1
|
Bhattarai S, Kaufmann E, Liang F, Zheng Y, Gusev E, Hamid Q, Ding J, Divangahi M, Petrof B. Characterization of SARS-CoV-2 Entry Genes in Skeletal Muscle and Impacts of In Vitro Versus In Vivo Infection. J Cachexia Sarcopenia Muscle 2025; 16:e13705. [PMID: 39871399 PMCID: PMC11772215 DOI: 10.1002/jcsm.13705] [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: 05/11/2024] [Revised: 12/19/2024] [Accepted: 01/02/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND COVID-19 has been associated with both respiratory (diaphragm) and non-respiratory (limb) muscle atrophy. It is unclear if SARS-CoV-2 infection of skeletal muscle plays a role in these changes. This study sought to: 1) determine if cells comprising skeletal muscle tissue, particularly myofibres, express the molecular components required for SARS-CoV-2 infection; 2) assess the capacity for direct SARS-CoV-2 infection and its impact on atrophy pathway genes in myogenic cells; and 3) in an animal model of COVID-19, examine the relationship between viral infection of skeletal muscle and myofibre atrophy within the diaphragm and limb muscles. METHODS We used in silico bioinformatics analysis of published human single cell RNA-seq datasets, as well as direct qPCR examination of human myotubes and diaphragm biopsies, to assess expression of key genes involved in SARS-CoV-2 cellular entry. In Vitro, we determined the ability of SARS-CoV-2 to directly infect myogenic cells and employed qPCR to assess the impact on muscle atrophy pathway genes (ubiquitin-proteasome, autophagy). In vivo, the diaphragm and quadriceps of Roborovski hamsters with SARS-CoV-2 respiratory infection were examined at day 3 post-inoculation to evaluate the relationship between atrophy pathway and SARS-CoV-2 transcripts by qPCR, as well as histological measurements of myofibre morphology. RESULTS Angiotensin converting enzyme 2 (ACE2), the primary receptor for SARS-CoV-2, as well as cooperating proteases (furin, cathepsins B and L), are expressed by myofibres. ACE2 expression was increased 5-fold (p = 0.01) in the diaphragms of mechanically ventilated human subjects compared to controls. In Vitro, a time-dependent increase of SARS-CoV-2 transcript levels was observed in myotubes directly exposed to the virus (p = 0.002). This was associated with downregulation of the ubiquitin ligase MuRF1 (by 64%, p = 0.002) and the autophagy gene LC3B (by 31%, p = 0.009). In contrast, in vivo infection led to upregulation of MuRF1 in quadriceps (23-fold, p = 0.0007) and autophagy genes in both quadriceps (5.2-fold for Gabarapl1, p = 0.03; 7-fold for p62, p = 0.0002) and diaphragm (2.2-fold for Gabarapl1, p = 0.03; 2.3-fold for p62, p = 0.057). In infected hamsters the diaphragm lacked viral transcripts but exhibited atrophy (48% decrease in myofibre area; p = 0.02), whereas the quadriceps lacked myofibre atrophy despite elevated viral transcripts in the muscle. CONCLUSIONS Although myogenic cells express the genes required for SARS-CoV-2 entry and can be directly infected, there was no evident relationship between viral transcript levels and manifestations of atrophy, either in vitro or in vivo. Our results do not support direct myofibre infection by SARS-CoV-2 as a likely cause of atrophy in COVID-19.
Collapse
Affiliation(s)
- Salyan Bhattarai
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
- Respiratory DivisionDepartment of MedicineMcGill UniversityMontrealQuebecCanada
| | - Eva Kaufmann
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
- Department of Biomedical and Molecular SciencesQueens UniversityKingstonOntarioCanada
| | - Feng Liang
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
- Respiratory DivisionDepartment of MedicineMcGill UniversityMontrealQuebecCanada
| | - Yumin Zheng
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
| | - Ekaterina Gusev
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
- Respiratory DivisionDepartment of MedicineMcGill UniversityMontrealQuebecCanada
| | - Qutayba Hamid
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
- Respiratory DivisionDepartment of MedicineMcGill UniversityMontrealQuebecCanada
- Sharjah Institute for Medical ResearchUniversity of SharjahUAE
| | - Jun Ding
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
- Respiratory DivisionDepartment of MedicineMcGill UniversityMontrealQuebecCanada
| | - Maziar Divangahi
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
- Respiratory DivisionDepartment of MedicineMcGill UniversityMontrealQuebecCanada
| | - Basil J. Petrof
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontrealQuebecCanada
- Respiratory DivisionDepartment of MedicineMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
2
|
Bahadoran Z, Mirmiran P, Ghasemi A. Type 2 diabetes-related sarcopenia: role of nitric oxide. Nutr Metab (Lond) 2024; 21:107. [PMID: 39695784 DOI: 10.1186/s12986-024-00883-z] [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: 09/07/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
Sarcopenia, characterized by progressive and generalized loss of skeletal muscle (SkM) mass, strength, and physical performance, is a prevalent complication in type 2 diabetes (T2D). Nitric oxide (NO), a multifunctional gasotransmitter involved in whole-body glucose and insulin homeostasis, plays key roles in normal SkM physiology and function. Here, we highlight the role of NO in SkM mass maintenance and its potential contribution to the development of T2D-related sarcopenia. Physiologic NO level, primarily produced by sarcolemmal neuronal nitric oxide synthase (nNOSμ isoform), is involved in protein synthesis in muscle fibers and maintenance of SkM mass. The observed effect of nNOSμ on SkM mass is muscle-type specific and sex-dependent. Impaired NO homeostasis [due to a diminished nNOSμ-NO availability and excessive NO production through inducible NOS (iNOS) in response to atrophic stimuli, e.g., inflammatory cytokines] in SkM occurred during the development and progression of T2D, may cause sarcopenia. Theoretically, restoration of NO through nNOS overexpression, supplying NOS substrates (e.g., L-arginine and L-citrulline), phosphodiesterase (PDE) inhibition, and supplementation with NO donors (e.g., inorganic nitrate) may be potential therapeutic approaches to preserve SkM mass and prevents sarcopenia in T2D.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Micronutrient Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Sahid-Erabi St, Yemen St, Chamran Exp, P.O. Box 19395-4763, Tehran, Iran.
