1
|
Castellví-Font A, Goligher EC, Dianti J. Lung and Diaphragm Protection During Mechanical Ventilation in Patients with Acute Respiratory Distress Syndrome. Clin Chest Med 2024; 45:863-875. [PMID: 39443003 DOI: 10.1016/j.ccm.2024.08.007] [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] [Indexed: 10/25/2024]
Abstract
Patients with acute respiratory distress syndrome often require mechanical ventilation to maintain adequate gas exchange and to reduce the workload of the respiratory muscles. Although lifesaving, positive pressure mechanical ventilation can potentially injure the lungs and diaphragm, further worsening patient outcomes. While the effect of mechanical ventilation on the risk of developing lung injury is widely appreciated, its potentially deleterious effects on the diaphragm have only recently come to be considered by the broader intensive care unit community. Importantly, both ventilator-induced lung injury and ventilator-induced diaphragm dysfunction are associated with worse patient-centered outcomes.
Collapse
Affiliation(s)
- Andrea Castellví-Font
- Critical Care Department, Hospital del Mar de Barcelona, Critical Illness Research Group (GREPAC), Hospital del Mar Research Institute (IMIM), Passeig Marítim de la Barceloneta 25-29, Ciutat Vella, 08003, Barcelona, Spain; Interdepartmental Division of Critical Care Medicine, University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada; Division of Respirology, Department of Medicine, University Health Network, Toronto, Canada
| | - Ewan C Goligher
- Interdepartmental Division of Critical Care Medicine, University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada; Division of Respirology, Department of Medicine, University Health Network, Toronto, Canada; University Health Network/Sinai Health System, University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada; Toronto General Hospital Research Institute, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada; Department of Physiology, University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada.
| | - Jose Dianti
- Critical Care Medicine Department, Centro de Educación Médica e Investigaciones Clínicas "Norberto Quirno" (CEMIC), Av. E. Galván 4102, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
2
|
Rohrs EC, Fernandez KC, Bassi TG, Nicholas M, Wittmann J, Ornowska M, Gani M, Dakin I, Reynolds SC. Transvenous phrenic nerve stimulation reduces diaphragm injury during controlled mechanical ventilation in a preclinical model of ARDS. J Appl Physiol (1985) 2024; 137:1175-1181. [PMID: 39262337 PMCID: PMC11573254 DOI: 10.1152/japplphysiol.00884.2023] [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: 12/11/2023] [Revised: 06/11/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
Patients with acute respiratory distress syndrome (ARDS) require periods of deep sedation and mechanical ventilation, leading to diaphragm dysfunction. Our study seeks to determine whether the combination of temporary transvenous diaphragm neurostimulation (TTDN) and mechanical ventilation changes the degree of diaphragm injury and cytokines concentration in a preclinical ARDS model. Moderate ARDS was induced in pigs using oleic acid, followed by ventilation for 12 h post-injury with volume-control at 8 mL/kg, positive end-expiratory pressure (PEEP) 5 cmH2O, respiratory rate and [Formula: see text] set to achieve normal arterial blood gases. Two groups received TTDN: every second breath (MV + TTDN50%, n = 6) or every breath (MV + TTDN100%, n = 6). One group received ventilation only (MV, n = 6). Full-thickness diaphragm and quadricep muscle biopsies were taken at study end. Samples were fixed and stained with hematoxylin and eosin and a point-counting technique was applied to calculate abnormal muscle area fraction. Cytokine concentrations were measured in homogenized tissue using porcine-specific enzyme-linked immunosorbent assay (ELISA) and compared with serum samples. Percentage of abnormal diaphragm tissue was different between MV [8.1% (6.0-8.8)] versus MV + TTDN50% [3.4% (2.1-4.8)], P = 0.010 and MV versus MV + TTDN100% [3.1% (2.5-4.0)], P = 0.005. Percentage of abnormal quadriceps tissue was not different between groups. Cytokine concentration patterns in diaphragm samples were different between all groups (P < 0.001) and the interaction between TTDN application and resultant cytokine concentration pattern was significant (P = 0.025). TTDN, delivered in synchrony with mechanical ventilation, mitigated diaphragm injury, as evidenced by less abnormal tissue in the diaphragm samples, in pigs with oleic acid-induced ARDS and is an exciting tool for lung and diaphragm-protective ventilation.NEW & NOTEWORTHY This study adds to our understanding of applying transvenous diaphragm neurostimulation synchronously with mechanical ventilation by examining its effects on diaphragm muscle injury and cytokine concentration patterns in pigs with acute respiratory distress syndrome (ARDS). We observed that using this therapy for 12 h post lung injury mitigated ventilator-induced diaphragm injury and changed the pattern of cytokine concentration measured in diaphragm tissue. These findings suggest that transvenous diaphragm neurostimulation is an exciting tool for lung and diaphragm protective ventilation.
Collapse
Affiliation(s)
- Elizabeth C Rohrs
- Simon Fraser University, Burnaby, British Columbia, Canada
- RCHF Advancing Innovation in Medicine Institute, New Westminster, British Columbia, Canada
| | | | - Thiago G Bassi
- Simon Fraser University, Burnaby, British Columbia, Canada
- Lungpacer Medical USA, Inc., Exton, Pennsylvania, United States
| | | | - Jessica Wittmann
- RCHF Advancing Innovation in Medicine Institute, New Westminster, British Columbia, Canada
| | - Marlena Ornowska
- Simon Fraser University, Burnaby, British Columbia, Canada
- RCHF Advancing Innovation in Medicine Institute, New Westminster, British Columbia, Canada
| | - Matt Gani
- Lungpacer Medical USA, Inc., Exton, Pennsylvania, United States
| | | | - Steven C Reynolds
- Simon Fraser University, Burnaby, British Columbia, Canada
- RCHF Advancing Innovation in Medicine Institute, New Westminster, British Columbia, Canada
| |
Collapse
|
3
|
Machado-Junior PA, Dias MSS, de Souza ABF, Lopes LSE, Menezes TP, Talvani A, Brochard L, Bezerra FS. A short duration of mechanical ventilation alters redox status in the diaphragm and aggravates inflammation in septic mice. Respir Physiol Neurobiol 2024; 331:104361. [PMID: 39433197 DOI: 10.1016/j.resp.2024.104361] [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: 09/04/2024] [Revised: 10/07/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND Mechanical ventilation (MV) is a life support method used to treat patients with respiratory failure. High tidal volumes during MV can cause ventilator-induced lung injury (VILI), but also affect other organs, such as the diaphragm (Dia) causing ventilator-induced diaphragmatic dysfunction (VIDD). VIDD is often associated with a complicated course on MV. Sepsis can induce inflammation and oxidative stress, contributing to the impairment of the Dia and worsening of the prognosis. This study evaluated the additive or synergistic effects of a short course of mechanical ventilation on Dia in healthy and septic adult mice. METHODS 32 adult male C57BL/6 mice were randomly into four groups: Control (CG), non-ventilated animals instilled with saline solution (PBS1x); Lipopolysaccharide (LPS), non-ventilated animals instilled with PBS solution containing lipopolysaccharide; Mechanical Ventilation (MV) for 1 h, ventilated animals instilled with PBS solution; and Mechanical Ventilation and LPS (MV+LPS), ventilated animals instilled with PBS solution containing LPS. At the end of the experimental protocol, the animals were euthanized, then blood and diaphragm tissue samples were collected. RESULTS Evaluation of leukocyte/blood parameters and diaphragm muscle showed that MV, LPS and the combination of both were able to increase neutrophil count, creatine kinase, inflammatory mediators and oxidative stress in all groups compared to the control. MV and sepsis combined had additive effects on inflammation and lipid peroxidation. CONCLUSIONS A short course of Mechanical ventilation promotes inflammation and oxidative stress and, its combination with sepsis further increases local and systemic inflammation.
Collapse
Affiliation(s)
- Pedro Alves Machado-Junior
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Marcelo Santiago Soares Dias
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Leonardo Spinelli Estevão Lopes
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Tatiana Prata Menezes
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Laurent Brochard
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences and Center of Research in Biological Sciences, Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Nascimento MS, Leite FS, Silva PAL, Zamberlan GC, de Souza PM, Eid RAC, Zacharias RSB, do Prado C, Batista CL. Reliability and reference values for diaphragmatic excursion, thickness, and thickening fraction and quadriceps femoris muscle thickness in full-term newborns evaluated by ultrasound. Eur J Pediatr 2024; 183:3453-3460. [PMID: 38771374 DOI: 10.1007/s00431-024-05608-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 05/22/2024]
Abstract
To determine the diaphragm thickness, thickening fraction, and excursion and thickness of the quadriceps femoris muscle in full-term newborns and to evaluate the intra- and interrater reliability of these measurements. This was a prospective, observational clinical study including full-term newborns born within the first 48 h after birth. Serial measurements of the thickness, thickening fraction, and mobility of the diaphragm muscles and the thickness of the quadriceps muscle were obtained using ultrasound images. A total of 69 newborns with a mean gestational age of 39 weeks were included. The following measurements were obtained and are expressed as the mean (standard deviation): inspiratory diaphragm thickness, 0.19 cm (0.04); expiratory diaphragm thickness, 0.16 cm (0.04); diaphragm thickness fraction, 16.70 cm (10.27); diaphragmatic excursion, 0.68 cm (0.22); and quadriceps thickness, 0.99 cm (0.14). Intrarater reliability was assessed using intraclass correlation coefficients (ICCs). Excellent intrarater agreement was observed for the two groups of operators (ICC > 0.86, p < 0.001) for all measurements except for the diaphragm thickening fraction, which showed good agreement for both operator groups (ICC = 0.70, p < 0.001). Regarding interrater reliability, moderate agreement between the raters was observed in the means of all measures (ICC > 0.49, p < 0.001), except for the diaphragm thickening fraction, which showed poor agreement. Conclusion: Good intrarater and moderate interrater reliability were achieved in ultrasound evaluations of the thickness and mobility of the diaphragm and quadriceps femoris muscles in full-term newborns, demonstrating the feasibility of this technique for clinical use. This pioneering study offers reference values for these muscles in a single study, allowing comparisons between different clinical conditions. What is Known: • Ultrasound is a highly reliable tool for muscle assessment that can be used to assess muscular atrophy in critically ill patients. • Muscle atrophy worsens the patient's condition and has been associated with worse outcomes. What is New: • To our knowledge, this is the first study to jointly evaluate the diaphragm and quadriceps muscle thickness and evaluate the reliability of all measurements. • Our study presents reference values for both muscles, enabling comparisons between different clinical conditions.
Collapse
Affiliation(s)
- Milena Siciliano Nascimento
- Departamento de Práticas Assistenciais, Hospital Israelita Albert Einstein, Avenue Albert Einstein, 627-701, 05651-901, São Paulo, Brazil.
| | - Flávia Sales Leite
- Departamento de Pacientes Graves, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | - Priscila Monteiro de Souza
- Departamento de Práticas Assistenciais, Hospital Israelita Albert Einstein, Avenue Albert Einstein, 627-701, 05651-901, São Paulo, Brazil
- Corpo Discente da Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, Brazil
| | | | | | - Cristiane do Prado
- Departamento Materno-infantil, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Carla Luciana Batista
- Departamento de Pacientes Graves, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
5
|
Itagaki T, Akimoto Y, Takashima T, Oto J. Ultrasonographic Assessment of the Diaphragm. Diagnostics (Basel) 2024; 14:1481. [PMID: 39061618 PMCID: PMC11276413 DOI: 10.3390/diagnostics14141481] [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: 06/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Mechanical ventilation injures not only the lungs but also the diaphragm, resulting in dysfunction associated with poor outcomes. Diaphragm ultrasonography is a noninvasive, cost-effective, and reproducible diagnostic method used to monitor the condition and function of the diaphragm. With advances in ultrasound technology and the expansion of its clinical applications, diaphragm ultrasonography has become increasingly important as a tool to visualize and quantify diaphragmatic morphology and function across multiple medical specialties, including pulmonology, critical care, and rehabilitation medicine. This comprehensive review aims to provide an in-depth analysis of the role and limitations of ultrasonography in assessing the diaphragm, especially among critically ill patients. Furthermore, we discuss a recently published expert consensus and provide a perspective for the future.
