarily, to evaluate the effects on plasma systemic inflammation, catabolic responses, and clinical outcomes. Methods We performed a randomized clinical trial in critically ill TBI patients. The control group received only conventional physiotherapy, while the NMES group additionally underwent daily NMES for 14 days in the lower limb muscles. Participants were assessed at baseline and on days 3, 7, and 14 of their stay in the intensive care unit. The primary outcomes were assessed with muscle ultrasound, neuromuscular electrophysiology, and evoked peak force, and the secondary outcomes with plasma cytokines, matrix metalloproteinases, and clinical outcomes. Results Sixty participants were randomized, and twenty completed the trial from each group. After 14 days, the control group presented a significant reduction in muscle thickness of tibialis anterior and rectus femoris, mean of - 0.33 mm (- 14%) and - 0.49 mm (- 21%), p < 0.0001, respectively, while muscle thickness was preserved in the NMES group. The control group presented a higher incidence of NED: 47% vs. 0% in the NMES group, p < 0.0001, risk ratio of 16, and the NMES group demonstrated an increase in the evoked peak force (2.34 kg/f, p < 0.0001), in contrast to the control group (- 1.55 kg/f, p < 0.0001). The time needed for the NMES protocol to prevent muscle architecture disorders and treat weakness was at least 7 days, and 14 days to treat NED. The secondary outcomes exhibited less precise results, with confidence intervals that spanned worthwhile or trivial effects. Conclusions NMES applied daily for fourteen consecutive days reduced muscle atrophy, the incidence of NED, and muscle weakness in critically ill TBI patients. At least 7 days of NMES were required to elicit the first significant results. Trial registration The trial was registered at ensaiosclinicos.gov.br under protocol RBR-8kdrbz on 17 January 2016.
Collapse
|
38
|
García-Martínez MÁ, Montejo González JC, García-de-Lorenzo Y Mateos A, Teijeira S. Muscle weakness: Understanding the principles of myopathy and neuropathy in the critically ill patient and the management options. Clin Nutr 2019; 39:1331-1344. [PMID: 31255348 DOI: 10.1016/j.clnu.2019.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/12/2019] [Accepted: 05/31/2019] [Indexed: 12/11/2022]
Abstract
Myo-neuropathy of the critically ill patient is a difficult nosological entity to understand and manage. It appears soon after injury, and it is estimated that 20-30% of patients admitted to Intensive Care Units will develop it in some degree. Although muscular and nervous involvement are related, the former has a better prognosis. Myo-neuropathy associates to more morbidity, longer stay in Intensive Care Unit and in hospital, and also to higher costs and mortality. It is considered part of the main determinants of the new entities: the Chronic Critical Patient and the Post Intensive Care Syndrome. This update focuses on aetiology, pathophysiology, diagnosis and strategies that can prevent, alleviate and/or improve muscle (or muscle-nerve) weakness.
Collapse
Affiliation(s)
- Miguel Ángel García-Martínez
- Department of Intensive Care Medicine, Hospital Universitario de Torrevieja, Ctra. Torrevieja a San Miguel de Salinas s/n, 03186, Torrevieja, Alicante, Spain.
| | - Juan Carlos Montejo González
- Department of Intensive Care Medicine, Hospital Universitario, 12 de Octubre, Av. Cordoba, s/n, 28041, Madrid, Spain
| | | | - Susana Teijeira
- Rare Diseases & Pediatric Medicine Research Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Complejo Hospitalario Universitario de Vigo, Calle de Clara Campoamor, 341, 36312, Vigo, Pontevedra, Spain
| |
Collapse
|
39
|
Mignemi NA, McClatchey PM, Kilchrist KV, Williams IM, Millis BA, Syring KE, Duvall CL, Wasserman DH, McGuinness OP. Rapid changes in the microvascular circulation of skeletal muscle impair insulin delivery during sepsis. Am J Physiol Endocrinol Metab 2019; 316:E1012-E1023. [PMID: 30860883 PMCID: PMC6620574 DOI: 10.1152/ajpendo.00501.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/21/2019] [Accepted: 03/07/2019] [Indexed: 01/07/2023]
Abstract
Sepsis costs the healthcare system $23 billion annually and has a mortality rate between 10 and 40%. An early indication of sepsis is the onset of hyperglycemia, which is the result of sepsis-induced insulin resistance in skeletal muscle. Previous investigations have focused on events in the myocyte (e.g., insulin signaling and glucose transport and subsequent metabolism) as the causes for this insulin-resistant state. However, the delivery of insulin to the skeletal muscle is also an important determinant of insulin action. Skeletal muscle microvascular blood flow, which delivers the insulin to the muscle, is known to be decreased during sepsis. Here we test whether the reduced capillary blood flow to skeletal muscle belies the sepsis-induced insulin resistance by reducing insulin delivery to the myocyte. We hypothesize that decreased capillary flow and consequent decrease in insulin delivery is an early event that precedes gross cardiovascular alterations seen with sepsis. This hypothesis was examined in mice treated with either lipopolysaccharide (LPS) or polymicrobial sepsis followed by intravital microscopy of the skeletal muscle microcirculation. We calculated insulin delivery to the myocyte using two independent methods and found that LPS and sepsis rapidly reduce insulin delivery to the skeletal muscle by ~50%; this was driven by decreases in capillary flow velocity and the number of perfused capillaries. Furthermore, the changes in skeletal muscle microcirculation occur before changes in both cardiac output and arterial blood pressure. These data suggest that a rapid reduction in skeletal muscle insulin delivery contributes to the induction of insulin resistance during sepsis.