| |
Collapse
|
3
|
Hashimoto H, Okazaki T, Honkura Y, Ren Y, Ngamsnae P, Hisaoka T, Koshiba Y, Suzuki J, Ebihara S, Katori Y. Nrf2 Deficiency Exacerbates the Decline in Swallowing and Respiratory Muscle Mass and Function in Mice with Aspiration Pneumonia. Int J Mol Sci 2024; 25:11829. [PMID: 39519380 PMCID: PMC11546094 DOI: 10.3390/ijms252111829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Aspiration pneumonia exacerbates swallowing and respiratory muscle atrophy. It induces respiratory muscle atrophy through three steps: proinflammatory cytokine production, caspase-3 and calpain, and then ubiquitin-proteasome activations. In addition, autophagy induces swallowing muscle atrophy. Nrf2 is the central detoxifying and antioxidant gene whose function in aspiration pneumonia is unclear. We explored the role of Nrf2 in aspiration pneumonia by examining swallowing and respiratory muscle mass and function using wild-type and Nrf2-knockout mice. Pepsin and lipopolysaccharide aspiration challenges caused aspiration pneumonia. The swallowing (digastric muscles) and respiratory (diaphragm) muscles were isolated. Quantitative RT-PCR and Western blotting were used to assess their proteolysis cascade. Pathological and videofluoroscopic examinations evaluated atrophy and swallowing function, respectively. Nrf2-knockouts showed exacerbated aspiration pneumonia compared with wild-types. Nrf2-knockouts exhibited more persistent and intense proinflammatory cytokine elevation than wild-types. In both mice, the challenge activated calpains and caspase-3 in the diaphragm but not in the digastric muscles. The digastric muscles showed extended autophagy activation in Nrf2-knockouts compared to wild-types. The diaphragms exhibited autophagy activation only in Nrf2-knockouts. Nrf2-knockouts showed worsened muscle atrophies and swallowing function compared with wild-types. Thus, activation of Nrf2 may alleviate inflammation, muscle atrophy, and function in aspiration pneumonia, a major health problem for the aging population, and may become a therapeutic target.
Collapse
Affiliation(s)
- Hikaru Hashimoto
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (H.H.)
| | - Tatsuma Okazaki
- Department of Rehabilitation Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan (S.E.)
- Center for Dysphagia of Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yohei Honkura
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (H.H.)
- Center for Dysphagia of Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yuzhuo Ren
- Department of Rehabilitation Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan (S.E.)
| | - Peerada Ngamsnae
- Department of Rehabilitation Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan (S.E.)
| | - Takuma Hisaoka
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (H.H.)
- Center for Dysphagia of Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yasutoshi Koshiba
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (H.H.)
- Center for Dysphagia of Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Jun Suzuki
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (H.H.)
- Center for Dysphagia of Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Satoru Ebihara
- Department of Rehabilitation Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan (S.E.)
- Center for Dysphagia of Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yukio Katori
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan; (H.H.)
- Center for Dysphagia of Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| |
Collapse
|
4
|
Pu H, Doig GS, Lv Y, Wu X, Yang F, Zhang S, Liang Z, Zhou Y, Kang Y. Modifiable risk factors for ventilator associated diaphragmatic dysfunction: a multicenter observational study. BMC Pulm Med 2023; 23:343. [PMID: 37700263 PMCID: PMC10498609 DOI: 10.1186/s12890-023-02633-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Diaphragmatic dysfunction is known to be associated with difficulties weaning from invasive mechanical ventilation and is related to worse patient outcomes yet our understanding of how to prevent diaphragmatic dysfunction remains incomplete. We examined potentially modifiable risk factors for diaphragmatic dysfunction and attempted to estimate benefits attributable to altering these modifiable risk factors. METHODS This prospective multicenter observational study was undertaken in the general ICUs of two tertiary care teaching hospitals. Critically ill adults expected to receive invasive mechanical ventilation for at least 48 h were enrolled. Diaphragm function was assessed by ultrasound each study day, with dysfunction defined as thickening fraction less than 20%. RESULTS From January to December 2019, 856 patients were screened and 126 patients were enrolled. Overall, 40.5% (51/126) of patients experienced diaphragmatic dysfunction during invasive mechanical ventilation. Patients with diaphragmatic dysfunction were more likely to develop ventilator associated pneumonia (risk difference [RD] + 12.9%, 95% Confidence Interval [CI] 1.4 to 24.4%, P = 0.028), were more likely to experience extubation failure (RD + 8.5%, 95% CI 0.4 to 16.6%, P = 0.039) and required a longer duration of invasive mechanical ventilation (RD + 1.3 days, 95% CI 0.1 to 2.5 days, P = 0.035). They also required a longer hospital stay (RD + 1.2 days, 95% CI 0.04 to 2.4 days, P = 0.041) and were more likely to die before hospital discharge (RD + 18.1%, 95% CI 3.7 to 32.5%, P = 0.014). Multivariable analysis considered the impact of age, sex, pre-existing nutritional status, caloric intake, amino acid intake, acute disease severity, modes of mechanical ventilation, measures of respiratory status, sedation, pain control and baseline diaphragm thickness. Only SOFA score (P = 0.008) and early amino acid intake (P = 0.001) remained significant independent risk factors for the onset of diaphragmatic dysfunction. Causal path modeling suggested early amino acid intake may significantly reduce diaphragmatic dysfunction (RRR 29%, 95% CI 10% to 48%, P = 0.003) and may also reduce mortality (RRR 49%, 95% CI 25% to 73%, P < 0.0001). CONCLUSIONS Amino acid intake during the first 24 h of ICU stay may represent an important, modifiable risk factor for diaphragmatic dysfunction and may have a direct causal effect on mortality. We recommend additional research on this topic.