Collapse
Affiliation(s)
- Taiga Itagaki
- Department of Emergency and Disaster Medicine, Tokushima University Hospital, 2-50-1 Kuramoto, Tokushima 770-8503, Japan
| | - Yusuke Akimoto
- Emergency Department, Tokushima Prefectural Miyoshi Hospital, 815-2 Ikedacho Shima, Miyoshi 778-0005, Japan;
| | - Takuya Takashima
- Department of Emergency and Critical Care Medicine, Tokushima University Graduate Hospital of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima 770-8503, Japan; (T.T.); (J.O.)
| | - Jun Oto
- Department of Emergency and Critical Care Medicine, Tokushima University Graduate Hospital of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima 770-8503, Japan; (T.T.); (J.O.)
| |
Collapse
|
6
|
Sedraoui S, Leduc-Gaudet JP, Mayaki D, Moamer A, Huck L, Gouspillou G, Petrof BJ, Hussain S. Lack of compensatory mitophagy in skeletal muscles during sepsis. J Physiol 2024; 602:2823-2838. [PMID: 38748778 DOI: 10.1113/jp286216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/02/2024] [Indexed: 06/15/2024] Open
Abstract
Skeletal muscle dysfunction is a major problem in critically ill patients suffering from sepsis. This condition is associated with mitochondrial dysfunction and increased autophagy in skeletal muscles. Autophagy is a proteolytic mechanism involved in eliminating dysfunctional cellular components, including mitochondria. The latter process, referred to as mitophagy, is essential for maintaining mitochondrial quality and skeletal muscle health. Recently, a fluorescent reporter system called mito-QC (i.e. mitochondrial quality control) was developed to specifically quantify mitophagy levels. In the present study, we used mito-QC transgenic mice and confocal microscopy to morphologically monitor mitophagy levels during sepsis. To induce sepsis, Mito-QC mice received Escherichia coli lipopolysaccharide (10 mg kg-1 i.p.) or phosphate-buffered saline and skeletal muscles (hindlimb and diaphragm) were excised 48 h later. In control groups, there was a negative correlation between the basal mitophagy level and overall muscle mitochondrial content. Sepsis increased general autophagy in both limb muscles and diaphragm but had no effect on mitophagy levels. Sepsis was associated with a downregulation of certain mitophagy receptors (Fundc1, Bcl2L13, Fkbp8 and Phbb2). The present study suggests that general autophagy and mitophagy can be dissociated from one another, and that the characteristic accumulation of damaged mitochondria in skeletal muscles under the condition of sepsis may reflect a failure of adequate compensatory mitophagy. KEY POINTS: There was a negative correlation between the basal level of skeletal muscle mitophagy and the mitochondrial content of individual muscles. Mitophagy levels in limb muscles and the diaphragm were unaffected by lipopolysaccharide (LPS)-induced sepsis. With the exception of BNIP3 in sepsis, LPS administration induced either no change or a downregulation of mitophagy receptors in skeletal muscles.
Collapse
Affiliation(s)
- Sami Sedraoui
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Jean-Philippe Leduc-Gaudet
- Department of Medical Biology, Faculty of Health Sciences, Université du Québec à Trois-Rivieres, Trois-Rivieres, QC, Canada
| | - Dominique Mayaki
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Alaa Moamer
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Laurent Huck
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Gilles Gouspillou
- Département des Sciences de l'Activité Physique, Faculté des Sciences, Université du Québec à Montréal, Montréal, QC, Canada
| | - Basil J Petrof
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
| | - Sabah Hussain
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montreal, QC, Canada
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montral, QC, Canada
- Department of Critical Care Medicine, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
7
|
Farley C, Brooks D, Newman ANL. The effects of inspiratory muscle training on physical function in critically ill adults: Protocol for a systematic review and meta-analysis. PLoS One 2024; 19:e0300605. [PMID: 38517914 PMCID: PMC10959358 DOI: 10.1371/journal.pone.0300605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/29/2024] [Indexed: 03/24/2024] Open
Abstract
INTRODUCTION Inspiratory muscle training (IMT) is one possible strategy to ameliorate respiratory muscle weakness due to invasive mechanical ventilation. Recent systematic reviews have focused on respiratory outcomes with minimal attention to physical function. The newest systematic review searched the literature until September 2017 and a recent preliminary search identified 5 new randomized controlled trials focusing on IMT in critical care. As such, a new systematic review is warranted to summarize the current body of evidence and to investigate the effect of IMT on physical function in critical care. MATERIALS AND METHODS We will search for three main concepts ("critical illness", "inspiratory muscle training", "RCT") across six databases from their inception (MEDLINE, EMBASE, Emcare, AMED, CINAHL, CENTRAL) and ClinicalTrials.gov. Two reviewers will independently screen titles, abstracts, and full texts for eligibility using the Covidence web-based software. Eligible studies must include: (1) adult (≥18 years) patients admitted to the intensive care unit (ICU) who required invasive mechanical ventilation for ≥24 hours, (2) an IMT intervention using a threshold device with the goal of improving inspiratory muscle strength, with or without usual care, and (3) randomized controlled trial design. The primary outcome of interest will be physical function. We will use the Cochrane Risk of Bias Tools (ROB2) and will assess the quality of the evidence using the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) tool. This protocol has been reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols (PRISMA- P) guidelines and is registered with the International Prospective Register of Systematic Reviews (PROSPERO). CONCLUSION Results will summarize the body of evidence of the effect of IMT on physical function in critically ill patients. We will submit our findings to a peer-reviewed journal and share our results at conferences.
Collapse
Affiliation(s)
- Christopher Farley
- Faculty of Health Science, School of Rehabilitation Science, McMaster University, Hamilton, ON, Canada
| | - Dina Brooks
- Faculty of Health Science, School of Rehabilitation Science, McMaster University, Hamilton, ON, Canada
- Department of Respiratory Medicine, West Park Healthcare Centre, Toronto, ON, Canada
- Faculty of Medicine, Department of Physical Therapy, University of Toronto, Toronto, ON, Canada
- Rehabilitation Sciences Institute, School of Graduate Studies, University of Toronto, Toronto, ON, Canada
- Faculty of Medicine, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anastasia N. L. Newman
- Faculty of Health Science, School of Rehabilitation Science, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
8
|
Yu R, Okazaki T, Ren Y, Okuyama J, Ebihara S, Izumi SI. Effects of body postures on respiratory muscle force and coughing in healthy people. Respir Physiol Neurobiol 2024; 319:104181. [PMID: 37866524 DOI: 10.1016/j.resp.2023.104181] [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: 07/21/2023] [Revised: 10/03/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
The respiratory muscle force determines the intensity of cough force. A greater cough force for cleaning the airways is essential for preventing and managing pneumonia. Body posture can affect the onset of aspiration pneumonia. However, the effects of body posture on the respiratory muscle and cough forces remain unclear. Thus, we aimed to explore the influence of the four body postures on respiratory muscle force, cough pressure, subjective ease of coughing, and pulmonary function in healthy individuals. Twenty healthy individuals were included in this study. Body postures were 0-degree supine, 30- and 60-degree semi-recumbent, and 90-degree sitting. The maximal inspiratory and expiratory pressures, maximal cough pressure, subjective ease of coughing, and pulmonary function, including peak expiratory flow, were evaluated. We set the measured values in the supine posture to 100% and showed the relative values. The 60-degree posture showed stronger inspiratory (125.1 ± 3.9%, mean ± standard error [SE]) and expiratory (116.4 ± 3.0%) muscle force, cough pressure, more subjective ease of coughing, and greater peak expiratory flow (113.4 ± 3.0%) than the supine posture. The sitting posture also showed greater inspiratory muscle force and peak expiratory flow than the supine posture. The correlation coefficient for the 60-degree posture showed that the maximal inspiratory pressure was moderately correlated with the maximal expiratory pressure (r = 0.512), cough pressure (r = 0.495), and peak expiratory flow (r = 0.558). The above findings suggest the advantage of keeping a 60-degree posture and avoiding the supine posture to generate a greater cough force in the prevention and management of pneumonia.
Collapse
Affiliation(s)
- Rui Yu
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Tatsuma Okazaki
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan.
| | - Yuzhuo Ren
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Junko Okuyama
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Satoru Ebihara
- Department of Internal Medicine and Rehabilitation Science, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shin-Ichi Izumi
- Center for Dysphagia of Tohoku University Hospital, Sendai, Miyagi, Japan; Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Biomedical Engineering, Sendai, Miyagi, Japan
| |
Collapse
|
9
|
Medrinal C, Machefert M, Lamia B, Bonnevie T, Gravier FE, Hilfiker R, Prieur G, Combret Y. Transcutaneous electrical diaphragmatic stimulation in mechanically ventilated patients: a randomised study. Crit Care 2023; 27:338. [PMID: 37649092 PMCID: PMC10469422 DOI: 10.1186/s13054-023-04597-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/01/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Few specific methods are available to reduce the risk of diaphragmatic dysfunction for patients under mechanical ventilation. The number of studies involving transcutaneous electrical stimulation of the diaphragm (TEDS) is increasing but none report results for diaphragmatic measurements, and they lack power. We hypothesised that the use of TEDS would decrease diaphragmatic dysfunction and improve respiratory muscle strength in patients in ICU. METHODS We conducted a controlled trial to assess the impact of daily active electrical stimulation versus sham stimulation on the prevention of diaphragm dysfunction during the weaning process from mechanical ventilation. The evaluation was based on ultrasound measurements of diaphragm thickening fraction during spontaneous breathing trials. We also measured maximal inspiratory muscle pressure (MIP), peak cough flow (PEF) and extubation failure. RESULTS Sixty-six patients were included and randomised using a 1:1 ratio. The mean number of days of mechanical ventilation was 10 ± 6.8. Diaphragm thickening fraction was > 30% at the SBT for 67% of participants in the TEDS group and 54% of the Sham group (OR1.55, 95% CI 0.47-5.1; p = 0.47). MIP and PEF were similar in the TEDS and Sham groups (respectively 35.5 ± 11.9 vs 29.7 ± 11.7 cmH20; p = 0.469 and 83.2 ± 39.5 vs. 75.3 ± 34.08 L/min; p = 0.83). Rate of extubation failure was not different between groups. CONCLUSION TEDS did not prevent diaphragm dysfunction or improve inspiratory muscle strength in mechanically ventilated patients. TRIAL REGISTRATION Prospectively registered on the 20th November 2019 on ClinicalTrials.gov Identifier NCT04171024.
Collapse
Affiliation(s)
- Clément Medrinal
- Université Paris-Saclay, UVSQ, Erphan, 78000, Versailles, France.
- Intensive Care Unit Department, Le Havre Hospital, Avenue Pierre Mendes France, 76290, Montivilliers, France.
| | - Margaux Machefert
- Université Paris-Saclay, UVSQ, Erphan, 78000, Versailles, France
- Physiotherapy Department, Le Havre Hospital, Avenue Pierre Mendes France, 76290, Montivilliers, France
| | - Bouchra Lamia
- Normandie Univ, UNIROUEN, EA3830-GRHV, 76 000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76 000, Rouen, France
- Pulmonology Department, Le Havre Hospital, Avenue Pierre Mendes France, 76290, Montivilliers, France
- Pulmonology, Respiratory Department, Rouen University Hospital, Rouen, France
| | - Tristan Bonnevie
- Normandie Univ, UNIROUEN, EA3830-GRHV, 76 000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76 000, Rouen, France
- Adir Association, 76230, Bois Guillaume, France
| | - Francis-Edouard Gravier
- Normandie Univ, UNIROUEN, EA3830-GRHV, 76 000, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), 76 000, Rouen, France
- Adir Association, 76230, Bois Guillaume, France
| | - Roger Hilfiker
- Research and Independent Studies in Private Physiotherapy (RISE), 3902, Brig, Switzerland
| | - Guillaume Prieur
- Intensive Care Unit Department, Le Havre Hospital, Avenue Pierre Mendes France, 76290, Montivilliers, France
- Pulmonology Department, Le Havre Hospital, Avenue Pierre Mendes France, 76290, Montivilliers, France
| | - Yann Combret
- Université Paris-Saclay, UVSQ, Erphan, 78000, Versailles, France
- Intensive Care Unit Department, Le Havre Hospital, Avenue Pierre Mendes France, 76290, Montivilliers, France
- Pulmonology Department, Le Havre Hospital, Avenue Pierre Mendes France, 76290, Montivilliers, France
| |
Collapse
|
10
|
Mirea L, Cobilinschi C, Ungureanu R, Cotae AM, Darie R, Tincu R, Avram O, Constantinescu S, Minoiu C, Baetu A, Grintescu IM. A Trend towards Diaphragmatic Muscle Waste after Invasive Mechanical Ventilation in Multiple Trauma Patients-What to Expect? J Clin Med 2023; 12:jcm12093338. [PMID: 37176778 PMCID: PMC10179085 DOI: 10.3390/jcm12093338] [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: 12/26/2022] [Revised: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Considering the prioritization of life-threatening injuries in trauma care, secondary dysfunctions such as ventilator-induced diaphragmatic dysfunction (VIDD) are often overlooked. VIDD is an entity induced by muscle inactivity during invasive mechanical ventilation, associated with a profound loss of diaphragm muscle mass. In order to assess the incidence of VIDD in polytrauma patients, we performed an observational, retrospective, longitudinal study that included 24 polytraumatized patients. All included patients were mechanically ventilated for at least 48 h and underwent two chest CT scans during their ICU stay. Diaphragmatic thickness was measured by two independent radiologists on coronal and axial images at the level of celiac plexus. The thickness of the diaphragm was significantly decreased on both the left and right sides (left side: -0.82 mm axial p = 0.034; -0.79 mm coronal p = 0.05; right side: -0.94 mm axial p = 0.016; -0.91 coronal p = 0.013). In addition, we obtained a positive correlation between the number of days of mechanical ventilation and the difference between the two measurements of the diaphragm thickness on both sides (r =0.5; p = 0.02). There was no statistically significant correlation between the body mass indexes on admission, the use of vitamin C or N-acetyl cysteine, and the differences in diaphragmatic thickness.