Collapse
Affiliation(s)
- Nicholas A Mignemi
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - P Mason McClatchey
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Kameron V Kilchrist
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - Ian M Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Bryan A Millis
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Biophotonics Center, Vanderbilt University , Nashville, Tennessee
| | - Kristen E Syring
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Mouse Metabolic Phenotyping Center , Nashville, Tennessee
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Mouse Metabolic Phenotyping Center , Nashville, Tennessee
| |
Collapse
|
40
|
Lotri-Koffi A, Pauly M, Lemarié E, Godin-Ribuot D, Tamisier R, Pépin JL, Vivodtzev I. Chronic neuromuscular electrical stimulation improves muscle mass and insulin sensitivity in a mouse model. Sci Rep 2019; 9:7252. [PMID: 31076597 PMCID: PMC6510751 DOI: 10.1038/s41598-019-43696-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 04/10/2019] [Indexed: 12/03/2022] Open
Abstract
Muscle wasting reduces functional capacity and increases cardiometabolic risk in chronic disease. Neuromuscular electrical stimulation (NMES) of the lower limb has been shown to reverse muscle wasting in these patients but its effect on cardiometabolic health is unclear. We investigated a mouse model of in-vivo non-invasive chronic NMES on muscle mass, insulin sensitivity and arterial blood pressure (BP). Twenty-three C57BL6 mice underwent unilateral NMES or sham training over 2.5 weeks while anesthetized by isoflurane. Lower limb muscle mass and the stimulated limb to non-stimulated limb muscle mass ratio were compared between groups (NMES vs. sham). Insulin sensitivity was assessed 48 h after training using an intraperitoneal insulin tolerance test (ITT) and BP was assessed before and after training using the tail-cuff technique. After training, muscle mass increased in NMES vs. sham (416 ± 6 vs. 397 ± 6 mg, p = 0.04) along with the ratio of muscle mass (+3 ± 1% vs. −1 ± 1% p = 0.04). Moreover, insulin sensitivity improved in NMES vs. sham (average blood glucose during ITT: 139.6 ± 8.5 vs. 161.9 ± 9.0 mg/dl blood, p = 0.01). BP was decreased in both groups, although it is likely that the effect of NMES on BP was dampened by repetitive anesthesia. The metabolic benefit of NMES training could be of great utility in patients with chronic disease. Moreover, the clinical-like mouse model of NMES is an effective tool to investigate the systemic effects of local muscle strengthening.
Collapse
Affiliation(s)
- Adiel Lotri-Koffi
- Univ. Grenoble Alpes, Inserm 1042, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Marion Pauly
- Univ. Grenoble Alpes, Inserm 1042, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Emeline Lemarié
- Univ. Grenoble Alpes, Inserm 1042, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Diane Godin-Ribuot
- Univ. Grenoble Alpes, Inserm 1042, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Renaud Tamisier
- Univ. Grenoble Alpes, Inserm 1042, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Jean-Louis Pépin
- Univ. Grenoble Alpes, Inserm 1042, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France
| | - Isabelle Vivodtzev
- Univ. Grenoble Alpes, Inserm 1042, CHU Grenoble Alpes, HP2 Laboratory, Grenoble, France. .,Cardiovascular Research Laboratory, Spaulding Rehabilitation Hospital, Cambridge, Massachusetts, USA. .,Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
41
|
Zhao L, Pascual F, Bacudio L, Suchanek AL, Young PA, Li LO, Martin SA, Camporez JP, Perry RJ, Shulman GI, Klett EL, Coleman RA. Defective fatty acid oxidation in mice with muscle-specific acyl-CoA synthetase 1 deficiency increases amino acid use and impairs muscle function. J Biol Chem 2019; 294:8819-8833. [PMID: 30975900 DOI: 10.1074/jbc.ra118.006790] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/21/2019] [Indexed: 01/07/2023] Open
Abstract
Loss of long-chain acyl-CoA synthetase isoform-1 (ACSL1) in mouse skeletal muscle (Acsl1M -/-) severely reduces acyl-CoA synthetase activity and fatty acid oxidation. However, the effects of decreased fatty acid oxidation on skeletal muscle function, histology, use of alternative fuels, and mitochondrial function and morphology are unclear. We observed that Acsl1M -/- mice have impaired voluntary running capacity and muscle grip strength and that their gastrocnemius muscle contains myocytes with central nuclei, indicating muscle regeneration. We also found that plasma creatine kinase and aspartate aminotransferase levels in Acsl1M -/- mice are 3.4- and 1.5-fold greater, respectively, than in control mice (Acsl1flox/flox ), indicating muscle damage, even without exercise, in the Acsl1M -/- mice. Moreover, caspase-3 protein expression exclusively in Acsl1M -/- skeletal muscle and the presence of cleaved caspase-3 suggested myocyte apoptosis. Mitochondria in Acsl1M -/- skeletal muscle were swollen with abnormal cristae, and mitochondrial biogenesis was increased. Glucose uptake did not increase in Acsl1M -/- skeletal muscle, and pyruvate oxidation was similar in gastrocnemius homogenates from Acsl1M -/- and control mice. The rate of protein synthesis in Acsl1M -/- glycolytic muscle was 2.1-fold greater 30 min after exercise than in the controls, suggesting resynthesis of proteins catabolized for fuel during the exercise. At this time, mTOR complex 1 was activated, and autophagy was blocked. These results suggest that fatty acid oxidation is critical for normal skeletal muscle homeostasis during both rest and exercise. We conclude that ACSL1 deficiency produces an overall defect in muscle fuel metabolism that increases protein catabolism, resulting in exercise intolerance, muscle weakness, and myocyte apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei O Li