Collapse
Affiliation(s)
- Hong Pu
- Department of Critical Care Medicine, West China Medical School, West China Hospital, Sichuan University, Chengdu, PR China
| | - Gordon S Doig
- Northern Clinical School Intensive Care Research Unit, Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| | - Yu Lv
- Healthcare-Associated Infection Control Center, Sichuan Academy of Medical Sciences, School of Medicine, Sichuan People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xiaoxiao Wu
- Department of Critical Care Medicine, Sichuan Academy of Medical Sciences, School of Medicine, Sichuan People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Fu Yang
- Department of Critical Care Medicine, West China Medical School, West China Hospital, Sichuan University, Chengdu, PR China
| | - Shurong Zhang
- Department of Critical Care Medicine, West China Medical School, West China Hospital, Sichuan University, Chengdu, PR China
| | - Zongan Liang
- Department of Respiratory and Critical Care Medicine, West China Medical School, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yan Zhou
- Department of Critical Care Medicine, West China Medical School, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yan Kang
- Department of Critical Care Medicine, West China Medical School, West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
5
|
Prolonged Mechanical Ventilation: Outcomes and Management. J Clin Med 2022; 11:jcm11092451. [PMID: 35566577 PMCID: PMC9103623 DOI: 10.3390/jcm11092451] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/23/2022] [Accepted: 04/24/2022] [Indexed: 02/01/2023] Open
Abstract
The number of patients requiring prolonged mechanical ventilation (PMV) is increasing worldwide, placing a burden on healthcare systems. Therefore, investigating the pathophysiology, risk factors, and treatment for PMV is crucial. Various underlying comorbidities have been associated with PMV. The pathophysiology of PMV includes the presence of an abnormal respiratory drive or ventilator-induced diaphragm dysfunction. Numerous studies have demonstrated that ventilator-induced diaphragm dysfunction is related to increases in in-hospital deaths, nosocomial pneumonia, oxidative stress, lung tissue hypoxia, ventilator dependence, and costs. Thus far, the pathophysiologic evidence for PMV has been derived from clinical human studies and experimental studies in animals. Moreover, recent studies have demonstrated the outcome benefits of pharmacological agents and rehabilitative programs for patients requiring PMV. However, methodological limitations affected these studies. Controlled prospective studies with an adequate number of participants are necessary to provide evidence of the mechanism, prognosis, and treatment of PMV. The great epidemiologic impact of PMV and the potential development of treatment make this a key research field.
Collapse
|
6
|
Kyriakoudi A, Rovina N, Koltsida O, Kostakou E, Konstantelou E, Kardara M, Kompoti M, Palamidas A, Kaltsakas G, Koutsoukou A. Weaning Failure in Critically Ill Patients Is Related to the Persistence of Sepsis Inflammation. Diagnostics (Basel) 2021; 12:diagnostics12010092. [PMID: 35054259 PMCID: PMC8774440 DOI: 10.3390/diagnostics12010092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/16/2021] [Accepted: 12/25/2021] [Indexed: 12/02/2022] Open
Abstract
Introduction: Septic patients undergoing mechanical ventilation (MV) often experience difficulty in weaning. Th aim of this study was to determine whether inflammatory biomarkers of sepsis could be indicative of the failure or success of spontaneous breathing trial (SBT) in these patients. Methods: Sixty-five patients on MV (42 septic and 23 intubated for other reasons) fulfilling the criteria for SBT were included in the study. Blood samples were collected right before, at the end of (30 min) and 24 h after the SBT. Serum inflammatory mediators associated with sepsis (IL-18, IL-18BP, TNF) were determined and correlated with the outcome of SBT. Results: A successful SBT was achieved in 45 patients (69.2%). Septic patients had a higher percentage of SBT failure as compared to non-septic patients (85% vs. 15%, p = 0.026), with an odds ratio for failing 4.5 times (OR = 4.5 95%CI: 1.16–17.68, p 0.022). IL-18 levels and the relative mRNA expression in serum were significantly higher in septic as compared to non-septic patients (p < 0.05). Sepsis was independently associated with higher serum IL-18 and TNF levels in two time-point GEE models (53–723, p = 0.023 and 0.3–64, p = 0.048, respectively). IL-18BP displayed independent negative association with rapid shallow breathing index (RSBI) (95% CI: −17.6 to −4, p = 0.002). Conclusion: Sustained increased levels of IL-18 and IL-18BP, acknowledged markers of sepsis, were found to be indicative of SBT failure in patients recovering from sepsis. Our results show that, although subclinical, remaining septic inflammation that sustaines for a long time complicates the weaning procedure. Biomarkers for the estimation of the septic burden and the right time for weaning are needed.