Collapse
Affiliation(s)
- Liliana Mirea
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Cobilinschi
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Raluca Ungureanu
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ana-Maria Cotae
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Raluca Darie
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | - Radu Tincu
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
- Department of Clinical Toxicology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Oana Avram
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
- Department of Clinical Toxicology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Sorin Constantinescu
- Department of Radiology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology, Victor Atanasiu National Aviation and Space Medicine Institute, 010825 Bucharest, Romania
| | - Costin Minoiu
- Department of Radiology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
| | - Alexandru Baetu
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Anesthesiology and Intensive Care, Grigore Alexandrescu Clinical Emergency Hospital for Children, 011743 Bucharest, Romania
| | - Ioana Marina Grintescu
- Department of Anesthesiology and Intensive Care, Clinical Emergency Hospital Bucharest, 014461 Bucharest, Romania
- Department of Anesthesiology and Intensive Care II, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
11
|
Laguado-Nieto MA, Roberto-Avilán SL, Naranjo-Junoy F, Meléndez-Flórez HJ, Lozada-Martinez ID, Domínguez-Alvarado GA, Campos-Castillo VA, Ríos-Orozco SU, Narváez-Rojas AR. Diaphragmatic Dynamics and Thickness Parameters Assessed by Ultrasonography Predict Extubation Success in Critically Ill Patients. CLINICAL MEDICINE INSIGHTS: CIRCULATORY, RESPIRATORY AND PULMONARY MEDICINE 2023; 17:11795484231165940. [PMID: 37008792 PMCID: PMC10052899 DOI: 10.1177/11795484231165940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 03/08/2023] [Indexed: 03/29/2023] Open
Abstract
INTRODUCTION A frequent cause of weaning and extubation failure in critically ill mechanically ventilated patients is diaphragm muscle dysfunction. Ultrasound (US) evaluation of the diaphragm yields important data regarding its thickness (diaphragm thickening fraction [TFdi]) and its movement or excursion (diaphragmatic dynamics) that reveal the presence of diaphragmatic dysfunction. METHODS Cross-sectional study, which included patients older than 18 years with invasive mechanical ventilation with an expected duration of more than 48 h, in a tertiary referral center in Colombia. The excursion of the diaphragm, inspiratory and expiratory thickness, and TFdi were evaluated by US. Prevalence and use of medications were evaluated, and the association with failure in ventilatory weaning and extubation was analyzed. RESULTS Sixty-one patients were included. The median age and APACHE IV score were 62.42 years and 78.23, respectively. The prevalence of diaphragmatic dysfunction (assessed by excursion and TFdi) was 40.98%. The sensibility, specificity, positive predictive value, and negative predictive value for TFdi < 20% was 86%, 24%, 75%, and 40%, respectively, with an area under the receiver operating characteristic (ROC) curve of 0.6. The ultrasonographic analysis of excursion of the diaphragm, inspiratory and expiratory thickness, and TFdi (>20%) allow in its set and with normal values, predict success or failure for the extubation with an area under the ROC curve of 0.87. CONCLUSION Diaphragmatic dynamics and thickness parameters together assessed by ultrasonography could predict the success of extubation in critically ill patients in Colombia, based on the finding of diaphragmatic dysfunction.
Collapse
Affiliation(s)
| | | | - Francisco Naranjo-Junoy
- Department of Critical Medicine and Intensive Care, FOSCAL International Clinic, Bucaramanga, Colombia
| | | | - Ivan David Lozada-Martinez
- Medical and Surgical Research Center, Future Surgeons Chapter, Colombian Surgery Association, Bogotá, Colombia
- Grupo Prometheus y Biomedicina Aplicada a las Ciencias Clínicas, School of Medicine, Universidad de Cartagena, Cartagena, Colombia
- International Coalition on Surgical Research, Universidad Nacional Autónoma de Nicaragua, Managua, Nicaragua
| | | | | | | | - Alexis Rafael Narváez-Rojas
- International Coalition on Surgical Research, Universidad Nacional Autónoma de Nicaragua, Managua, Nicaragua
- Division of Breast Surgical Oncology, DeWitt Daughtry Family Department of Surgery, Jackson Health System / University of Miami Miller School of Medicine, Miami, FL, USA
- Alexis Rafael Narvaez-Rojas, International Coalition on Surgical Research, Universidad Nacional Autónoma de Nicaragua, Managua, Nicaragua; Breast Surgical Oncology Division, DeWitt Daughtry Family Department of Surgery, Jackson Health System / University of Miami Miller School of Medicine, FL, USA.
| |
Collapse
|
12
|
Miyatake M, Okazaki T, Suzukamo Y, Matsuyama S, Tsuji I, Izumi SI. High Mortality in an Older Japanese Population with Low Forced Vital Capacity and Gender-Dependent Potential Impact of Muscle Strength: Longitudinal Cohort Study. J Clin Med 2022; 11:jcm11185264. [PMID: 36142910 PMCID: PMC9505108 DOI: 10.3390/jcm11185264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/21/2022] Open
Abstract
Generally, weak muscle power is associated with high mortality. We aimed to evaluate the unknown association between % predicted value forced vital capacity (FVC% predicted) and mortality in asymptomatic older people, and the impact of muscle power on this association. We analyzed the Tsurugaya cohort that enrolled Japanese people aged ≥70 for 15 years with Cox proportional hazards model. Exposure variables were FVC% predicted and leg power. The outcome was all-cause mortality. The subjects were divided into quartiles by FVC% predicted or leg power, or into two groups by 80% for FVC% predicted or by the strongest 25% for leg power. Across 985 subjects, 262 died. The males with lower FVC% predicted exhibited higher mortality risks. The hazard ratio (HR) was 2.03 (95% CI 1.30−3.18) at the lowest relative to the highest groups. The addition of leg power reduced the HR to 1.78 (95% CI 1.12−2.80). In females, FVC% predicted under 80% was a risk factor and the HR was 1.67 (95% CI 1.05−2.64) without the effect of leg power. In FVC% predicted <80% males HRs were 2.44 (95% CI 1.48−4.02) in weak and 1.38 (95% CI 0.52−3.64) in strong leg power males, relative to ≥80% and strong leg power males. Low FVC% predicted was associated with high mortality with potential unfavorable effects of weak leg power in males.
Collapse
Affiliation(s)
- Midori Miyatake
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Tatsuma Okazaki
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Dysphagia of Tohoku University Hospital, Sendai 980-8575, Japan
- Correspondence: ; Tel.: +81-22-717-7338
| | - Yoshimi Suzukamo
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Sanae Matsuyama
- Division of Epidemiology, Department of Health Informatics and Public Health, School of Public Health, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Ichiro Tsuji
- Division of Epidemiology, Department of Health Informatics and Public Health, School of Public Health, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Shin-Ichi Izumi
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
- Center for Dysphagia of Tohoku University Hospital, Sendai 980-8575, Japan
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Biomedical Engineering, Sendai 980-8575, Japan
| |
Collapse
|
13
|
Lecronier M, Jung B, Molinari N, Pinot J, Similowski T, Jaber S, Demoule A, Dres M. Severe but reversible impaired diaphragm function in septic mechanically ventilated patients. Ann Intensive Care 2022; 12:34. [PMID: 35403916 PMCID: PMC9001790 DOI: 10.1186/s13613-022-01005-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Whether sepsis-associated diaphragm dysfunction may improve despite the exposure of mechanical ventilation in critically ill patients is unclear. This study aims at describing the diaphragm function time course of septic and non-septic mechanically ventilated patients. Methods Secondary analysis of two prospective observational studies of mechanically ventilated patients in whom diaphragm function was assessed twice: within the 24 h after intubation and when patients were switched to pressure support mode, by measuring the endotracheal pressure in response to bilateral anterior magnetic phrenic nerve stimulation (Ptr,stim). Change in diaphragm function was expressed as the difference between Ptr,stim measured under pressure support mode and Ptr,stim measured within the 24 h after intubation. Sepsis was defined according to the Sepsis-3 international guidelines upon inclusion. In a sub-group of patients, the right hemidiaphragm thickness was measured by ultrasound. Results Ninety-two patients were enrolled in the study. Sepsis upon intubation was present in 51 (55%) patients. In septic patients, primary reason for ventilation was acute respiratory failure related to pneumonia (37/51; 73%). In non-septic patients, main reasons for ventilation were acute respiratory failure not related to pneumonia (16/41; 39%), coma (13/41; 32%) and cardiac arrest (6/41; 15%). Ptr,stim within 24 h after intubation was lower in septic patients as compared to non-septic patients: 6.3 (4.9–8.7) cmH2O vs. 9.8 (7.0–14.2) cmH2O (p = 0.004), respectively. The median (interquartile) duration of mechanical ventilation between first and second diaphragm evaluation was 4 (2–6) days in septic patients and 3 (2–4) days in non-septic patients (p = 0.073). Between first and second measurements, the change in Ptr,stim was + 19% (− 13–61) in septic patients and − 7% (− 40–12) in non-septic patients (p = 0.005). In the sub-group of patients with ultrasound measurements, end-expiratory diaphragm thickness decreased in both, septic and non-septic patients. The 28-day mortality was higher in patients with decrease or no change in diaphragm function. Conclusion Septic patients were associated with a more severe but reversible impaired diaphragm function as compared to non-septic patients. Increase in diaphragm function was associated with a better survival. Supplementary Information The online version contains supplementary material available at 10.1186/s13613-022-01005-9.
Collapse
Affiliation(s)
- Marie Lecronier
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France. .,Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM-UMR S 1158, Sorbonne Université, Paris, France.
| | - Boris Jung
- Département de Médecine Intensive - Réanimation, CHU Montpellier, Montpellier, France.,Laboratoire de Physiologie et Médecine Expérimentale du cœur et des Muscles, INSERM U1046-CNRS UMR 9214, Université de Montpellier, Montpellier, France
| | - Nicolas Molinari
- Department of Medical Information, Hôpital Arnaud de Villeneuve, IMAG U5149, Université de Montpellier, Montpellier, France
| | - Jérôme Pinot
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Thomas Similowski
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France.,Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM-UMR S 1158, Sorbonne Université, Paris, France
| | - Samir Jaber
- Département de Médecine Intensive - Réanimation, CHU Montpellier, Montpellier, France.,Laboratoire de Physiologie et Médecine Expérimentale du cœur et des Muscles, INSERM U1046-CNRS UMR 9214, Université de Montpellier, Montpellier, France
| | - Alexandre Demoule
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France.,Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM-UMR S 1158, Sorbonne Université, Paris, France
| | - Martin Dres
- Médecine Intensive - Réanimation (Département "R3S"), APHP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France.,Neurophysiologie Respiratoire Expérimentale et Clinique, INSERM-UMR S 1158, Sorbonne Université, Paris, France
| |
Collapse
|
14
|
Margotti W, Goldim MPDS, Machado RS, Bagio E, Dacoregio C, Bernades G, Lanzzarin E, Stork S, Cidreira T, Denicol TL, Joaquim L, Danielski LG, Metzker KLL, Bonfante S, Margotti E, Petronilho F. Oxidative stress in multiple organs after sepsis in elderly rats. Exp Gerontol 2022; 160:111705. [DOI: 10.1016/j.exger.2022.111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 11/04/2022]
|
15
|
Li LF, Yu CC, Wu HP, Chu CM, Huang CY, Liu PC, Liu YY. Reduction in Ventilation-Induced Diaphragmatic Mitochondrial Injury through Hypoxia-Inducible Factor 1α in a Murine Endotoxemia Model. Int J Mol Sci 2022; 23:ijms23031083. [PMID: 35163007 PMCID: PMC8835058 DOI: 10.3390/ijms23031083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022] Open
Abstract
Mechanical ventilation (MV) is essential for patients with sepsis-related respiratory failure but can cause ventilator-induced diaphragm dysfunction (VIDD), which involves diaphragmatic myofiber atrophy and contractile inactivity. Mitochondrial DNA, oxidative stress, mitochondrial dynamics, and biogenesis are associated with VIDD. Hypoxia-inducible factor 1α (HIF-1α) is crucial in the modulation of diaphragm immune responses. The mechanism through which HIF-1α and mitochondria affect sepsis-related diaphragm injury is unknown. We hypothesized that MV with or without endotoxin administration would aggravate diaphragmatic and mitochondrial injuries through HIF-1α. C57BL/6 mice, either wild-type or HIF-1α-deficient, were exposed to MV with or without endotoxemia for 8 h. MV with endotoxemia augmented VIDD and mitochondrial damage, which presented as increased oxidative loads, dynamin-related protein 1 level, mitochondrial DNA level, and the expressions of HIF-1α and light chain 3-II. Furthermore, disarrayed myofibrils; disorganized mitochondria; increased autophagosome numbers; and substantially decreased diaphragm contractility, electron transport chain activities, mitofusin 2, mitochondrial transcription factor A, peroxisome proliferator activated receptor-g coactivator-1α, and prolyl hydroxylase domain 2 were observed (p < 0.05). Endotoxin-stimulated VIDD and mitochondrial injuries were alleviated in HIF-1α-deficient mice (p < 0.05). Our data revealed that endotoxin aggravated MV-induced diaphragmatic dysfunction and mitochondrial damages, partially through the HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Li-Fu Li
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Chieh Yu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Huang-Pin Wu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chien-Ming Chu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chih-Yu Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ping-Chi Liu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Correspondence: ; Tel.: 886-2-28712121 (ext. 3071); Fax: 886-2-28757858
| |
Collapse
|
16
|
Itagaki T. Diaphragm-protective mechanical ventilation in acute respiratory failure. THE JOURNAL OF MEDICAL INVESTIGATION 2022; 69:165-172. [DOI: 10.2152/jmi.69.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
17
|
Rationale and design of a mechanistic clinical trial of JAK inhibition to prevent ventilator-induced diaphragm dysfunction. Respir Med 2021; 189:106620. [PMID: 34655959 DOI: 10.1016/j.rmed.2021.106620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/14/2021] [Accepted: 09/18/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Ventilator-induced diaphragm dysfunction (VIDD) is an important phenomenon that has been repeatedly demonstrated in experimental and clinical models of mechanical ventilation. Even a few hours of MV initiates signaling cascades that result in, first, reduced specific force, and later, atrophy of diaphragm muscle fibers. This severe, progressive weakness of the critical ventilatory muscle results in increased duration of MV and thus increased MV-associated complications/deaths. A drug that could prevent VIDD would likely have a major positive impact on intensive care unit outcomes. We identified the JAK/STAT pathway as important in VIDD and then demonstrated that JAK inhibition prevents VIDD in rats. We subsequently developed a clinical model of VIDD demonstrating reduced contractile force of isolated diaphragm fibers harvested after ∼7 vs ∼1 h of MV during a thoracic surgical procedure. MATERIALS AND METHODS The NIH-funded clinical trial that has been initiated is a prospective, placebo controlled trial: subjects undergoing esophagectomy are randomized to receive 6 preoperative doses of the FDA-approved JAK inhibitor Tofacitinib (commonly used for rheumatoid arthritis) vs. placebo. The primary outcome variable will be the difference in the reduction that occurs in force generation of diaphragm single muscle fibers (normalized to their cross-sectional area), in the Tofacitinib vs. placebo subjects, over 6 h of MV. DISCUSSION This trial represents a first-in-human, mechanistic clinical trial of a drug to prevent VIDD. It will provide proof-of-concept in human subjects whether JAK inhibition prevents clinical VIDD, and if successful, will support an ICU-based clinical trial that would determine whether JAK inhibition impacts clinical outcome variables such as duration of MV and mortality.