- From the Departments of Nutrition and
| | - Sarah A Martin
- the Department of Molecular Genetics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | | | - Rachel J Perry
- the Departments of Internal Medicine and.,Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Gerald I Shulman
- the Departments of Internal Medicine and.,Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Eric L Klett
- Medicine, University of North Carolina, Chapel Hill, North Carolina 27599
| | | |
Collapse
|
42
|
Tamion F, Bohé J. Comment je prends en charge la nutrition d’un patient en état de choc. MEDECINE INTENSIVE REANIMATION 2018. [DOI: 10.3166/rea-2018-0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
La réponse métabolique à l’agression correspond à un ensemble de réactions à la base de l’adaptation de l’organisme aux nouvelles conditions. Ces modifications concernent des aspects métaboliques spécifiques comme le maintien de la masse protéique et/ou l’état des réserves énergétiques. L’une des principales difficultés de l’optimisation du support métabolique consiste à distinguer les changements métaboliques bénéfiques de ceux qui sont délétères pour l’organisme. Dans ce contexte, les objectifs thérapeutiques peuvent se limiter à une approche nutritionnelle s’attachant à limiter le déficit énergétique et les pertes protéiques et musculaires. Ils peuvent être plus ambitieux en essayant d’adapter les apports aux différents besoins d’un point de vue quantitatif comme qualitatif. La limitation du déficit énergétique semble être un objectif raisonnable à atteindre selon les données de la littérature. Enfin, essayer d’interférer avec la réponse métabolique à l’agression (immunomodulation, manipulations pharmacologiques des voies métaboliques, etc.) représente le degré d’intervention métabolique le plus élaboré et, si quelques données ont pu être encourageantes, il n’est pas possible d’affirmer que cet objectif soit complètement réaliste, voire même bénéfique. Les apports nutritionnels doivent être intégrés à la stratégie thérapeutique globale de prise en charge. La réponse optimale du support nutritionnel a pour but « de donner les moyens métaboliques » de la guérison.
Collapse
|
43
|
Puthucheary ZA, Astin R, Mcphail MJW, Saeed S, Pasha Y, Bear DE, Constantin D, Velloso C, Manning S, Calvert L, Singer M, Batterham RL, Gomez-Romero M, Holmes E, Steiner MC, Atherton PJ, Greenhaff P, Edwards LM, Smith K, Harridge SD, Hart N, Montgomery HE. Metabolic phenotype of skeletal muscle in early critical illness. Thorax 2018; 73:926-935. [PMID: 29980655 DOI: 10.1136/thoraxjnl-2017-211073] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 05/17/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To characterise the sketetal muscle metabolic phenotype during early critical illness. METHODS Vastus lateralis muscle biopsies and serum samples (days 1 and 7) were obtained from 63 intensive care patients (59% male, 54.7±18.0 years, Acute Physiology and Chronic Health Evaluation II score 23.5±6.5). MEASUREMENTS AND MAIN RESULTS From day 1 to 7, there was a reduction in mitochondrial beta-oxidation enzyme concentrations, mitochondrial biogenesis markers (PGC1α messenger mRNA expression (-27.4CN (95% CI -123.9 to 14.3); n=23; p=0.025) and mitochondrial DNA copy number (-1859CN (IQR -5557-1325); n=35; p=0.032). Intramuscular ATP content was reduced compared tocompared with controls on day 1 (17.7mmol/kg /dry weight (dw) (95% CI 15.3 to 20.0) vs. 21.7 mmol/kg /dw (95% CI 20.4 to 22.9); p<0.001) and decreased over 7 days (-4.8 mmol/kg dw (IQR -8.0-1.2); n=33; p=0.001). In addition, the ratio of phosphorylated:total AMP-K (the bioenergetic sensor) increased (0.52 (IQR -0.09-2.6); n=31; p<0.001). There was an increase in intramuscular phosphocholine (847.2AU (IQR 232.5-1672); n=15; p=0.022), intramuscular tumour necrosis factor receptor 1 (0.66 µg (IQR -0.44-3.33); n=29; p=0.041) and IL-10 (13.6 ng (IQR 3.4-39.0); n=29; p=0.004). Serum adiponectin (10.3 µg (95% CI 6.8 to 13.7); p<0.001) and ghrelin (16.0 ng/mL (IQR -7-100); p=0.028) increased. Network analysis revealed a close and direct relationship between bioenergetic impairment and reduction in muscle mass and between intramuscular inflammation and impaired anabolic signaling. ATP content and muscle mass were unrelated to lipids delivered. CONCLUSIONS Decreased mitochondrial biogenesis and dysregulated lipid oxidation contribute to compromised skeletal muscle bioenergetic status. In addition, intramuscular inflammation was associated with impaired anabolic recovery with lipid delivery observed as bioenergetically inert. Future clinical work will focus on these key areas to ameliorate acute skeletal muscle wasting. TRIAL REGISTRATION NUMBER NCT01106300.