Collapse
Affiliation(s)
- Anna Kyriakoudi
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and “Sotiria” Chest Disease Hospital, 11527 Athens, Greece; (A.K.); (O.K.); (E.K.); (E.K.); (A.P.); (G.K.); (A.K.)
| | - Nikoletta Rovina
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and “Sotiria” Chest Disease Hospital, 11527 Athens, Greece; (A.K.); (O.K.); (E.K.); (E.K.); (A.P.); (G.K.); (A.K.)
- Correspondence: ; Tel.: +30-210-7763650
| | - Ourania Koltsida
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and “Sotiria” Chest Disease Hospital, 11527 Athens, Greece; (A.K.); (O.K.); (E.K.); (E.K.); (A.P.); (G.K.); (A.K.)
| | - Eirini Kostakou
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and “Sotiria” Chest Disease Hospital, 11527 Athens, Greece; (A.K.); (O.K.); (E.K.); (E.K.); (A.P.); (G.K.); (A.K.)
| | - Elissavet Konstantelou
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and “Sotiria” Chest Disease Hospital, 11527 Athens, Greece; (A.K.); (O.K.); (E.K.); (E.K.); (A.P.); (G.K.); (A.K.)
| | - Matina Kardara
- 1st Department of Critical Care Medicine & Pulmonary Services, National and Kapodistrian University of Athens, Medical School, Evangelismos Hospital, 10676 Athens, Greece;
| | - Maria Kompoti
- Intensive Care Unit, General Hospital of Eleusis Thriasio, 13674 Athens, Greece;
| | - Anastasios Palamidas
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and “Sotiria” Chest Disease Hospital, 11527 Athens, Greece; (A.K.); (O.K.); (E.K.); (E.K.); (A.P.); (G.K.); (A.K.)
| | - Georgios Kaltsakas
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and “Sotiria” Chest Disease Hospital, 11527 Athens, Greece; (A.K.); (O.K.); (E.K.); (E.K.); (A.P.); (G.K.); (A.K.)
| | - Antonia Koutsoukou
- 1st Department of Respiratory Medicine, Medical School, National and Kapodistrian University of Athens and “Sotiria” Chest Disease Hospital, 11527 Athens, Greece; (A.K.); (O.K.); (E.K.); (E.K.); (A.P.); (G.K.); (A.K.)
| |
Collapse
|
7
|
Preau S, Vodovar D, Jung B, Lancel S, Zafrani L, Flatres A, Oualha M, Voiriot G, Jouan Y, Joffre J, Huel F, De Prost N, Silva S, Azabou E, Radermacher P. Energetic dysfunction in sepsis: a narrative review. Ann Intensive Care 2021; 11:104. [PMID: 34216304 PMCID: PMC8254847 DOI: 10.1186/s13613-021-00893-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 06/24/2021] [Indexed: 02/07/2023] Open
Abstract
Background Growing evidence associates organ dysfunction(s) with impaired metabolism in sepsis. Recent research has increased our understanding of the role of substrate utilization and mitochondrial dysfunction in the pathophysiology of sepsis-related organ dysfunction. The purpose of this review is to present this evidence as a coherent whole and to highlight future research directions. Main text Sepsis is characterized by systemic and organ-specific changes in metabolism. Alterations of oxygen consumption, increased levels of circulating substrates, impaired glucose and lipid oxidation, and mitochondrial dysfunction are all associated with organ dysfunction and poor outcomes in both animal models and patients. The pathophysiological relevance of bioenergetics and metabolism in the specific examples of sepsis-related immunodeficiency, cerebral dysfunction, cardiomyopathy, acute kidney injury and diaphragmatic failure is also described. Conclusions Recent understandings in substrate utilization and mitochondrial dysfunction may pave the way for new diagnostic and therapeutic approaches. These findings could help physicians to identify distinct subgroups of sepsis and to develop personalized treatment strategies. Implications for their use as bioenergetic targets to identify metabolism- and mitochondria-targeted treatments need to be evaluated in future studies. Supplementary Information The online version contains supplementary material available at 10.1186/s13613-021-00893-7.