Collapse
|
18
|
Aarab Y, Flatres A, Garnier F, Capdevila M, Raynaud F, Lacampagne A, Chapeau D, Klouche K, Etienne P, Jaber S, Molinari N, Gamon L, Matecki S, Jung B. Shear Wave Elastography, A New Tool for Diaphragmatic Qualitative Assessment. A Translational Study. Am J Respir Crit Care Med 2021; 204:797-806. [PMID: 34255974 DOI: 10.1164/rccm.202011-4086oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Prolonged mechanical ventilation (MV) is often associated either with a decrease (known atrophy) or an increase (supposed injury) in diaphragmatic thickness. Shear wave elastography is a non-invasive technique that measures shear modulus, a surrogate of tissue stiffness and mechanical properties. OBJECTIVES To describe changes in shear modulus (SM) during the ICU stay and the relationship with alterations in muscle thickness. To perform a comprehensive ultrasound-based characterization of histological and force production changes occurring in the diaphragm. METHODS Translational study using critically ill patients and mechanically ventilated piglets. Serial ultrasound examination of the diaphragm collecting thickness and SM was performed in both patients and piglets. Transdiaphragmatic pressure and diaphragmatic biopsies were collected in piglets. MEASUREMENTS AND MAIN RESULTS We enrolled 102 patients, 88 of whom were invasively mechanically ventilated. At baseline, SM was 14.3+/-4.3 kPa and diaphragm end-expiratory thickness was 2.0+/-0.5 mm. Decrease or increase by more than 10% from baseline was reported in 86% of the patients for thickness and in 92% of the patients for shear modulus. An increase in diaphragmatic thickness during the stay was associated with a decrease in SM (β=-9.34±4.41; p=0.03) after multivariable analysis. In the piglet sample, a decrease in SM over 3 days of MV was associated with loss of force production, slow and fast fiber atrophy and increased lipid droplets accumulation. CONCLUSIONS Increases in diaphragm thickness during critical illness is associated with decreased tissue stiffness as demonstrated by shear wave ultrasound elastography, consistent with the development of muscle injury and weakness.
Collapse
Affiliation(s)
| | | | - Fanny Garnier
- Centre Hospitalier Regional Universitaire de Montpellier, 26905, Montpellier, France
| | - Mathieu Capdevila
- Montpellier University and Montpellier Teaching Hospital,, Saint Eloi Anesthesiology and Critical Care Medicine, Montpellier University and Montpellier Teaching Hospital, Montpellier, France , Montpellier, France.,Montpellier Universite d'Excellence, 539031, PhyMedExp, Montpellier, France
| | | | - Alain Lacampagne
- PhyMedExp, Montpellier University, INSERM, CNRS, Montpellier, France
| | - David Chapeau
- Lapeyronie University Hospital, Intensive Care Unit, Montpellier, France
| | - Kada Klouche
- Lapeyronie University Hospital, Intensive Care Unit, Montpellier, France
| | - Pascal Etienne
- Laboratoire Charles Coulomb, 131799, Montpellier, France
| | - Samir Jaber
- University hospital. CHU de MONTPELLIER HOPITAL SAINT ELOI, Intensive Care Unit and transplantation-Departement of Anesthesiology DAR B, Montpellier Cedex 5, France
| | - Nicolas Molinari
- CHU Montpellier - Hôpital la Colombière, DIM, Montpellier, France
| | - Lucie Gamon
- Montpellier University and Montpellier Teaching Hospital,, Saint Eloi Anesthesiology and Critical Care Medicine, Montpellier University and Montpellier Teaching Hospital, Montpellier, France
| | - Stefan Matecki
- Universite de Montpellier, 27037, 4. Pediatric Functional Exploration Unit, University Hospital of Montpellier, Montpellier, France
| | - Boris Jung
- Centre Hospitalier Regional Universitaire de Montpellier, 26905, medical ICU, Montpellier, France;
| |
Collapse
|
19
|
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: 54] [Impact Index Per Article: 13.5] [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
|
20
|
Crulli B, Kawaguchi A, Praud JP, Petrof BJ, Harrington K, Emeriaud G. Evolution of inspiratory muscle function in children during mechanical ventilation. Crit Care 2021; 25:229. [PMID: 34193216 PMCID: PMC8243304 DOI: 10.1186/s13054-021-03647-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There is no universally accepted method to assess the pressure-generating capacity of inspiratory muscles in children on mechanical ventilation (MV), and no study describing its evolution over time in this population. METHODS In this prospective observational study, we have assessed the function of the inspiratory muscles in children on various modes of MV. During brief airway occlusion maneuvers, we simultaneously recorded airway pressure depression at the endotracheal tube (ΔPaw, force generation) and electrical activity of the diaphragm (EAdi, central respiratory drive) over five consecutive inspiratory efforts. The neuro-mechanical efficiency ratio (NME, ΔPaw/EAdimax) was also computed. The evolution over time of these indices in a group of children in the pediatric intensive care unit (PICU) was primarily described. As a secondary objective, we compared these values to those measured in a group of children in the operating room (OR). RESULTS In the PICU group, although median NMEoccl decreased over time during MV (regression coefficient - 0.016, p = 0.03), maximum ΔPawmax remained unchanged (regression coefficient 0.109, p = 0.50). Median NMEoccl at the first measurement in the PICU group (after 21 h of MV) was significantly lower than at the only measurement in the OR group (1.8 cmH2O/µV, Q1-Q3 1.3-2.4 vs. 3.7 cmH2O/µV, Q1-Q3 3.5-4.2; p = 0.015). Maximum ΔPawmax in the PICU group was, however, not significantly different from the OR group (35.1 cmH2O, Q1-Q3 21-58 vs. 31.3 cmH2O, Q1-Q3 28.5-35.5; p = 0.982). CONCLUSIONS The function of inspiratory muscles can be monitored at the bedside of children on MV using brief airway occlusions. Inspiratory muscle efficiency was significantly lower in critically ill children than in children undergoing elective surgery, and it decreased over time during MV in critically ill children. This suggests that both critical illness and MV may have an impact on inspiratory muscle efficiency.
Collapse
Affiliation(s)
- Benjamin Crulli
- Pediatric Intensive Care Unit, CHU Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Atsushi Kawaguchi
- Pediatric Intensive Care Unit, CHU Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Pediatric Intensive Care Unit, Children's Hospital of Eastern Ontario, University of Ottawa, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Intensive Care Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Jean-Paul Praud
- Neonatal Respiratory Research Unit, Departments of Pediatrics and Pharmacology-Physiology, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC, J1H 5N4, Canada
| | - Basil J Petrof
- Meakins-Christie Laboratories and Translational Research in Respiratory Diseases Program, McGill University Health Centre and Research Institute, 1001 Boulevard Décarie, Montreal, QC, H4A 3J1, Canada
| | - Karen Harrington
- Pediatric Intensive Care Unit, CHU Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Guillaume Emeriaud
- Pediatric Intensive Care Unit, CHU Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
| |
Collapse
|
21
|
Fayssoil A, De Carnavalet MCDC, Mansencal N, Lofaso F, Davido B. Cardiopulmonary Pathophysiological Aspects in the Context of COVID-19 and Obesity. ACTA ACUST UNITED AC 2021; 3:1848-1857. [PMID: 34151188 PMCID: PMC8200316 DOI: 10.1007/s42399-021-00995-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Obesity is a significant public health concern associated with high morbidity. Obese patients are at risk of severe COVID-19 infection, and obesity is a high-risk factor for admission to the intensive care unit. We aimed to write a narrative review of cardiac and pulmonary pathophysiological aspects of obese patients in the context of COVID-19 infection. Obesity affects lung volume, with a decrease in expiratory reserve volume, which is associated with a decrease in lung and chest wall compliance, an increase in airway resistance, and an increase in work of breathing. Obesity affects cardiac structure and hemodynamics. Obesity is a risk factor for hypertension and cardiovascular disorders. Moreover, obesity is associated with a low-grade inflammatory state, endothelial dysfunction, hyperinsulinemia, and metabolic disorders. Obesity is associated with severe COVID-19 and invasive mechanical ventilation. These previous cardiopulmonary pathological aspects may explain the clinical severity in obese patients with COVID-19. Obese patients are at risk of severe COVID-19 infection. Understanding cardiorespiratory pathophysiological aspects may help physicians manage patients in hospitals.
Collapse
Affiliation(s)
- Abdallah Fayssoil
- Service de Cardiologie, Hôpital Raymond Poincaré, APHP, Garches, France.,Service de Cardiologie, Centre de Référence des Cardiomyopathies et des Troubles du Rythme Cardiaque Héréditaires ou Rares, Hôpital Ambroise Paré, AP-HP, Université Paris Saclay/UFR Simone Veil-Santé-Université de Versailles Saint Quentin en Yvelines, Boulogne-Billancourt, France.,INSERM U1179, END-ICAP, Montigny-le-Bretonneux, France
| | | | - Nicolas Mansencal
- Service de Cardiologie, Centre de Référence des Cardiomyopathies et des Troubles du Rythme Cardiaque Héréditaires ou Rares, Hôpital Ambroise Paré, AP-HP, Université Paris Saclay/UFR Simone Veil-Santé-Université de Versailles Saint Quentin en Yvelines, Boulogne-Billancourt, France.,INSERM U-1018, CESP, Clinical Epidemiology, UVSQ, Villejuif, France
| | - Frederic Lofaso
- Service de Physiologie - Explorations fonctionnelles, Hôpital Raymond Poincaré, APHP, Université Paris Saclay/UFR Simone Veil-Santé-Université de Versailles Saint Quentin en Yvelines, Garches, France
| | - Benjamin Davido
- Service de maladies infectieuses, Hôpital Raymond Poincaré, APHP, AP-HP, Université Paris Saclay/UFR Simone Veil-Santé-Université de Versailles Saint Quentin en Yvelines, Garches, France
| |
Collapse
|
22
|
Dong Z, Liu Y, Gai Y, Meng P, Lin H, Zhao Y, Xing J. Early rehabilitation relieves diaphragm dysfunction induced by prolonged mechanical ventilation: a randomised control study. BMC Pulm Med 2021; 21:106. [PMID: 33781259 PMCID: PMC8006630 DOI: 10.1186/s12890-021-01461-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/09/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Prolonged mechanical ventilation (MV) induces diaphragm dysfunction in patients in the intensive care units (ICUs). Our study aimed to explore the therapeutic efficacy of early rehabilitation therapy in patients with prolonged MV in the ICU. METHODS Eighty eligible patients who underwent MV for > 72 h in the ICU from June 2019 to March 2020 were enrolled in this prospective randomised controlled trial. The patients were randomly divided into a rehabilitation group (n = 39) and a control group (n = 41). Rehabilitation therapy included six levels of rehabilitation exercises. Diaphragm function was determined using ultrasound (US). RESULTS Diaphragmatic excursion (DE) and diaphragm thickening fraction (DTF) were significantly decreased in all patients in both groups after prolonged MV (p < 0.001). The rehabilitation group had significantly higher DTF (p = 0.008) and a smaller decrease in DTF (p = 0.026) than the control group after 3 days of rehabilitation training. The ventilator duration and intubation duration were significantly shorter in the rehabilitation group than in the control group (p = 0.045 and p = 0.037, respectively). There were no significant differences in the duration of ICU stay, proportion of patients undergoing tracheotomy, and proportion of recovered patients between the two groups. CONCLUSIONS Early rehabilitation is feasible and beneficial to ameliorate diaphragm dysfunction induced by prolonged MV and advance withdrawal from the ventilator and extubation in patients with MV. Diaphragm US is suggested for mechanically ventilated patients in the ICU. Trial registration Chinese Clinical Trial Registry, ID: ChiCTR1900024046, registered on 2019/06/23.