Collapse
Affiliation(s)
- Zudin A Puthucheary
- Institute for Sport, Exercise and Health, University College London, London, UK
- Department of Medicine, Centre for Human Health and Performance, University College London, London, UK
- Intensive Care Unit, Royal Free London NHS Foundation Trust, London, UK
- Centre for Human and Applied Physiological Sciences, King's College London, London, UK
| | - Ronan Astin
- Institute for Sport, Exercise and Health, University College London, London, UK
- Department of Medicine, Centre for Human Health and Performance, University College London, London, UK
| | - Mark J W Mcphail
- Hepatology and Gastroenterology, St Mary's Hospital, Imperial College London, London, UK
- Institute of Liver Studies, Kings College Hospital NHS Foundation Trust, London, UK
| | - Saima Saeed
- Wolfson Institute Centre for Intensive Care Medicine, University College London, London, UK
| | - Yasmin Pasha
- Hepatology and Gastroenterology, St Mary's Hospital, Imperial College London, London, UK
| | - Danielle E Bear
- Centre for Human and Applied Physiological Sciences, King's College London, London, UK
- Department of Nutrition and Dietetics, Guy's and St Thomas' NHS Foundation Trust, London
- Department of Critical Care, Guy's and St Thomas' NHS Foundation Trust, London
- Lane Fox Clinical Respiratory Physiology Research Centre, St Thomas' Hospital, Guy's and St Thomas' Foundation Trust, London, London, UK
| | - Despina Constantin
- Medical Research Council/Arthritis Research UK Centre for Musculoskeletal Aging Research, National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Cristiana Velloso
- Centre for Human and Applied Physiological Sciences, King's College London, London, UK
| | - Sean Manning
- Centre for Obesity Research, University College London, London, UK
- National Institute of Health Research, UCLH Biomedical Research Centre, University College London Hospitals, London
- School of Medicine, University College Cork, Cork, Ireland
| | - Lori Calvert
- Northwest Anglia foundation Trust, Peterborough City Hospital NHS Trust, Peterborough, UK
| | - Mervyn Singer
- Intensive Care Unit, Royal Free London NHS Foundation Trust, London, UK
- Wolfson Institute Centre for Intensive Care Medicine, University College London, London, UK
| | - Rachel L Batterham
- Centre for Obesity Research, University College London, London, UK
- National Institute of Health Research, UCLH Biomedical Research Centre, University College London Hospitals, London
| | - Maria Gomez-Romero
- Biomolecular Medicine, Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, London, UK
| | - Elaine Holmes
- Biomolecular Medicine, Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, London, UK
| | - Michael C Steiner
- Institute for Lung Health, Leicester NIHR Biomedical Research Centre-Respiratory, University of Leicester, Leicester, UK
| | - Philip J Atherton
- Medical Research Council/Arthritis Research UK Centre for Musculoskeletal Aging Research, National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Paul Greenhaff
- Medical Research Council/Arthritis Research UK Centre for Musculoskeletal Aging Research, National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Lindsay M Edwards
- Digital, Data & Analytics Unit, Respiratory Therapy Area, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Kenneth Smith
- Medical Research Council/Arthritis Research UK Centre for Musculoskeletal Aging Research, National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Stephen D Harridge
- Centre for Human and Applied Physiological Sciences, King's College London, London, UK
| | - Nicholas Hart
- Lane Fox Clinical Respiratory Physiology Research Centre, St Thomas' Hospital, Guy's and St Thomas' Foundation Trust, London, London, UK
- Lane Fox Respiratory Service, St Thomas' Hospital, Guy's and St Thomas' Foundation Trust, London, UK
| | - Hugh E Montgomery
- Institute for Sport, Exercise and Health, University College London, London, UK
- Department of Medicine, Centre for Human Health and Performance, University College London, London, UK
| |
Collapse
|
44
|
Diaphragm Weakness in the Critically Ill: Basic Mechanisms Reveal Therapeutic Opportunities. Chest 2018; 154:1395-1403. [PMID: 30144420 DOI: 10.1016/j.chest.2018.08.1028] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/23/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022] Open
Abstract
The diaphragm is the primary muscle of inspiration. Its capacity to respond to the load imposed by pulmonary disease is a major determining factor both in the onset of ventilatory failure and in the ability to successfully separate patients from ventilator support. It has recently been established that a very large proportion of critically ill patients exhibit major weakness of the diaphragm, which is associated with poor clinical outcomes. The two greatest risk factors for the development of diaphragm weakness in critical illness are the use of mechanical ventilation and the presence of sepsis. Loss of force production by the diaphragm under these conditions is caused by a combination of defective contractility and reduced diaphragm muscle mass. Importantly, many of the same molecular mechanisms are implicated in the diaphragm dysfunction associated with both mechanical ventilation and sepsis. This review outlines the primary cellular mechanisms identified thus far at the nexus of diaphragm dysfunction associated with mechanical ventilation and/or sepsis, and explores the potential for treatment or prevention of diaphragm weakness in critically ill patients through therapeutic manipulation of these final common pathway targets.