Collapse
Affiliation(s)
- Sebastien Preau
- U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France.
| | - Dominique Vodovar
- Centre AntiPoison de Paris, Hôpital Fernand Widal, APHP, 75010, Paris, France.,Faculté de pharmacie, UMRS 1144, 75006, Paris, France.,Université de Paris, UFR de Médecine, 75010, Paris, France
| | - Boris Jung
- Medical Intensive Care Unit, Lapeyronie Teaching Hospital, Montpellier University Hospital and PhyMedExp, University of Montpellier, Montpellier, France
| | - Steve Lancel
- U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Lara Zafrani
- Médecine Intensive Réanimation, Hôpital Saint-Louis, AP-HP, Université de Paris, Paris, France.,INSERM UMR 976, Hôpital Saint Louis, Université de Paris, Paris, France
| | | | - Mehdi Oualha
- Pediatric Intensive Care Unit, Necker Hospital, APHP, Centre - Paris University, Paris, France
| | - Guillaume Voiriot
- Service de Médecine Intensive Réanimation, Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Hôpital Tenon, Paris, France
| | - Youenn Jouan
- Service de Médecine Intensive Réanimation, CHRU Tours, Tours, France.,Faculté de Médecine de Tours, INSERM U1100 Centre d'Etudes des Pathologies Respiratoires, Tours, France
| | - Jeremie Joffre
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, 94143, USA
| | - Fabrice Huel
- Réanimation médico-chirurgicale, Université de Paris, Assistance Publique - Hôpitaux de Paris, Hôpital Louis Mourier, Paris, France
| | - Nicolas De Prost
- Service de Réanimation Médicale, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Cedex 94010, Créteil, France
| | - Stein Silva
- Réanimation URM CHU Purpan, Cedex 31300, Toulouse, France.,Toulouse NeuroImaging Center INSERM1214, Cedex 31300, Toulouse, France
| | - Eric Azabou
- Clinical Neurophysiology and Neuromodulation Unit, Departments of Physiology and Critical Care Medicine, Raymond Poincaré Hospital, AP-HP, Inserm UMR 1173, Infection and Inflammation (2I), University of Versailles (UVSQ), Paris-Saclay University, Paris, France
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum, Ulm, Germany
| |
Collapse
|
8
|
Corradi F, Isirdi A, Malacarne P, Santori G, Barbieri G, Romei C, Bove T, Vetrugno L, Falcone M, Bertini P, Guarracino F, Landoni G, Forfori F. Low diaphragm muscle mass predicts adverse outcome in patients hospitalized for COVID-19 pneumonia: an exploratory pilot study. Minerva Anestesiol 2021; 87:432-438. [PMID: 33594871 DOI: 10.23736/s0375-9393.21.15129-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND The aim of this study was to evaluate whether measurement of diaphragm thickness (DT) by ultrasonography may be a clinically useful noninvasive method for identifying patients at risk of adverse outcomes defined as need of invasive mechanical ventilation or death. METHODS We prospectively enrolled 77 patients with laboratory-confirmed COVID-19 infection admitted to our intermediate care unit in Pisa between March 5 and March 30, 2020, with follow-up until hospital discharge or death. Logistic regression was used identify variables potentially associated with adverse outcomes and those P<0.10 were entered into a multivariate logistic regression model. Cumulative probability for lack of adverse outcomes in patients with or without low baseline diaphragm muscle mass was calculated with the Kaplan-Meier product-limit estimator. RESULTS The main findings of this study are that: 1) patients who developed adverse outcomes had thinner diaphragm than those who did not (2.0 vs. 2.2 mm, P=0.001); and 2) DT and lymphocyte count were independent significant predictors of adverse outcomes, with end-expiratory DT being the strongest (ß=-708; OR=0.492; P=0.018). CONCLUSIONS Diaphragmatic ultrasound may be a valid tool to evaluate the risk of respiratory failure. Evaluating the need of mechanical ventilation treatment should be based not only on PaO<inf>2</inf>/FiO<inf>2</inf>, but on a more comprehensive assessment including DT because if the lungs become less compliant a thinner diaphragm, albeit free of intrinsic abnormality, may become exhausted, thus contributing to severe respiratory failure.
Collapse
Affiliation(s)
- Francesco Corradi
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy - .,E.O. Ospedali Galliera, Genoa, Italy -
| | - Alessandro Isirdi
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy.,E.O. Ospedali Galliera, Genoa, Italy
| | | | - Gregorio Santori
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Greta Barbieri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Tiziana Bove
- Anesthesiology and Intensive Care Clinic, Department of Medicine, University of Udine, Udine, Italy
| | - Luigi Vetrugno
- Anesthesiology and Intensive Care Clinic, Department of Medicine, University of Udine, Udine, Italy
| | - Marco Falcone
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | | - Giovanni Landoni
- Department of Anesthesiology and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy.,School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Forfori
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy.,E.O. Ospedali Galliera, Genoa, Italy
| | | |
Collapse
|
9
|
Hydrocortisone mitigates ICU-AW by fine-tuning of muscle atrophic and hypertrophic signaling pathways in a sepsis model with limb immobilization. Life Sci 2020; 261:118366. [PMID: 32871182 DOI: 10.1016/j.lfs.2020.118366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/14/2020] [Accepted: 08/27/2020] [Indexed: 12/29/2022]
Abstract
AIMS Intensive care unit-acquired weakness (ICU-AW) is a complex spectrum of disability that delays recovery of critically ill-immobilized patients with sepsis. Much discrepancy remain on the use of corticosteroids and their impact on muscle regeneration in critical illness management. Therefore, the aim of this study is to investigate whether hydrocortisone (HCT) modulates muscle mass turnover in ICU-AW induced by sepsis with limb immobilization (SI). MAIN METHODS Sepsis by cecal ligation puncture (CLP) with forelimb-immobilization were performed in rats. The study consisted of four groups: Sham (left forelimb-immobilization), Sham HCT (left forelimb-immobilization + HCT), SI (CLP + left forelimb-immobilization) and SI HCT (CLP + left forelimb-immobilization + HCT). Motor force, blood and muscle sampling were assessed. KEY FINDINGS HCT prevented body weight loss associated with SI and attenuated systemic and muscular inflammation. Besides, myosin was restituted in SI HCT group in conjunction to muscle mass and strength restoration. Pro-hypertrophic calcineurin (PP2B-Aβ) and nuclear factor of activated T-cells C3 (NFATc3) but not protein kinase B (Akt) were re-activated by HCT. Finally, pro-atrophic extracellular signal-regulated kinases (ERK1/2) and p38 mitogen-activated protein kinases (p38) but not nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) were inhibited in SI HCT group. SIGNIFICANCE This study unravels new molecular events thought to control muscle protein synthesis in ICU-AW induced by sepsis and limb immobilization. HCT has a potential to fine-tune muscle-signaling pathways and to reduce the negative outcomes of ICU-AW.