Collapse
Affiliation(s)
- Zehua Dong
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, 266000 Shandong China
| | - Ying Liu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, 266000 Shandong China
| | - Yubiao Gai
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, 266000 Shandong China
| | - Pingping Meng
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000 Shandong China
| | - Hui Lin
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, 266000 Shandong China
| | - Yuxiao Zhao
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, 266000 Shandong China
| | - Jinyan Xing
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Qingdao, 266000 Shandong China
| |
Collapse
|
23
|
Frija-Masson J, Bancal C, Plantier L, Benzaquen H, Mangin L, Penaud D, Arnoult F, Flamant M, d’Ortho MP. Alteration of Diffusion Capacity After SARS-CoV-2 Infection: A Pathophysiological Approach. Front Physiol 2021; 12:624062. [PMID: 33841169 PMCID: PMC8030254 DOI: 10.3389/fphys.2021.624062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/27/2021] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has affected millions of people worldwide, and pneumonia affects 90% of patients. This raises the possibility of millions of people with altered lung function. Few data exist to date on pulmonary function after SARS-CoV-2 infection, but alteration of diffusion capacity of CO (D LCO) is the most frequently described abnormality. First, we present original data on lung function at 3 months after SARS-CoV-2 infection and discuss the effect of using European Coal and Steel Community (ECSC) or Global Lung Function Initiative (GLI) reference equations to diagnose diffusion capacity. Second, we review existing data on D LCO alteration after SARS-CoV-2 infection and discuss the implication of restrictive disorder in D LCO alteration. Last, we discuss the pathophysiology of D LCO alteration and try to disentangle vascular damage and fibrosis.
Collapse
Affiliation(s)
- Justine Frija-Masson
- Neurodiderot, INSERM, Université de Paris, Paris, France
- Physiologie-Explorations Fonctionnelles, FHU APOLLO, Assistance Publique Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France
| | - Catherine Bancal
- Physiologie-Explorations Fonctionnelles, FHU APOLLO, Assistance Publique Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France
| | - Laurent Plantier
- Centre de Ressources et de Compétences de la Mucoviscidose, Service de Pneumologie et Explorations Fonctionnelles Respiratoires, CHRU de Tours, Tours, France
- CEPR/INSERM, UMR 1100, Université de Tours, Tours, France
| | - Hélène Benzaquen
- Physiologie-Explorations Fonctionnelles, FHU APOLLO, Assistance Publique Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France
| | - Laurence Mangin
- Physiologie-Explorations Fonctionnelles, FHU APOLLO, Assistance Publique Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France
- Laboratoire Matière et Systèmes Complexes, UMR 7505, CNRS, Paris, France
| | - Dominique Penaud
- Physiologie-Explorations Fonctionnelles, FHU APOLLO, Assistance Publique Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France
| | - Florence Arnoult
- Physiologie-Explorations Fonctionnelles, FHU APOLLO, Assistance Publique Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France
| | - Martin Flamant
- Physiologie-Explorations Fonctionnelles, FHU APOLLO, Assistance Publique Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France
- INSERM U 1149, Center for Research in Inflammation, Université de Paris, Paris, France
| | - Marie-Pia d’Ortho
- Neurodiderot, INSERM, Université de Paris, Paris, France
- Physiologie-Explorations Fonctionnelles, FHU APOLLO, Assistance Publique Hôpitaux de Paris, Hôpital Bichat-Claude Bernard, Paris, France
| |
Collapse
|
24
|
Okazaki T, Suzukamo Y, Miyatake M, Komatsu R, Yaekashiwa M, Nihei M, Izumi S, Ebihara T. Respiratory Muscle Weakness as a Risk Factor for Pneumonia in Older People. Gerontology 2021; 67:581-590. [PMID: 33621975 DOI: 10.1159/000514007] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/21/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The respiratory muscle strength regulates the effectiveness of coughing, which clears the airways and protects people from pneumonia. Sarcopenia is an aging-related loss of muscle mass and function, the worsening of which is associated with malnutrition. The loss of respiratory and swallowing muscle strength occurs with aging, but its effect on pneumonia is unclear. This study aimed to determine the risks of respiratory muscle weakness on the onset and relapse of pneumonia in older people in conjunction with other muscle-related factors such as malnutrition. METHODS We conducted a longitudinal study with 47 pneumonia inpatients and 35 non-pneumonia controls aged 70 years and older. We evaluated the strength of respiratory and swallowing muscles, muscle mass, and malnutrition (assessed by serum albumin levels and somatic fat) during admission and confirmed pneumonia relapse within 6 months. The maximal inspiratory and expiratory pressures determined the respiratory muscle strength. Swallowing muscle strength was evaluated by tongue pressure. Bioelectrical impedance analysis was used to evaluate the muscle and fat mass. RESULTS The respiratory muscle strength, body trunk muscle mass, serum albumin level, somatic fat mass, and tongue pressure were significantly lower in pneumonia patients than in controls. Risk factors for the onset of pneumonia were low inspiratory respiratory muscle strength (odds ratio [OR], 6.85; 95% confidence interval [CI], 1.56-30.11), low body trunk muscle mass divided by height2 (OR, 6.86; 95% CI, 1.49-31.65), and low serum albumin level (OR, 5.46; 95% CI, 1.51-19.79). For the relapse of pneumonia, low somatic fat mass divided by height2 was a risk factor (OR, 20.10; 95% CI, 2.10-192.42). DISCUSSION/CONCLUSIONS Respiratory muscle weakness, lower body trunk muscle mass, and malnutrition were risk factors for the onset of pneumonia in older people. For the relapse of pneumonia, malnutrition was a risk factor.
Collapse
Affiliation(s)
- Tatsuma Okazaki
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Japan, .,Center for Dysphagia of Tohoku University Hospital, Sendai, Japan,
| | - Yoshimi Suzukamo
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Midori Miyatake
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Riyo Komatsu
- Department of Respiratory Medicine, Hiraka General Hospital, Yokote, Akita, Japan
| | | | - Mayumi Nihei
- Department of Respiratory Medicine, Sendai City Hospital, Sendai, Japan
| | - Shinichi Izumi
- Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Medicine, Sendai, Japan.,Center for Dysphagia of Tohoku University Hospital, Sendai, Japan.,Department of Physical Medicine and Rehabilitation, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Takae Ebihara
- Department of Geriatric Medicine, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| |
Collapse
|
25
|
Li LF, Yu CC, Huang HY, Wu HP, Chu CM, Huang CY, Liu PC, Liu YY. Suppression of Hypoxia-Inducible Factor 1α by Low-Molecular-Weight Heparin Mitigates Ventilation-Induced Diaphragm Dysfunction in a Murine Endotoxemia Model. Int J Mol Sci 2021; 22:ijms22041702. [PMID: 33567713 PMCID: PMC7914863 DOI: 10.3390/ijms22041702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/29/2022] Open
Abstract
Mechanical ventilation (MV) is required to maintain life for patients with sepsis-related acute lung injury but can cause diaphragmatic myotrauma with muscle damage and weakness, known as ventilator-induced diaphragm dysfunction (VIDD). Hypoxia-inducible factor 1α (HIF-1α) plays a crucial role in inducing inflammation and apoptosis. Low-molecular-weight heparin (LMWH) was proven to have anti-inflammatory properties. However, HIF-1α and LMWH affect sepsis-related diaphragm injury has not been investigated. We hypothesized that LMWH would reduce endotoxin-augmented VIDD through HIF-1α. C57BL/6 mice, either wild-type or HIF-1α–deficient, were exposed to MV with or without endotoxemia for 8 h. Enoxaparin (4 mg/kg) was administered subcutaneously 30 min before MV. MV with endotoxemia aggravated VIDD, as demonstrated by increased interleukin-6 and macrophage inflammatory protein-2 levels, oxidative loads, and the expression of HIF-1α, calpain, caspase-3, atrogin-1, muscle ring finger-1, and microtubule-associated protein light chain 3-II. Disorganized myofibrils, disrupted mitochondria, increased numbers of autophagic and apoptotic mediators, substantial apoptosis of diaphragm muscle fibers, and decreased diaphragm function were also observed (p < 0.05). Endotoxin-exacerbated VIDD and myonuclear apoptosis were attenuated by pharmacologic inhibition by LMWH and in HIF-1α–deficient mice (p < 0.05). Our data indicate that enoxaparin reduces endotoxin-augmented MV-induced diaphragmatic injury, partially through HIF-1α pathway inhibition.
Collapse
Affiliation(s)
- Li-Fu Li
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Chieh Yu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Hung-Yu Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Huang-Pin Wu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chien-Ming Chu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chih-Yu Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ping-Chi Liu
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (L.-F.L.); (C.-C.Y.); (H.-Y.H.); (H.-P.W.); (C.-M.C.); (C.-Y.H.); (P.-C.L.)
- Department of Internal Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Correspondence: ; Tel.: +(886)-2-28712121 (ext. 3071); Fax: +(886)-2-28757858
| |
Collapse
|
26
|
Goligher EC, Dres M, Patel BK, Sahetya SK, Beitler JR, Telias I, Yoshida T, Vaporidi K, Grieco DL, Schepens T, Grasselli G, Spadaro S, Dianti J, Amato M, Bellani G, Demoule A, Fan E, Ferguson ND, Georgopoulos D, Guérin C, Khemani RG, Laghi F, Mercat A, Mojoli F, Ottenheijm CAC, Jaber S, Heunks L, Mancebo J, Mauri T, Pesenti A, Brochard L. Lung- and Diaphragm-Protective Ventilation. Am J Respir Crit Care Med 2020; 202:950-961. [PMID: 32516052 DOI: 10.1164/rccm.202003-0655cp] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mechanical ventilation can cause acute diaphragm atrophy and injury, and this is associated with poor clinical outcomes. Although the importance and impact of lung-protective ventilation is widely appreciated and well established, the concept of diaphragm-protective ventilation has recently emerged as a potential complementary therapeutic strategy. This Perspective, developed from discussions at a meeting of international experts convened by PLUG (the Pleural Pressure Working Group) of the European Society of Intensive Care Medicine, outlines a conceptual framework for an integrated lung- and diaphragm-protective approach to mechanical ventilation on the basis of growing evidence about mechanisms of injury. We propose targets for diaphragm protection based on respiratory effort and patient-ventilator synchrony. The potential for conflict between diaphragm protection and lung protection under certain conditions is discussed; we emphasize that when conflicts arise, lung protection must be prioritized over diaphragm protection. Monitoring respiratory effort is essential to concomitantly protect both the diaphragm and the lung during mechanical ventilation. To implement lung- and diaphragm-protective ventilation, new approaches to monitoring, to setting the ventilator, and to titrating sedation will be required. Adjunctive interventions, including extracorporeal life support techniques, phrenic nerve stimulation, and clinical decision-support systems, may also play an important role in selected patients in the future. Evaluating the clinical impact of this new paradigm will be challenging, owing to the complexity of the intervention. The concept of lung- and diaphragm-protective ventilation presents a new opportunity to potentially improve clinical outcomes for critically ill patients.