Collapse
|
45
|
Schaller SJ, Nagashima M, Schönfelder M, Sasakawa T, Schulz F, Khan MAS, Kem WR, Schneider G, Schlegel J, Lewald H, Blobner M, Jeevendra Martyn JA. GTS-21 attenuates loss of body mass, muscle mass, and function in rats having systemic inflammation with and without disuse atrophy. Pflugers Arch 2018; 470:1647-1657. [PMID: 30006848 DOI: 10.1007/s00424-018-2180-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/19/2018] [Accepted: 07/05/2018] [Indexed: 12/19/2022]
Abstract
Muscle changes of critical illness are attributed to systemic inflammatory responses and disuse atrophy. GTS-21 (3-(2,4-dimethoxy-benzylidene)anabaseine), also known as DMBX-A) is a synthetic derivative of the natural product anabaseine that acts as an agonist at α7-acetylcholine receptors (α7nAChRs). Hypothesis tested was that modulation of inflammation by agonist GTS-21 (10 mg/kg b.i.d. intraperitoneally) will attenuate body weight (BW) and muscle changes. Systemic sham inflammation was produced in 125 rats by Cornyebacterium parvum (C.p.) or saline injection on days 0/4/8. Seventy-four rats had one immobilized-limb producing disuse atrophy. GTS-21 effects on BW, tibialis muscle mass (TMM), and function were assessed on day 12. Systemically, methemoglobin levels increased 26-fold with C.p. (p < 0.001) and decreased significantly (p < 0.033) with GTS-21. Control BW increased (+ 30 ± 9 g, mean ± SD) at day 12, but decreased with C.p. and superimposed disuse (p = 0.005). GTS-21 attenuated BW loss in C.p. (p = 0.005). Compared to controls, TMM decreased with C.p. (0.43 ± 0.06 g to 0.26 ± 0.03 g) and with superimposed disuse (0.18 ± 0.04 g); GTS-21 ameliorated TMM loss to 0.32 ± 0.04 (no disuse, p = 0.028) and to 0.22 ± 0.03 (with disuse, p = 0.004). Tetanic tensions decreased with C.p. or disuse and GTS-21 attenuated tension decrease in animals with disuse (p = 0.006) and in animals with C.p. and disuse (p = 0.029). C.p.-induced 11-fold increased muscle α7nAChR expression was decreased by > 60% with GTS-21 treatment. In conclusion, GTS-21 modulates systemic inflammation, evidenced by both decreased methemoglobin levels and decrease of α7nAChR expression, and mitigates inflammation-mediated loss of BW, TMM, fiber size, and function.
Collapse
Affiliation(s)
- Stefan J Schaller
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA.
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany.
| | - Michio Nagashima
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA
- Department of Intensive Care Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Martin Schönfelder
- Institute of Exercise Biology, Technische Universität München, Georg-Brauchle-Ring 60/62, 80992, Munich, Germany
| | - Tomoki Sasakawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA
- Department of Anesthesiology and Critical Care Medicine, Asahikawa Medical University, 1 Chome-1-1 Midorigaoka Higashi 2 Jō, Asahikawa-shi, Hokkaidō, 078-8802, Japan
| | - Fabian Schulz
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany
| | - Mohammed A S Khan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA
| | - William R Kem
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, P.O. Box 100267, Gainesville, FL, 32610-0267, USA
| | - Gerhard Schneider
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany
| | - Jürgen Schlegel
- Institute of Pathology, Technische Universität München, Ismaningerstr. 22, 81675, Munich, Germany
| | - Heidrun Lewald
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany
| | - Manfred Blobner
- Klinik für Anaesthesiologie, Klinikum rechts der Isar, Technische Universität München, Ismaningertr. 22, 81675, Munich, Germany
| | - J A Jeevendra Martyn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children®-Boston, Harvard Medical School, 55 Fruit St, Boston, MA, 02114, USA
| |
Collapse
|
46
|
Medrinal C, Combret Y, Prieur G, Robledo Quesada A, Bonnevie T, Gravier FE, Dupuis Lozeron E, Frenoy E, Contal O, Lamia B. Comparison of exercise intensity during four early rehabilitation techniques in sedated and ventilated patients in ICU: a randomised cross-over trial. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:110. [PMID: 29703223 PMCID: PMC5923017 DOI: 10.1186/s13054-018-2030-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 04/06/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND In the ICU, out-of-bed rehabilitation is often delayed and in-bed exercises are generally low-intensity. Since the majority of rehabilitation is carried out in bed, it is essential to carry out the exercises that have the highest intensity. The aim of this study was to compare the physiological effects of four common types of bed exercise in intubated, sedated patients confined to bed in the ICU, in order to determine which was the most intensive. METHODS A randomised, single-blind, placebo-controlled crossover trial was carried out to evaluate the effects of four bed exercises (passive range of movements (PROM), passive