Collapse
|
10
|
Dres M, Jung B, Molinari N, Manna F, Dubé BP, Chanques G, Similowski T, Jaber S, Demoule A. Respective contribution of intensive care unit-acquired limb muscle and severe diaphragm weakness on weaning outcome and mortality: a post hoc analysis of two cohorts. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:370. [PMID: 31752937 PMCID: PMC6873450 DOI: 10.1186/s13054-019-2650-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/16/2019] [Indexed: 01/16/2023]
Abstract
Background Intensive care unit (ICU)-acquired weakness (ICU-AW) and ICU-acquired diaphragm dysfunction (ICU-DD) occur frequently in mechanically ventilated (MV) patients. It is unknown whether they have different risk factors and different impacts on outcome. This study was designed to (1) describe the respective risk factors associated with ICU-AW and severe ICU-DD and (2) evaluate the respective impact of ICU-AW and severe ICU-DD on outcome. Methods Post hoc analysis of two prospective cohort studies conducted in two ICUs. In patients mechanically ventilated for at least 24 h undergoing a first spontaneous breathing trial, severe ICU-DD was defined as diaphragm twitch pressure < 7 cmH2O and ICU-AW was defined as Medical Research Council Score < 48. Results One hundred sixteen patients were assessed. Factors independently associated with severe ICU-DD were age, longer duration of MV, and exposure to sufentanil, and those factors associated with ICU-AW were longer duration of MV and exposure to norepinephrine. Severe ICU-DD (OR 3.56, p = 0.008), but not ICU-AW, was independently associated with weaning failure (59%). ICU-AW (OR 4.30, p = 0.033), but not severe ICU-DD, was associated with ICU mortality. Weaning failure and mortality rate were higher in patients with both severe ICU-DD and ICU-AW (86% and 39%, respectively) than in patients with either severe ICU-DD (64% and 0%) or ICU-AW (63% and 13%). Conclusion Severe ICU-DD and ICU-AW have different risk factors and different impacts on weaning failure and mortality. The impact of the combination of ICU-DD and ICU-AW is more pronounced than their individual impact.
Collapse
Affiliation(s)
- Martin Dres
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Boris Jung
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Medical Intensive Care Unit, Lapeyronie University Hospital, Montpellier, France
| | - Nicolas Molinari
- Department of Statistics, CHU Montpellier, IMAG, CNRS, Univ Montpellier, Montpellier, France
| | - Federico Manna
- Department of Statistics, CHU Montpellier, IMAG, CNRS, Univ Montpellier, Montpellier, France
| | - Bruno-Pierre Dubé
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Gerald Chanques
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| | - Thomas Similowski
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Samir Jaber
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| | - Alexandre Demoule
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France. .,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France. .,Service de Pneumologie, Médecine Intensive et Réanimation, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France.
| |
Collapse
|
11
|
Dres M, Jung B, Molinari N, Manna F, Dubé BP, Chanques G, Similowski T, Jaber S, Demoule A. Respective contribution of intensive care unit-acquired limb muscle and severe diaphragm weakness on weaning outcome and mortality: a post hoc analysis of two cohorts. CRITICAL CARE (LONDON, ENGLAND) 2019. [PMID: 31752937 DOI: 10.1186/s13054-019-2650-z].] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Intensive care unit (ICU)-acquired weakness (ICU-AW) and ICU-acquired diaphragm dysfunction (ICU-DD) occur frequently in mechanically ventilated (MV) patients. It is unknown whether they have different risk factors and different impacts on outcome. This study was designed to (1) describe the respective risk factors associated with ICU-AW and severe ICU-DD and (2) evaluate the respective impact of ICU-AW and severe ICU-DD on outcome. METHODS Post hoc analysis of two prospective cohort studies conducted in two ICUs. In patients mechanically ventilated for at least 24 h undergoing a first spontaneous breathing trial, severe ICU-DD was defined as diaphragm twitch pressure < 7 cmH2O and ICU-AW was defined as Medical Research Council Score < 48. RESULTS One hundred sixteen patients were assessed. Factors independently associated with severe ICU-DD were age, longer duration of MV, and exposure to sufentanil, and those factors associated with ICU-AW were longer duration of MV and exposure to norepinephrine. Severe ICU-DD (OR 3.56, p = 0.008), but not ICU-AW, was independently associated with weaning failure (59%). ICU-AW (OR 4.30, p = 0.033), but not severe ICU-DD, was associated with ICU mortality. Weaning failure and mortality rate were higher in patients with both severe ICU-DD and ICU-AW (86% and 39%, respectively) than in patients with either severe ICU-DD (64% and 0%) or ICU-AW (63% and 13%). CONCLUSION Severe ICU-DD and ICU-AW have different risk factors and different impacts on weaning failure and mortality. The impact of the combination of ICU-DD and ICU-AW is more pronounced than their individual impact.