Collapse
Affiliation(s)
- Ewan C Goligher
- Interdepartmental Division of Critical Care Medicine.,Division of Respirology, Department of Medicine, University Health Network, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Martin Dres
- Service de Pneumologie, Médecine Intensive et Réanimation (Département R3S), Assistance Publique-Hopitaux de Paris, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Paris, France.,Unite Mixte de Recherche-Sorbonne 1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Institut National de la Sante et de la Recherche Medicale, Sorbonne Université, Paris, France
| | - Bhakti K Patel
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Sarina K Sahetya
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Jeremy R Beitler
- Division of Pulmonary, Allergy, and Critical Care Medicine, Center for Acute Respiratory Failure, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Irene Telias
- Interdepartmental Division of Critical Care Medicine.,Division of Respirology, Department of Medicine, University Health Network, Toronto, Ontario, Canada.,Keenan Centre for Biomedical Research, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Takeshi Yoshida
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Katerina Vaporidi
- Department of Intensive Care Medicine, University Hospital of Heraklion, Medical School, University of Crete, Heraklion, Greece
| | - Domenico Luca Grieco
- Department of Anesthesiology and Intensive Care Medicine, Catholic University of the Sacred Heart, Rome, Italy.,Dipartimento di Medicina d'Urgenza e di Terapia Intensiva e Anestesia, Fondazione Policlinico Universitario, A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Tom Schepens
- Department of Critical Care Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Giacomo Grasselli
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Savino Spadaro
- Department Morphology, Surgery and Experimental Medicine, ICU, St. Anne's Archbishop Hospital, University of Ferrara, Ferrara, Italy
| | - Jose Dianti
- Interdepartmental Division of Critical Care Medicine.,Division of Respirology, Department of Medicine, University Health Network, Toronto, Ontario, Canada.,Intensive Care Unit, Department of Medicine, Italian Hospital of Buenos Aires, Buenos Aires, Argentina
| | - Marcelo Amato
- Laboratório de Pneumologia, Laboratório de Investicação Médica 9, Disciplina de Pneumologia, Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Giacomo Bellani
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
| | - Alexandre Demoule
- Service de Pneumologie, Médecine Intensive et Réanimation (Département R3S), Assistance Publique-Hopitaux de Paris, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Paris, France.,Unite Mixte de Recherche-Sorbonne 1158 Neurophysiologie Respiratoire Expérimentale et Clinique, Institut National de la Sante et de la Recherche Medicale, Sorbonne Université, Paris, France
| | - Eddy Fan
- Interdepartmental Division of Critical Care Medicine.,Institute for Health Policy, Management, and Evaluation, and.,Division of Respirology, Department of Medicine, University Health Network, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Niall D Ferguson
- Interdepartmental Division of Critical Care Medicine.,Institute for Health Policy, Management, and Evaluation, and.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Division of Respirology, Department of Medicine, University Health Network, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Dimitrios Georgopoulos
- Department of Intensive Care Medicine, University Hospital of Heraklion, Medical School, University of Crete, Heraklion, Greece
| | - Claude Guérin
- Médecine Intensive-Réanimation, Hopital Edouard Herriot Lyon, Faculté de Médecine Lyon-Est, Université de Lyon, Institut National de la Santé et de la Recherche Médicale 955 Créteil, Lyon, France
| | - Robinder G Khemani
- Department of Anesthesiology and Critical Care, Children's Hospital Los Angeles, Los Angeles, California.,Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Franco Laghi
- Division of Pulmonary and Critical Care Medicine, Stritch School of Medicine, Loyola University, Maywood, Illinois.,Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital, Hines, Illinois
| | - Alain Mercat
- Département de Médecine Intensive-Réanimation et Médecine Hyperbare, Centre Hospitalier d'Angers, Angers, France
| | - Francesco Mojoli
- Department of Anesthesia and Intensive Care, Scientific Hospitalization and Care Institute, San Matteo Polyclinic Foundation, University of Pavia, Pavia, Italy
| | | | - Samir Jaber
- Anesthesiology and Intensive Care, Anesthesia and Critical Care Department B, Saint Eloi Teaching Hospital, PhyMedExp, Montpellier University Hospital Center, University of Montpellier, Joint Research Unit 9214, National Institute of Health and Medical Research U1046, National Scientific Research Center, Montpellier, France; and
| | - Leo Heunks
- Department of Intensive Care, Vrije University Location, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Jordi Mancebo
- Servei de Medicina Intensiva Hospital de Sant Pau, Barcelona, Spain
| | - Tommaso Mauri
- Dipartimento di Medicina d'Urgenza e di Terapia Intensiva e Anestesia, Fondazione Policlinico Universitario, A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Department of Critical Care Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Antonio Pesenti
- Dipartimento di Medicina d'Urgenza e di Terapia Intensiva e Anestesia, Fondazione Policlinico Universitario, A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy.,Department of Critical Care Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Laurent Brochard
- Interdepartmental Division of Critical Care Medicine.,Keenan Centre for Biomedical Research, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
27
|
van Steveninck AL, Imming LM. Diaphragm dysfunction prior to intubation in a patient with Covid-19 pneumonia; assessment by point of care ultrasound and potential implications for patient monitoring. Respir Med Case Rep 2020; 31:101284. [PMID: 33200066 PMCID: PMC7654235 DOI: 10.1016/j.rmcr.2020.101284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/09/2020] [Accepted: 11/02/2020] [Indexed: 11/29/2022] Open
Abstract
The clinical research described in this case report was initiated because of the recognized need for early identification of Covid-19 patients at risk of respiratory failure. We used point of care ultrasound to identify diaphragm dysfunction in a spontaneously breathing Covid-19 patient. Measurements of diaphragm thickness and thickening fraction indicated diaphragm dysfunction prior to intubation while respiratory failure was not yet evident from arterial blood gas analysis. Recovery of diaphragm contractility was demonstrated within two days of controlled mechanical ventilation when the patient was switched to a pressure support mode. With recovery of the diaphragm very large fractional shortening was seen after discontinuation of rocuronium, which was associated with a reduced dynamic compliance. In conclusion, this case report illustrates the need to be aware of potential diaphragm dysfunction in spontaneously breathing Covid-19 patients. With recovery, point of care ultrasound allows repeated evaluation of diaphragm function which appears to be responsive to changes in pulmonary compliance.
Collapse
Affiliation(s)
| | - Leonie M Imming
- Department of Pulmonary Medicine, Deventer Hospital, Deventer, the Netherlands
| |
Collapse
|
28
|
Hong Y, Woo S, Kim Y, Lee JJ, Hong JY. Plasma concentrations of NOX4 are predictive of successful liberation from mechanical ventilation and 28-day mortality in intubated patients. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1376. [PMID: 33313121 PMCID: PMC7723573 DOI: 10.21037/atm-20-4252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) enzymes play important roles in generating reactive oxygen species (ROS); in particular, NOX4 plays a distinct role in regulating lung inflammation and apoptosis. Methods We determined whether plasma NOX4 level can be used as a prognostic biomarker to guide weaning from mechanical ventilation and to predict mortality in intubated patients. Plasma levels of NOX4 were measured at days 1 (NOX4 D1) and 7 (NOX4 D7) after initiation of mechanical ventilation in 184 patients. Results With increase in day 7 NOX4 quartile, the success of weaning tended to decrease and 28-day mortality tended to increase. On multivariate logistic regression, Acute Physiology, Age, Chronic Health Evaluation II (APACHE II) [odds ratio (OR): 1.10; 95% CI, 1.02–1.18], duration of mechanical ventilation (OR: 1.12; 95% CI: 1.06–1.18), and NOX4 D7 levels >18.2 ng/mL (OR: 4.40; 95% CI: 1.91–10.06) were independently associated with weaning failure. Also, Cox-hazard proportional model showed that NOX4 D7 level >18.2 ng/mL (hazard ratio [HR], 2.29; 95% CI, 1.26–4.16), APACHE II (HR: 1.07; 95% CI: 1.02–1.14), Sequential Organ Failure Assessment (SOFA) (HR: 1.10; 95% CI: 1.01–1.20) and coexisting cancer (HR: 1.99; 95% CI, 1.01–3.94), were independently associated with 28-day mortality. The longitudinal trend of NOX4 level varied according to the clinical outcomes. Conclusions An increased plasma NOX4 D7 level was associated with weaning failure and 28-day mortality in patients with mechanical ventilation. Our results suggest that NOX4-directed management may lead to improved outcomes in patients with mechanical ventilation.
Collapse
Affiliation(s)
- Yoonki Hong
- Department of Internal Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Seongji Woo
- Institute of New frontier Research, Hallym University College of Medicine, Republic of Korea
| | - Youngmi Kim
- Institute of New frontier Research, Hallym University College of Medicine, Republic of Korea
| | - Jae Jun Lee
- Institute of New frontier Research, Hallym University College of Medicine, Republic of Korea
| | - Ji Young Hong
- Institute of New frontier Research, Hallym University College of Medicine, Republic of Korea.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Chuncheon Sacred Heart Hospital, Hallym University Medical Center, Chuncheon, Gangwon-do, Republic of Korea.,Lung Research Institute of Hallym University College of Medicine, Chuncheon, Republic of Korea
| |
Collapse
|
29
|
Respiratory Neurophysiology in Intensive Care Unit. J Clin Neurophysiol 2020; 37:208-210. [PMID: 32358247 DOI: 10.1097/wnp.0000000000000663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Patients with intensive care unit-acquired weakness have an increased risk of prolonged mechanical ventilation, which is a risk factor for prolonged stay and mortality. The most common cause of this problem is weakness of the diaphragm, which can derive from phrenic nerve injury associated with critical neuropathy, or with the complex multiorgan failure/systemic respiratory response syndrome causing muscle fiber lesion. Two conventional neurophysiological techniques are useful to investigate the respiratory muscles, phrenic nerve conduction, and needle electromyography of the accessory respiratory muscles and diaphragm. Phrenic nerve stimulation is a standard noninvasive technique; amplitude of the motor response can be reduced because of muscle fiber inexcitability or axonal loss. Electromyography of the diaphragm is an invasive method but is safe if performed as indicated. It can reveal neurogenic or myopathic motor units. Although these neurophysiological methods have limitations in the investigation of intensive care unit patients with severe respiratory involvement, normal phrenic nerve responses should exclude marked axonal loss and indicate a better prognosis.
Collapse
|
30
|
Neuregulin-1 β Protects the Rat Diaphragm during Sepsis against Oxidative Stress and Inflammation by Activating the PI3K/Akt Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1720961. [PMID: 32765805 PMCID: PMC7387979 DOI: 10.1155/2020/1720961] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/15/2020] [Accepted: 07/01/2020] [Indexed: 11/18/2022]
Abstract
Sepsis-induced diaphragm dysfunction (SIDD) which is mainly characterized by decrease in diaphragmatic contractility has been identified to cause great harms to patients. Therefore, there is an important and pressing need to find effective treatments for improving SIDD. In addition, acetylcholinesterase (AChE) activity is a vital property of the diaphragm, so we evaluated both diaphragmatic contractility and AChE activity. Though neuregulin-1β (NRG-1β) is known to exert organ-protective effects in some inflammatory diseases, little is known about the potential of NRG-1β therapy in the diaphragm during sepsis. Our study was aimed at exploring the effects of NRG-1β application on diaphragmatic contractility and AChE activity during sepsis. Proinflammatory cytokines, muscle injury biomarkers in serum, contractile force, AChE activity, proinflammatory cytokines, oxidative parameters, histological condition, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining, and expression of phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB/Akt) signaling proteins in the diaphragm were measured and compared between nonseptic and septic groups with or without NRG-1β treatment. In vitro, the effects of NRG-1β on reactive oxygen species (ROS) production in the lipopolysaccharide- (LPS-) stimulated L6 rat muscle skeletal cells with or without the Akt inhibitor MK-2206 were detected. NRG-1β inhibited proinflammatory cytokine release and muscle injury biomarkers soaring in serum and improved the sepsis-induced diaphragm dysfunction and AChE activity decrease significantly during sepsis. Meanwhile, the inflammatory response, oxidative stress, pathological impairment, and cell apoptosis in the diaphragm were mitigated by NRG-1β. And NRG-1β activated the PI3K/Akt signaling in the diaphragm of septic rats. Elevated ROS production in the LPS-stimulated L6 rat skeletal muscle cells was reduced after treatment with NRG-1β, while MK-2206 blocked these effects of NRG-1β. In conclusion, our findings underlined that NRG-1β could reduce circulating levels of proinflammatory cytokines in rats with sepsis, adjust diaphragmatic proinflammatory cytokine level, mitigate diaphragmatic oxidative injury, and lessen diaphragm cell apoptosis, thereby improving diaphragmatic function, and play a role in diaphragmatic protection by activating PI3K/Akt signaling.
Collapse
|
31
|
Is Mitochondrial Oxidative Stress the Key Contributor to Diaphragm Atrophy and Dysfunction in Critically Ill Patients? Crit Care Res Pract 2020; 2020:8672939. [PMID: 32377432 PMCID: PMC7191397 DOI: 10.1155/2020/8672939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 02/08/2023] Open
Abstract
Diaphragm dysfunction is prevalent in the progress of respiratory dysfunction in various critical illnesses. Respiratory muscle weakness may result in insufficient ventilation, coughing reflection suppression, pulmonary infection, and difficulty in weaning off respirators. All of these further induce respiratory dysfunction and even threaten the patients' survival. The potential mechanisms of diaphragm atrophy and dysfunction include impairment of myofiber protein anabolism, enhancement of myofiber protein degradation, release of inflammatory mediators, imbalance of metabolic hormones, myonuclear apoptosis, autophagy, and oxidative stress. Among these contributors, mitochondrial oxidative stress is strongly implicated to play a key role in the process as it modulates diaphragm protein synthesis and degradation, induces protein oxidation and functional alteration, enhances apoptosis and autophagy, reduces mitochondrial energy supply, and is regulated by inflammatory cytokines via related signaling molecules. This review aims to provide a concise overview of pathological mechanisms of diaphragmatic dysfunction in critically ill patients, with special emphasis on the role and modulating mechanisms of mitochondrial oxidative stress.
Collapse
|
32
|
Nouet J, Himelman E, Lahey KC, Zhao Q, Fraidenraich D. Connexin-43 reduction prevents muscle defects in a mouse model of manifesting Duchenne muscular dystrophy female carriers. Sci Rep 2020; 10:5683. [PMID: 32231219 PMCID: PMC7105483 DOI: 10.1038/s41598-020-62844-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/18/2020] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe X-linked neuromuscular disorder that affects males. However, 8% of female carriers are symptomatic and underrepresented in research due to the lack of animal models. We generated a symptomatic mouse model of DMD carriers via injection of mdx (murine DMD) embryonic stem cells (ESCs) into wild-type (WT) blastocysts (mdx/WT chimera). mdx/WT chimeras developed cardiomyopathic features and dystrophic skeletal muscle phenotypes including elevated mononuclear invasion, central nucleation, fibrosis and declined forelimb grip strength. The disease was accompanied by connexin-43 (Cx43) aberrantly enhanced in both cardiac and skeletal muscles and remodeled in the heart. Genetic reduction of Cx43-copy number in mdx/WT-Cx43(+/-) chimeras protected them from both cardiac and skeletal muscle fiber damage. In dystrophic skeletal muscle, Cx43 expression was not seen in the fibers but in adjacent F4/80+ mononuclear cells. Ethidium Bromide uptake in purified F4/80+/CD11b+ mdx macrophages revealed functional activity of Cx43, which was inhibited by administration of Gap19 peptide mimetic, a Cx43 hemichannel-specific inhibitor. Thus, we suggest that Cx43 reduction in symptomatic DMD carrier mice leads to prevention of Cx43 remodeling in the heart and prevention of aberrant Cx43 hemichannel activity in the skeletal muscle macrophages neighboring Cx43 non-expressing fibers.