cycle-ergometry, quadriceps electrical stimulation and functional electrical stimulation (FES) cycling) on cardiac output. Each exercise was carried out for ten minutes in ventilated, sedated patients. Cardiac output was recorded using cardiac Doppler ultrasound. The secondary aims were to evaluate right heart function and pulmonary and systemic artery pressures during the exercises, and the microcirculation of the vastus lateralis muscle. RESULTS The results were analysed in 19 patients. FES cycling was the only exercise that increased cardiac output, with a mean increase of 1 L/min (15%). There was a concomitant increase in muscle oxygen uptake, suggesting that muscle work occurred. FES cycling thus constitutes an effective early rehabilitation intervention. No muscle or systemic effects were induced by the passive techniques. CONCLUSION Most bed exercises were low-intensity and induced low levels of muscle work. FES cycling was the only exercise that increased cardiac output and produced sufficient intensity of muscle work. Longer-term studies of the effect of FES cycling on functional outcomes should be carried out. TRIAL REGISTRATION ClinicalTrials.gov, NCT02920684 . Registered on 30 September 2016. Prospectively registered.
Collapse
Affiliation(s)
- Clément Medrinal
- Normandie Univ, UNIROUEN, UPRES EA3830 - GRHV, Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France. .,Intensive Care Unit Department, Groupe Hospitalier du Havre, Hôpital Jacques Monod, Pierre Mendes France, 76290, Montivilliers, France.
| | - Yann Combret
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle de Pneumologie, ORL & Dermatologie, Université Catholique de Louvain, 1200, Brussels, Belgium.,Physiotherapy Department, Groupe Hospitalier du Havre, avenue Pierre Mendes France, 76290, Montivilliers, France
| | - Guillaume Prieur
- Intensive Care Unit Department, Groupe Hospitalier du Havre, Hôpital Jacques Monod, Pierre Mendes France, 76290, Montivilliers, France
| | - Aurora Robledo Quesada
- Intensive Care Unit Department, Groupe Hospitalier du Havre, Hôpital Jacques Monod, Pierre Mendes France, 76290, Montivilliers, France
| | - Tristan Bonnevie
- Normandie Univ, UNIROUEN, EA3830 - GRHV, 76000, Rouen, France.,Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France.,ADIR Association, Bois Guillaume, France
| | | | - Elise Dupuis Lozeron
- Division of Clinical Epidemiology, Geneva University Hospitals, Geneva, Switzerland
| | - Eric Frenoy
- Intensive Care Unit Department Department, Groupe Hospitalier du Havre, Hôpital Jacques Monod, 76290, Montivilliers, France
| | - Olivier Contal
- University of Applied Sciences and Arts Western Switzerland (HES-SO), avenue de Beaumont, 1011, Lausanne, Switzerland
| | - Bouchra Lamia
- Normandie Univ, UNIROUEN, EA3830 - GRHV, 76000, Rouen, France.,Institute for Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France.,Intensive Care Unit, Respiratory Department, Rouen University Hospital, Rouen, France.,Pulmonology Department, Groupe Hospitalier du Havre, avenue Pierre Mendes France, 76290, Montivilliers, France
| |
Collapse
|
47
|
Maffiuletti NA, Gondin J, Place N, Stevens-Lapsley J, Vivodtzev I, Minetto MA. Clinical Use of Neuromuscular Electrical Stimulation for Neuromuscular Rehabilitation: What Are We Overlooking? Arch Phys Med Rehabil 2017; 99:806-812. [PMID: 29233625 DOI: 10.1016/j.apmr.2017.10.028] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/18/2017] [Accepted: 10/31/2017] [Indexed: 12/19/2022]
Abstract
The clinical success of neuromuscular electrical stimulation (NMES) for neuromuscular rehabilitation is greatly compromised by the poor consideration of different physiological and methodological issues that are not always obvious to the clinicians. Therefore, the aim of this narrative review is to reexamine some of these fundamental aspects of NMES using a tripartite model perspective. First, we contend that NMES does not actually bypass the central nervous system but results in a multitude of neurally mediated responses that contribute substantially to force generation and may engender neural adaptations. Second, we argue that too much emphasis is generally placed on externally controllable stimulation parameters while the major determinant of NMES effectiveness is the intrinsically determined muscle tension generated by the current (ie, evoked force). Third, we believe that a more systematic approach to NMES therapy is required in the clinic and this implies a better identification of the patient-specific impairment and of the potential "responders" to NMES therapy. On the basis of these considerations, we suggest that the crucial steps to ensure the clinical effectiveness of NMES treatment should consist of (1) identifying the neuromuscular impairment with clinical assessment and (2) implementing algorithm-based NMES therapy while (3) properly dosing the treatment with tension-controlled NMES and eventually amplifying its neural effects.