Collapse
Affiliation(s)
- Martin Dres
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Boris Jung
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Medical Intensive Care Unit, Lapeyronie University Hospital, Montpellier, France
| | - Nicolas Molinari
- Department of Statistics, CHU Montpellier, IMAG, CNRS, Univ Montpellier, Montpellier, France
| | - Federico Manna
- Department of Statistics, CHU Montpellier, IMAG, CNRS, Univ Montpellier, Montpellier, France
| | - Bruno-Pierre Dubé
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Gerald Chanques
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| | - Thomas Similowski
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Samir Jaber
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| | - Alexandre Demoule
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France. .,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France. .,Service de Pneumologie, Médecine Intensive et Réanimation, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France.
| |
Collapse
|
12
|
Dres M, Jung B, Molinari N, Manna F, Dubé BP, Chanques G, Similowski T, Jaber S, Demoule A. Respective contribution of intensive care unit-acquired limb muscle and severe diaphragm weakness on weaning outcome and mortality: a post hoc analysis of two cohorts. CRITICAL CARE (LONDON, ENGLAND) 2019. [PMID: 31752937 DOI: 10.1186/s13054-019-2650-z]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Intensive care unit (ICU)-acquired weakness (ICU-AW) and ICU-acquired diaphragm dysfunction (ICU-DD) occur frequently in mechanically ventilated (MV) patients. It is unknown whether they have different risk factors and different impacts on outcome. This study was designed to (1) describe the respective risk factors associated with ICU-AW and severe ICU-DD and (2) evaluate the respective impact of ICU-AW and severe ICU-DD on outcome. METHODS Post hoc analysis of two prospective cohort studies conducted in two ICUs. In patients mechanically ventilated for at least 24 h undergoing a first spontaneous breathing trial, severe ICU-DD was defined as diaphragm twitch pressure < 7 cmH2O and ICU-AW was defined as Medical Research Council Score < 48. RESULTS One hundred sixteen patients were assessed. Factors independently associated with severe ICU-DD were age, longer duration of MV, and exposure to sufentanil, and those factors associated with ICU-AW were longer duration of MV and exposure to norepinephrine. Severe ICU-DD (OR 3.56, p = 0.008), but not ICU-AW, was independently associated with weaning failure (59%). ICU-AW (OR 4.30, p = 0.033), but not severe ICU-DD, was associated with ICU mortality. Weaning failure and mortality rate were higher in patients with both severe ICU-DD and ICU-AW (86% and 39%, respectively) than in patients with either severe ICU-DD (64% and 0%) or ICU-AW (63% and 13%). CONCLUSION Severe ICU-DD and ICU-AW have different risk factors and different impacts on weaning failure and mortality. The impact of the combination of ICU-DD and ICU-AW is more pronounced than their individual impact.
Collapse
Affiliation(s)
- Martin Dres
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Boris Jung
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Medical Intensive Care Unit, Lapeyronie University Hospital, Montpellier, France
| | - Nicolas Molinari
- Department of Statistics, CHU Montpellier, IMAG, CNRS, Univ Montpellier, Montpellier, France
| | - Federico Manna
- Department of Statistics, CHU Montpellier, IMAG, CNRS, Univ Montpellier, Montpellier, France
| | - Bruno-Pierre Dubé
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Gerald Chanques
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| | - Thomas Similowski
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France
| | - Samir Jaber
- Montpellier School of Medicine, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France.,Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| | - Alexandre Demoule
- AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Pneumologie, Médecine intensive - Réanimation (Département "R3S"), F-75013, Paris, France. .,AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Intensive Care Unit and Respiratory Division (Département "R3S"), F-75013, Paris, France. .,Service de Pneumologie, Médecine Intensive et Réanimation, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France.
| |
Collapse
|
13
|
Lu Z, Ge H, Xu L, Guo F, Zhang G, Wu Y. Alterations in diaphragmatic function assessed by ultrasonography in mechanically ventilated patients with sepsis. JOURNAL OF CLINICAL ULTRASOUND : JCU 2019; 47:206-211. [PMID: 30671990 DOI: 10.1002/jcu.22690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 12/01/2018] [Accepted: 12/25/2018] [Indexed: 06/09/2023]
Abstract
PURPOSE To assess alteration of diaphragmatic function by ultrasonography in a population of mechanically ventilated patients with or without sepsis. METHODS We performed a prospective, 6-month, single-center, observational cohort study. Mechanically ventilated septic and nonseptic patients were studied within 24 hours following intubation and before the moment of ventilator liberation. Diaphragm thickness and contractile activity (quantified by diaphragmatic thickening fraction, DTF) were measured by ultrasonography at the zone of apposition. Intraobserver and interobserver reproducibility were measured. RESULTS Fifty-two critically ill patients were included, 28 with sepsis and 24 without sepsis. Upon initiation of ventilation, DTF was lower in septic than that in nonseptic patients (P = 0.03). No difference was observed between septic and nonseptic patients for diaphragm thickness. Mean 188 ± 111 hours after the first measurement, both diaphragm thickness and DTF decreased significantly compared with first measurements in septic and nonseptic patients, all P < 0.001. Diaphragm thickness decreased by 9.1 ± 10.7% in nonseptic and by 16.0 ± 13.5% in septic patients, P = 0.049. DTF decreased by 15.2 ± 21.3% in nonseptic and by 30.7 ± 22.0% in septic patients, P = 0.013. CONCLUSIONS Mechanically ventilated patients with sepsis were associated with an earlier and more severe diaphragm dysfunction compared with patients without sepsis.