Collapse
Affiliation(s)
- Julie Nouet
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, USA
| | - Eric Himelman
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, USA
| | - Kevin C Lahey
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, USA
| | - Qingshi Zhao
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, USA
| | - Diego Fraidenraich
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
33
|
Supinski GS, Wang L, Schroder EA, Callahan LAP. SS31, a mitochondrially targeted antioxidant, prevents sepsis-induced reductions in diaphragm strength and endurance. J Appl Physiol (1985) 2020; 128:463-472. [PMID: 31944887 PMCID: PMC7099438 DOI: 10.1152/japplphysiol.00240.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 01/14/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Sepsis-induced diaphragm dysfunction contributes to respiratory failure and mortality in critical illness. There are no treatments for this form of diaphragm weakness. Studies show that sepsis-induced muscle dysfunction is triggered by enhanced mitochondrial free radical generation. We tested the hypothesis that SS31, a mitochondrially targeted antioxidant, would attenuate sepsis-induced diaphragm dysfunction. Four groups of mice were studied: 1) sham-operated controls, 2) sham-operated+SS31 (10 mg·kg-1·day-1), 3) cecal ligation puncture (CLP), and 4) CLP+SS31. Forty-eight hours postoperatively, diaphragm strips with attached phrenic nerves were isolated, and the following were assessed: muscle-field-stimulated force-frequency curves, nerve-stimulated force-frequency curves, and muscle fatigue. We also measured calpain activity, 20S proteasomal activity, myosin heavy chain (MHC) levels, mitochondrial function, and aconitase activity, an index of mitochondrial superoxide generation. Sepsis markedly reduced diaphragm force generation; SS31 prevented these decrements. Diaphragm-specific force generation averaged 30.2 ± 1.4, 9.4 ± 1.8, 25.5 ± 2.3, and 27.9 ± 0.6 N/cm2 for sham, CLP, sham+SS31, and CLP+SS31 groups (P < 0.001). Similarly, with phrenic nerve stimulation, CLP depressed diaphragm force generation, effects prevented by SS31. During endurance trials, force was significantly reduced with CLP, and SS31 prevented these reductions (P < 0.001). Sepsis also increased diaphragm calpain activity, increased 20S proteasomal activity, decreased MHC levels, reduced mitochondrial function (state 3 rates and ATP generation), and reduced aconitase activity; SS31 prevented each of these sepsis-induced alterations (P ≤ 0.017 for all indices). SS31 prevents sepsis-induced diaphragm dysfunction, preserving force generation, endurance, and mitochondrial function. Compounds with similar mechanisms of action may be useful therapeutically to preserve diaphragm function in patients who are septic and critically ill.NEW & NOTEWORTHY Sepsis-induced diaphragm dysfunction is a major contributor to mortality and morbidity in patients with critical illness in intensive care units. Currently, there is no proven pharmacological treatment for this problem. This study provides the novel finding that administration of SS31, a mitochondrially targeted antioxidant, preserves diaphragm myosin heavy chain content and mitochondrial function, thereby preventing diaphragm weakness and fatigue in sepsis.
Collapse
Affiliation(s)
- Gerald S Supinski
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Lin Wang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Elizabeth A Schroder
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| | - Leigh Ann P Callahan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
34
|
Utility of Plasma GDF-15 for Diagnosis and Prognosis Assessment of ICU-Acquired Weakness in Mechanically Ventilated Patients: Prospective Observational Study. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3630568. [PMID: 32104689 PMCID: PMC7036092 DOI: 10.1155/2020/3630568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/24/2019] [Accepted: 09/03/2019] [Indexed: 12/25/2022]
Abstract
Objective To identify the clinical correlations between plasma growth differentiation factor-15 (GDF-15), skeletal muscle function, and acute muscle wasting in ICU patients with mechanical ventilation. In addition, to investigate its diagnostic value for ICU-acquired weakness (ICU-AW) and its predictive value for 90-day survival in mechanically ventilated patients. Methods 95 patients with acute respiratory failure, who required mechanical ventilation therapy, were randomly selected among hospitalized patients from June 2017 to January 2019. The plasma GDF-15 level was detected by ELISA, the rectus femoris cross-sectional area (RFcsa) was measured by ultrasound, and the patient's muscle strength was assessed using the British Medical Research Council (MRC) muscle strength score on day 1, day 4, and day 7. Patients were divided into an ICU-AW group and a non-ICU-AW group according to their MRC-score on the 7th day. The differences in plasma GDF-15 level, MRC-score, and RFcsa between the two groups were compared on the 1st, 4th, and 7th day after being admitted to the ICU. Then, the correlations between plasma GDF-15 level, RFcsa loss, and MRC-score on day 7 were investigated. The receiver operating characteristic curve (ROC) was used to analyze the plasma GDF-15 level, RFcsa loss, and % decrease in RFcsa on the 7th day to the diagnosis of ICU-AW in mechanically ventilated patients. Moreover, the predictive value of GDF-15 on the 90-day survival status of patients was assessed using patient survival curves. Results Based on whether the 7th day MRC-score was <48, 50 cases were included in the ICU-AW group and 45 cases in the non-ICU-AW group. The length of mechanical ventilation, ICU length of stay, and hospital length of stay were significantly longer in the ICU-AW group than in the non-ICU-AW group (all P < 0.05), while the other baseline indicators were not statistically significant between the two groups. As the treatment time increased, the plasma GDF-15 level was significantly increased, the ICU-AW group demonstrated a significant decreasing trend in the MRC-score and RFcsa, while no significant changes were found in the non-ICU-AW group. In the ICU-AW group, the plasma GDF-15 level was significantly higher than that in the non-ICU-AW group, while the RFcsa and the MRC-score were significantly lower than those in the non-ICU-AW group (GDF-15 (pg/ml): 2542.44 ± 629.38 vs. 1542.86 ± 502.86; RFcsa (cm2): 2.04 ± 0.64 vs. 2.34 ± 0.61; MRC-score: 41.22 ± 3.42 vs. 51.42 ± 2.72, all P < 0.05), while the other baseline indicators were not statistically significant between the two groups. As the treatment time increased, the plasma GDF-15 level was significantly increased, the ICU-AW group demonstrated a significant decreasing trend in the MRC-score and RFcsa, while no significant changes were found in the non-ICU-AW group. In the ICU-AW group, the plasma GDF-15 level was significantly higher than that in the non-ICU-AW group, while the RFcsa and the MRC-score were significantly lower than those in the non-ICU-AW group (GDF-15 (pg/ml): 2542.44 ± 629.38 vs. 1542.86 ± 502.86; RFcsa (cm2): 2.04 ± 0.64 vs. 2.34 ± 0.61; MRC-score: 41.22 ± 3.42 vs. 51.42 ± 2.72, all r = −0.60), while it was significantly positively correlated with the RFcsa loss (r = −0.60), while it was significantly positively correlated with the RFcsa loss (r = −0.60), while it was significantly positively correlated with the RFcsa loss (r = −0.60), while it was significantly positively correlated with the RFcsa loss (P < 0.05), while the other baseline indicators were not statistically significant between the two groups. As the treatment time increased, the plasma GDF-15 level was significantly increased, the ICU-AW group demonstrated a significant decreasing trend in the MRC-score and RFcsa, while no significant changes were found in the non-ICU-AW group. In the ICU-AW group, the plasma GDF-15 level was significantly higher than that in the non-ICU-AW group, while the RFcsa and the MRC-score were significantly lower than those in the non-ICU-AW group (GDF-15 (pg/ml): 2542.44 ± 629.38 vs. 1542.86 ± 502.86; RFcsa (cm2): 2.04 ± 0.64 vs. 2.34 ± 0.61; MRC-score: 41.22 ± 3.42 vs. 51.42 ± 2.72, all P < 0.05), while the other baseline indicators were not statistically significant between the two groups. As the treatment time increased, the plasma GDF-15 level was significantly increased, the ICU-AW group demonstrated a significant decreasing trend in the MRC-score and RFcsa, while no significant changes were found in the non-ICU-AW group. In the ICU-AW group, the plasma GDF-15 level was significantly higher than that in the non-ICU-AW group, while the RFcsa and the MRC-score were significantly lower than those in the non-ICU-AW group (GDF-15 (pg/ml): 2542.44 ± 629.38 vs. 1542.86 ± 502.86; RFcsa (cm2): 2.04 ± 0.64 vs. 2.34 ± 0.61; MRC-score: 41.22 ± 3.42 vs. 51.42 ± 2.72, all P < 0.05), while the other baseline indicators were not statistically significant between the two groups. As the treatment time increased, the plasma GDF-15 level was significantly increased, the ICU-AW group demonstrated a significant decreasing trend in the MRC-score and RFcsa, while no significant changes were found in the non-ICU-AW group. In the ICU-AW group, the plasma GDF-15 level was significantly higher than that in the non-ICU-AW group, while the RFcsa and the MRC-score were significantly lower than those in the non-ICU-AW group (GDF-15 (pg/ml): 2542.44 ± 629.38 vs. 1542.86 ± 502.86; RFcsa (cm2): 2.04 ± 0.64 vs. 2.34 ± 0.61; MRC-score: 41.22 ± 3.42 vs. 51.42 ± 2.72, all Conclusion The plasma GDF-15 concentration level was significantly associated with skeletal muscle function and muscle wasting on day 7 in ICU patients with mechanical ventilation. Therefore, it can be concluded that the plasma GDF-15 level on the 7th day has a high diagnostic yield for ICU-acquired muscle weakness, and it can predict the 90-day survival status of ICU mechanically ventilated patients.
Collapse
|
35
|
Abstract
WHAT WE ALREADY KNOW ABOUT THIS TOPIC Muscle atrophy is common in the critically ill, and diaphragm atrophy occurs during mechanical ventilation. It is not known whether wasting of diaphragm and nondiaphragm muscle is related. WHAT THIS ARTICLE TELLS US THAT IS NEW Ultrasound was used for serial assessment of diaphragm and pectoral muscle in 97 critically ill patients. Diaphragm and pectoral atrophy occurred in 48% and 29%, respectively, and was associated with septic shock (diaphragm) and steroid use (pectoral); atrophy of the two muscle types appears unrelated. BACKGROUND Muscle atrophy occurs early during critical illnesses. Although diffuse, this atrophy may specifically affect the diaphragm under artificial inactivity accompanying invasive mechanical ventilation. The primary objective of this study was to highlight diaphragm atrophy during the first 5 days of critical illness. Monitoring of pectoral thickness (a nonpostural muscle with mainly phasic function) served as a control. METHODS Diaphragm and pectoral thicknesses were measured by ultrasound within the first 24 h of admission in 97 critically ill patients, including 62 on mechanical ventilation. Thirty-five patients were reexamined at day 5. RESULTS Baseline median (interquartile) values of diaphragm and pectoral thicknesses at day 1 were 2.4 (2.0, 2.9) and 5.9 (4.7, 7.2) mm, respectively (n = 97). Higher values of diaphragm thickness at baseline were positively associated with male sex, chronic obstructive pulmonary disease, and diabetes. Diaphragm and pectoral atrophies (defined as a decrease of 10% or more between day 1 and day 5) were detected in 48% (17 of 35) and 29% (10 of 34) respectively, and were uncorrelated with each other. Diaphragm atrophy was significantly more frequent in patients with septic shock and in those with mechanical ventilation, as compared with their respective counterparts (71% [10 of 14] vs. 33% [7 of 21], P = 0.027 and 71% [17 of 28] vs. 0% [0 of 7], P = 0.004, respectively), whereas pectoral atrophy was more common in patients treated with steroids as compared with their counterparts (58% [7 of 12] vs. 14% [3 of 22], P = 0.006). A statistically significant association between diaphragm atrophy and outcome was not found. Pectoral atrophy seemed associated with less successful weaning from mechanical ventilation at day 14 (12% [1 of 8] vs. 58% [11 of 19], P = 0.043). CONCLUSIONS Ultrasound enables identification of specific early diaphragm atrophy that affects the majority of mechanically ventilated patients and septic shock patients. Diaphragm atrophy and pectoral muscle atrophy seem to be two unrelated processes.
Collapse
|
36
|
Li J, Scott JB, Duan J, Liu K, Fink JB. More than just a screen to liberate from mechanical ventilation: treat to keep extubated? ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S338. [PMID: 32016056 DOI: 10.21037/atm.2019.09.100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Jie Li
- Department of Cardiopulmonary Sciences, Division of Respiratory Care, Rush University Medical Center, Chicago, IL, USA
| | - J Brady Scott
- Department of Cardiopulmonary Sciences, Division of Respiratory Care, Rush University Medical Center, Chicago, IL, USA
| | - Jun Duan
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Kai Liu
- Department of Critical Care Medicine, Shanghai Zhongshan Hospital, Fu Dan University, Shanghai 200032, China
| | - James B Fink
- Department of Cardiopulmonary Sciences, Division of Respiratory Care, Rush University Medical Center, Chicago, IL, USA.,Aerogen Pharma Corp, San Mateo, CA, USA
| |
Collapse
|
37
|
Prevalence and development of chronic critical illness in acute patients admitted to a respiratory intensive care setting. Pulmonology 2019; 26:151-158. [PMID: 31672594 DOI: 10.1016/j.pulmoe.2019.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Chronic Critical Illness (chronic CI) is a condition associated to patients surviving an episode of acute respiratory failure (ARF). The prevalence and the factors associated with the development of chronic CI in the population admitted to a Respiratory Intensive Care Unit (RICU) have not yet been clarified. METHODS An observational prospective cohort study was undertaken at the RICU of the University Hospital of Modena (Italy). Patients mechanically ventilated with ARF in RICU were enrolled. Demographics, severity scores (APACHEII, SOFA, SAPSII), and clinical condition (septic shock, pneumonia, ARDS) were recorded on admission. Respiratory mechanics and inflammatory-metabolic blood parameters were measured both on admission and over the first week of stay. All variables were tested as predictors of chronic CI through univariate and multivariate analysis. RESULTS Chronic CI occurred in 33 out of 100 patients observed. Higher APACHEII, the presence of septic shock, diaphragmatic dysfunction (DD) at sonography, multidrug-resistant (MDR) bacterial infection, the occurrence of a second infection during stay, and a C-reactive protein (CRP) serum level inceasing 7 days over admission were associated with chronic CI. Septic shock was the strongest predictor of chronic CI (AUC = 0.92 p < 0.0001). CONCLUSIONS Chronic CI is frequent in patients admitted to RICU and mechanically ventilated due to ARF. Infection-related factors seem to play a major role as predictors of this syndrome.