Collapse
Affiliation(s)
| | - Julien Gondin
- NeuroMyoGene Institute, University Claude Bernard Lyon 1, INSERM U1217, CNRS UMR 5310, Villeurbanne, France
| | - Nicolas Place
- Institute of Sport Sciences, Faculty of Biology Medicine, University of Lausanne, Lausanne, Switzerland
| | - Jennifer Stevens-Lapsley
- Physical Therapy Program, Department of Physical Medicine and Rehabilitation, University of Colorado, Aurora, CO; Geriatric Research Education and Clinical Center, VA Eastern Colorado Healthcare System, Denver, CO
| | - Isabelle Vivodtzev
- INSERM U1042, HP2 Laboratory (Hypoxia: Pathophysiology), University Grenoble Alps, Grenoble, France
| | - Marco A Minetto
- Division of Physical Medicine and Rehabilitation, Department of Surgical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
48
|
Latronico N, Herridge M, Hopkins RO, Angus D, Hart N, Hermans G, Iwashyna T, Arabi Y, Citerio G, Ely EW, Hall J, Mehta S, Puntillo K, Van den Hoeven J, Wunsch H, Cook D, Dos Santos C, Rubenfeld G, Vincent JL, Van den Berghe G, Azoulay E, Needham DM. The ICM research agenda on intensive care unit-acquired weakness. Intensive Care Med 2017; 43:1270-1281. [PMID: 28289812 DOI: 10.1007/s00134-017-4757-5] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/02/2017] [Indexed: 12/23/2022]
Abstract
We present areas of uncertainty concerning intensive care unit-acquired weakness (ICUAW) and identify areas for future research. Age, pre-ICU functional and cognitive state, concurrent illness, frailty, and health trajectories impact outcomes and should be assessed to stratify patients. In the ICU, early assessment of limb and diaphragm muscle strength and function using nonvolitional tests may be useful, but comparison with established methods of global and specific muscle strength and physical function and determination of their reliability and normal values would be important to advance these techniques. Serial measurements of limb and respiratory muscle strength, and systematic screening for dysphagia, would be helpful to clarify if and how weakness of these muscle groups is independently associated with outcome. ICUAW, delirium, and sedatives and analgesics may interact with each other, amplifying the effects of each individual factor. Reduced mobility in patients with hypoactive delirium needs investigations into dysfunction of central and peripheral nervous system motor pathways. Interventional nutritional studies should include muscle mass, strength, and physical function as outcomes, and prioritize elucidation of mechanisms. At follow-up, ICU survivors may suffer from prolonged muscle weakness and wasting and other physical impairments, as well as fatigue without demonstrable weakness on examination. Further studies should evaluate the prevalence and severity of fatigue in ICU survivors and define its association with psychiatric disorders, pain, cognitive impairment, and axonal loss. Finally, methodological issues, including accounting for baseline status, handling of missing data, and inclusion of patient-centered outcome measures should be addressed in future studies.
Collapse
Affiliation(s)
- Nicola Latronico
- Department of Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy.
- Department of Anesthesia, Critical Care and Emergency, Spedali Civili University Hospital, Brescia, Italy.
| | - Margaret Herridge
- Interdepartmental Division of Critical Care Medicine, Institute of Medical Science, Toronto General Research Institute, University of Toronto, University Health Network, Toronto, Canada
| | - Ramona O Hopkins
- Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Murray, UT, USA
- Psychology Department and Neuroscience Center, Brigham Young University, Provo, UT, USA
| | - Derek Angus
- Department of Critical Care Medicine, University of Pittsburgh Schools of Medicine, Pittsburgh, PA, USA
| | - Nicholas Hart
- St Thomas' Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Greet Hermans
- Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
- Medical Intensive-Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Louvain, Belgium
| | - Theodore Iwashyna
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Center for Clinical Management Research, VA Ann Arbor Health System, Ann Arbor, MI, USA
| | - Yaseen Arabi
- Intensive Care Department, Respiratory Services, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Giuseppe Citerio
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- Neurointensive Care, San Gerardo Hospital, ASST-Monza, Monza, Italy
| | - E. Wesley Ely
- Department of Medicine, Pulmonary and Critical Care and Health Services, Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- The Tennessee Valley Veteran's Affairs Geriatric Research Education, Clinical Center (GRECC), Nashville, TN, USA
| | | | - Sangeeta Mehta
- Department of Medicine, Sinai Health System, Toronto, Canada
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada
| | - Kathleen Puntillo
- Department of Physiological Nursing, University of California, San Francisco, USA
| | | | - Hannah Wunsch
- Department of Critical Care Medicine, Sunnybrook Health Sciences Center, Toronto, ON, Canada
- Interdepartmental Division of Critical Care Medicine, Department of Anesthesia, University of Toronto, Toronto, ON, Canada
| | - Deborah Cook
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Department of Critical Care, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Claudia Dos Santos
- Keenan and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, ON, Canada