Collapse
|
38
|
Liu Y, Chen N, Chang C, Lin S, Kao K, Hu H, Chang G, Li L. Ethyl pyruvate attenuates ventilation-induced diaphragm dysfunction through high-mobility group box-1 in a murine endotoxaemia model. J Cell Mol Med 2019; 23:5679-5691. [PMID: 31339670 PMCID: PMC6652995 DOI: 10.1111/jcmm.14478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/12/2019] [Accepted: 05/22/2019] [Indexed: 01/05/2023] Open
Abstract
Mechanical ventilation (MV) can save the lives of patients with sepsis. However, MV in both animal and human studies has resulted in ventilator-induced diaphragm dysfunction (VIDD). Sepsis may promote skeletal muscle atrophy in critically ill patients. Elevated high-mobility group box-1 (HMGB1) levels are associated with patients requiring long-term MV. Ethyl pyruvate (EP) has been demonstrated to lengthen survival in patients with severe sepsis. We hypothesized that the administration of HMGB1 inhibitor EP or anti-HMGB1 antibody could attenuate sepsis-exacerbated VIDD by repressing HMGB1 signalling. Male C57BL/6 mice with or without endotoxaemia were exposed to MV (10 mL/kg) for 8 hours after administrating either 100 mg/kg of EP or 100 mg/kg of anti-HMGB1 antibody. Mice exposed to MV with endotoxaemia experienced augmented VIDD, as indicated by elevated proteolytic, apoptotic and autophagic parameters. Additionally, disarrayed myofibrils and disrupted mitochondrial ultrastructures, as well as increased HMGB1 mRNA and protein expression, and plasminogen activator inhibitor-1 protein, oxidative stress, autophagosomes and myonuclear apoptosis were also observed. However, MV suppressed mitochondrial cytochrome C and diaphragm contractility in mice with endotoxaemia (P < 0.05). These deleterious effects were alleviated by pharmacologic inhibition with EP or anti-HMGB1 antibody (P < 0.05). Our data suggest that EP attenuates endotoxin-enhanced VIDD by inhibiting HMGB1 signalling pathway.
Collapse
Affiliation(s)
- Yung‐Yang Liu
- Chest DepartmentTaipei Veterans General HospitalTaipeiTaiwan
- Institutes of Clinical MedicineSchool of MedicineNational Yang‐Ming UniversityTaipeiTaiwan
| | - Ning‐Hung Chen
- Department of Internal Medicine, Division of Pulmonary and Critical Care MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Internal MedicineChang Gung UniversityTaoyuanTaiwan
- Department of Respiratory TherapyChang Gung Memorial HospitalTaoyuanTaiwan
| | - Chih‐Hao Chang
- Department of Internal Medicine, Division of Pulmonary and Critical Care MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Internal MedicineChang Gung UniversityTaoyuanTaiwan
| | - Shih‐Wei Lin
- Department of Internal Medicine, Division of Pulmonary and Critical Care MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Internal MedicineChang Gung UniversityTaoyuanTaiwan
- Department of Respiratory TherapyChang Gung Memorial HospitalTaoyuanTaiwan
| | - Kuo‐Chin Kao
- Department of Internal Medicine, Division of Pulmonary and Critical Care MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Internal MedicineChang Gung UniversityTaoyuanTaiwan
- Department of Respiratory TherapyChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Respiratory Care, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Han‐Chung Hu
- Department of Internal Medicine, Division of Pulmonary and Critical Care MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Internal MedicineChang Gung UniversityTaoyuanTaiwan
- Department of Respiratory TherapyChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Respiratory Care, College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Gwo‐Jyh Chang
- Graduate Institute of Clinical Medical SciencesChang Gung UniversityTaoyuanTaiwan
| | - Li‐Fu Li
- Department of Internal Medicine, Division of Pulmonary and Critical Care MedicineChang Gung Memorial HospitalTaoyuanTaiwan
- Department of Internal MedicineChang Gung UniversityTaoyuanTaiwan
- Department of Respiratory TherapyChang Gung Memorial HospitalTaoyuanTaiwan
| |
Collapse
|
39
|
Piva S, Fagoni N, Latronico N. Intensive care unit-acquired weakness: unanswered questions and targets for future research. F1000Res 2019; 8. [PMID: 31069055 PMCID: PMC6480958 DOI: 10.12688/f1000research.17376.1] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2019] [Indexed: 12/23/2022] Open
Abstract
Intensive care unit-acquired weakness (ICU-AW) is the most common neuromuscular impairment in critically ill patients. We discuss critical aspects of ICU-AW that have not been completely defined or that are still under discussion. Critical illness polyneuropathy, myopathy, and muscle atrophy contribute in various proportions to ICU-AW. Diagnosis of ICU-AW is clinical and is based on Medical Research Council sum score and handgrip dynamometry for limb weakness and recognition of a patient's ventilator dependency or difficult weaning from artificial ventilation for diaphragmatic weakness (DW). ICU-AW can be caused by a critical illness polyneuropathy, a critical illness myopathy, or muscle disuse atrophy, alone or in combination. Its diagnosis requires both clinical assessment of muscle strength and complete electrophysiological evaluation of peripheral nerves and muscles. The peroneal nerve test (PENT) is a quick simplified electrophysiological test with high sensitivity and good specificity that can be used instead of complete electrophysiological evaluation as a screening test in non-cooperative patients. DW, assessed by bilateral phrenic nerve magnetic stimulation or diaphragm ultrasound, can be an isolated event without concurrent limb muscle involvement. Therefore, it remains uncertain whether DW and limb weakness are different manifestations of the same syndrome or are two distinct entities. Delirium is often associated with ICU-AW but a clear correlation between these two entities requires further studies. Artificial nutrition may have an impact on ICU-AW, but no study has assessed the impact of nutrition on ICU-AW as the primary outcome. Early mobilization improves activity limitation at hospital discharge if it is started early in the ICU, but beneficial long-term effects are not established. Determinants of ICU-AW can be many and can interact with each other. Therefore, future studies assessing early mobilization should consider a holistic patient approach with consideration of all components that may lead to muscle weakness.
Collapse
Affiliation(s)
- Simone Piva
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy, 25123, Italy.,Department of Anesthesia, Intensive Care and Emergency, ASST Spedali Civili University Hospital, Brescia, Italy, 25123, Italy
| | - Nazzareno Fagoni
- Department of Anesthesia, Intensive Care and Emergency, ASST Spedali Civili University Hospital, Brescia, Italy, 25123, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy, 25123, Italy
| | - Nicola Latronico
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy, 25123, Italy.,Department of Anesthesia, Intensive Care and Emergency, ASST Spedali Civili University Hospital, Brescia, Italy, 25123, Italy
| |
Collapse
|
40
|
EL PACIENTE CRÍTICO CRÓNICO. REVISTA MÉDICA CLÍNICA LAS CONDES 2019. [DOI: 10.1016/j.rmclc.2019.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
41
|
Goligher EC. Myotrauma in mechanically ventilated patients. Intensive Care Med 2019; 45:881-884. [PMID: 30741329 DOI: 10.1007/s00134-019-05557-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/02/2019] [Indexed: 10/27/2022]
Affiliation(s)
- Ewan C Goligher
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada. .,Department of Medicine, Division of Respirology, University Health Network, Toronto, Canada. .,Toronto General Hospital Research Institute, 585 University Ave., 11-PMB Room 192, Toronto, ON, M5G 2N2, Canada.
| |
Collapse
|
42
|
Schefold JC, Messmer AS, Wenger S, Müller L, von Haehling S, Doehner W, McPhee JS, Fux M, Rösler KM, Scheidegger O, Olariu R, Z’Graggen W, Rezzi S, Grathwohl D, Konz T, Takala J, Cuenoud B, Jakob SM. Nutrient pattern analysis in critically ill patients using Omics technology (NAChO) - Study protocol for a prospective observational study. Medicine (Baltimore) 2019; 98:e13937. [PMID: 30608424 PMCID: PMC6344160 DOI: 10.1097/md.0000000000013937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Intensive care unit-acquired weakness (ICU-AW) is often observed in critically ill patients with prolonged intensive care unit (ICU) stay. We hypothesized that evolving metabolic abnormalities during prolonged ICU stay are reflected by changing nutrient patterns in blood, urine and skeletal muscle, and that these patterns differ in patients with/without ICU-AW and between patients with/without sepsis. METHODS In a prospective single-center observational trial, we aim to recruit 100 critically ill patients (ICU length of stay ≥ 5 days) with severe sepsis/septic shock ("sepsis group", n = 50) or severe head trauma/intracerebral hemorrhage ("CNS group", n = 50). Patients will be sub-grouped for presence or absence of ICU-AW as determined by the Medical Research Council sum score. Blood and urine samples will be collected and subjected to comprehensive nutrient analysis at different time points by targeted quantitative mass spectrometric methods. In addition, changes in muscular tissue (biopsy, when available), muscular architecture (ultrasound), electrophysiology, body composition analyses (bioimpedance, cerebral magnetic resonance imaging), along with clinical status will be assessed. Patients will be followed-up for 180 and 360 days including assessment of quality of life. DISCUSSION Key objective of this trial is to assess changes in nutrient pattern in blood and urine over time in critically ill patients with/without ICU-AW by using quantitative nutrient analysis techniques. Peer-reviewed published NAChO data will allow for a better understanding of metabolic changes in critically ill patients on standard liquid enteral nutrition and will likely open up new avenues for future therapeutic and nutritional interventions.
Collapse
Affiliation(s)
- Joerg C. Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Anna S. Messmer
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Stefanie Wenger
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Lionel Müller
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Stephan von Haehling
- Metabolic Research Unit, Department Cardiology and Pneumology, University of Göttingen, Göttingen, Germany
| | - Wolfram Doehner
- Neuro Research Center, Charite University Medicine Berlin, Berlin, Germany
| | - Jamie S. McPhee
- Musculoskeletal Science and Sports Medicine Research Centre, Manchester Metropolitan University, Manchester, United Kingdom
| | - Michaela Fux
- Clinical Cytomics Facility, University Institute of Clinical Chemistry and Centre of Laboratory Medicine
| | | | | | | | - Werner Z’Graggen
- Depts. of Neurosurgery and Neurology, Inselspital, Bern University Hospital, University of Bern
| | - Serge Rezzi
- Nestlé Research, vers-chez-les-Blanc, Lausanne
- Swiss Vitamin Institute, Epalinges, Switzerland
| | | | - Tobias Konz
- Nestlé Research, vers-chez-les-Blanc, Lausanne
| | - Jukka Takala
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | | | - Stephan M. Jakob
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
| |
Collapse
|
43
|
Peña-López Y, Ramirez-Estrada S, Eshwara VK, Rello J. Limiting ventilator-associated complications in ICU intubated subjects: strategies to prevent ventilator-associated events and improve outcomes. Expert Rev Respir Med 2018; 12:1037-1050. [PMID: 30460868 DOI: 10.1080/17476348.2018.1549492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Intubation is required to maintain the airways in comatose patients and enhance oxygenation in hypoxemic or ventilation in hypercapnic subjects. Recently, the Centers of Disease Control (CDC) created new surveillance definitions designed to identify complications associated with poor outcomes. Areas covered: The new framework proposed by CDC, Ventilator-Associated Events (VAE), has a range of definitions encompassing Ventilator-Associated Conditions (VAC), Infection-related Ventilator-Associated Complications (IVAC), or Possible Ventilator-Associated Pneumonia - suggesting replacing the traditional definitions of Ventilator-Associated Tracheobronchitis (VAT) and Ventilator-Associated Pneumonia (VAP). They focused more on oxygenation variations than on Chest-X rays or inflammatory biomarkers. This article will review the spectrum of infectious (VAP & VAT) complications, as well as the main non-infectious complications, namely pulmonary edema, acute respiratory distress syndrome (ARDS) and atelectasis. Strategies to limit these complications and improve outcomes will be presented. Expert commentary: Improving outcomes should be the objective of implementing bundles of prevention, based on risk factors amenable of intervention. Promotion of measures that reduce the exposition or duration of intubation should be a priority.
Collapse
Affiliation(s)
- Yolanda Peña-López
- a Pediatric Critical Care Department , Vall d'Hebron Barcelona Hospital Campus , Barcelona , Spain
| | | | - Vandana Kalwaje Eshwara
- c Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education , Manipal University , Manipal , India
| | - Jordi Rello
- d Clinical Research/epidemiology In Pneumonia & Sepsis , Vall d'Hebron Institut of Research & Centro de Investigacion Biomedica en Red (CIBERES) , Barcelona , Spain
| |
Collapse
|