- Interdepartmental Division of Critical Care, Department of Medicine and Institute of Medical Sciences, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Gordon Rubenfeld
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada
- Program in Trauma, Emergency and Critical Care, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Greet Van den Berghe
- Department and Laboratory of Intensive Care Medicine, KU Leuven, Louvain, Belgium
| | - Elie Azoulay
- Medical Intensive Care Unit, Hôpital Saint-Louis, ECSTRA Team, Biostatistics and Clinical Epidemiology, UMR 1153 (Center of Epidemiology and Biostatistics Sorbonne Paris Cité, CRESS), INSERM, Paris Diderot Sorbonne University, Paris, France
| | - Dale M Needham
- Division of Pulmonary and Critical Care Medicine, Outcomes After Critical Illness and Surgery (OACIS) Group, Department of Physical Medicine and Rehabilitation, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
49
|
Abstract
OBJECTIVE In sepsis, the disease course of critically ill patients is often complicated by muscle failure leading to ICU-acquired weakness. The myokine transforming growth factor-β1 increases during inflammation and mediates muscle atrophy in vivo. We observed that the transforming growth factor-β1 inhibitor, secreted frizzled-related protein 2, was down-regulated in skeletal muscle of ICU-acquired weakness patients. We hypothesized that secreted frizzled-related protein 2 reduction enhances transforming growth factor-β1-mediated effects and investigated the interrelationship between transforming growth factor-β1 and secreted frizzled-related protein 2 in inflammation-induced atrophy. DESIGN Observational study and prospective animal trial. SETTING Two ICUs and research laboratory. PATIENTS/SUBJECTS Twenty-six critically ill patients with Sequential Organ Failure Assessment scores greater than or equal to 8 underwent a skeletal muscle biopsy from the vastus lateralis at median day 5 in ICU. Four patients undergoing elective orthopedic surgery served as controls. To search for signaling pathways enriched in muscle of ICU-acquired weakness patients, a gene set enrichment analysis of our recently published gene expression profiles was performed. Quantitative reverse transcriptase-polymerase chain reaction, Western blot, and immunohistochemistry were used to analyze secreted frizzled-related protein 2 expression and protein content. A mouse model of inflammation-induced skeletal muscle atrophy due to polymicrobial sepsis and cultured myocytes were used for mechanistic analyses. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Gene set enrichment analysis uncovered transforming growth factor-β1 signaling activation in vastus lateralis from ICU-acquired weakness patients. Muscular secreted frizzled-related protein 2 expression was reduced after 5 days in ICU. Likewise, muscular secreted frizzled-related protein 2 expression was decreased early and continuously in mice with inflammation-induced atrophy. In muscle, secreted frizzled-related protein 2 was predominantly contained in fast twitch/type II myofibers. Secreted frizzled-related protein 2 physically interacted and colocalized with transforming growth factor-β1 through its cysteine-rich domain. Finally, secreted frizzled-related protein 2 prevented transforming growth factor-β1-induced atrophy in C2C12 myotubes. CONCLUSIONS Muscular secreted frizzled-related protein 2 is down-regulated in ICU-acquired weakness patients and mice with inflammation-induced muscle atrophy. Decreased secreted frizzled-related protein 2 possibly establishes a positive feedback loop enhancing transforming growth factor-β1-mediated atrophic effects in inflammation-induced atrophy.
Collapse
|
50
|
Effect of neuromuscular stimulation and individualized rehabilitation on muscle strength in Intensive Care Unit survivors: A randomized trial. J Crit Care 2017; 40:76-82. [PMID: 28364678 DOI: 10.1016/j.jcrc.2017.03.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 02/20/2017] [Accepted: 03/19/2017] [Indexed: 01/16/2023]
Abstract
PURPOSE Intensive Care Unit (ICU) survivors experience muscle weakness leading to restrictions in functional ability. Neuromuscular electrical stimulation (NMES) has been an alternative to exercise in critically ill patients. The aim of our study was to investigate its effects along with individualized rehabilitation on muscle strength of ICU survivors. MATERIAL AND METHODS Following ICU discharge, 128 patients (age: 53±16years) were randomly assigned to daily NMES sessions and individualized rehabilitation (NMES group) or to control group. Muscle strength was assessed by the Medical Research Council (MRC) score and hand grip at hospital discharge. Secondary outcomes were functional ability and hospital length of stay. RESULTS MRC, handgrip, functional status and hospital length of stay did not differ at hospital discharge between groups (p>0.05). ΔMRC% one and two weeks after ICU discharge tended to be higher in NMES group, while it was significant higher in NMES group of patients with ICU-acquired weakness at two weeks (p=0.05). CONCLUSIONS NMES and personalized physiotherapy in ICU survivors did not result in greater improvement of muscle strength and functional status at hospital discharge. However, in patients with ICU-aw NMES may be effective. The potential benefits of rehabilitation strategies should be explored in larger number of patients in future studies. CLINICAL TRIAL REGISTRATION www.Clinicaltrials.gov: NCT01717833.
Collapse
|