Collapse
Affiliation(s)
- Zhihua Lu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiqing Ge
- Department of Respiratory Care, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lilong Xu
- Department of Diagnostic Ultrasound, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Guo
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ge Zhang
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinshan Wu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
14
|
Liu YY, Li LF. Ventilator-induced diaphragm dysfunction in critical illness. Exp Biol Med (Maywood) 2018; 243:1329-1337. [PMID: 30453774 DOI: 10.1177/1535370218811950] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
IMPACT STATEMENT Mechanical ventilation (MV) is life-saving for patients with acute respiratory failure but also causes difficult liberation of patients from ventilator due to rapid decrease of diaphragm muscle endurance and strength, which is termed ventilator-induced diaphragmatic damage (VIDD). Numerous studies have revealed that VIDD could increase extubation failure, ICU stay, ICU mortality, and healthcare expenditures. However, the mechanisms of VIDD, potentially involving a multistep process including muscle atrophy, oxidative loads, structural damage, and muscle fiber remodeling, are not fully elucidated. Further research is necessary to unravel mechanistic framework for understanding the molecular mechanisms underlying VIDD, especially mitochondrial dysfunction and increased mitochondrial oxidative stress, and develop better MV strategies, rehabilitative programs, and pharmacologic agents to translate this knowledge into clinical benefits.
Collapse
Affiliation(s)
- Yung-Yang Liu
- 1 Chest Department, Taipei Veterans General Hospital, Taipei 112, Taiwan.,2 Institutes of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Li-Fu Li
- 3 Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan.,4 Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
15
|
Attenuation of ventilation-induced diaphragm dysfunction through toll-like receptor 4 and nuclear factor-κB in a murine endotoxemia model. J Transl Med 2018; 98:1170-1183. [PMID: 29925937 DOI: 10.1038/s41374-018-0081-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 01/30/2023] Open
Abstract
Mechanical ventilation (MV) is often used to maintain life in patients with sepsis and sepsis-related acute lung injury. However, controlled MV may cause diaphragm weakness due to muscle injury and atrophy, an effect termed ventilator-induced diaphragm dysfunction (VIDD). Toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) signaling pathways may elicit sepsis-related acute inflammatory responses and muscle protein degradation and mediate the pathogenic mechanisms of VIDD. However, the mechanisms regulating the interactions between VIDD and endotoxemia are unclear. We hypothesized that mechanical stretch with or without endotoxin treatment would augment diaphragmatic structural damage, the production of free radicals, muscle proteolysis, mitochondrial dysfunction, and autophagy of the diaphragm via the TLR4/NF-κB pathway. Male C57BL/6 mice, either wild-type or TLR4-deficient, aged between 6 and 8 weeks were exposed to MV (6 mL/kg or 10 mL/kg) with or without endotoxemia for 8 h. Nonventilated mice were used as controls. MV with endotoxemia aggravated VIDD, as demonstrated by the increases in the expression levels of TLR4, caspase-3, atrogin-1, muscle ring finger-1, and microtubule-associated protein light chain 3-II. In addition, increased NF-κB phosphorylation and oxidative loads, disorganized myofibrils, disrupted mitochondria, autophagy, and myonuclear apoptosis were also observed. Furthermore, MV with endotoxemia reduced P62 levels and diaphragm muscle fiber size (P < 0.05). Endotoxin-exacerbated VIDD was attenuated by pharmacologic inhibition with a NF-κB inhibitor or in TLR4-deficient mice (P < 0.05). Our data indicate that endotoxin-augmented MV-induced diaphragmatic injury occurs through the activation of the TLR4/NF-κB signaling pathway.
Collapse
|
16
|
Diaphragm Weakness in the Critically Ill: Basic Mechanisms Reveal Therapeutic Opportunities. Chest 2018; 154:1395-1403. [PMID: 30144420 DOI: 10.1016/j.chest.2018.08.1028] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/23/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022] Open
Abstract
The diaphragm is the primary muscle of inspiration. Its capacity to respond to the load imposed by pulmonary disease is a major determining factor both in the onset of ventilatory failure and in the ability to successfully separate patients from ventilator support. It has recently been established that a very large proportion of critically ill patients exhibit major weakness of the diaphragm, which is associated with poor clinical outcomes. The two greatest risk factors for the development of diaphragm weakness in critical illness are the use of mechanical ventilation and the presence of sepsis. Loss of force production by the diaphragm under these conditions is caused by a combination of defective contractility and reduced diaphragm muscle mass. Importantly, many of the same molecular mechanisms are implicated in the diaphragm dysfunction associated with both mechanical ventilation and sepsis. This review outlines the primary cellular mechanisms identified thus far at the nexus of diaphragm dysfunction associated with mechanical ventilation and/or sepsis, and explores the potential for treatment or prevention of diaphragm weakness in critically ill patients through therapeutic manipulation of these final common pathway targets.
Collapse
|
17
|
Le Dinh M, Carreira S, Obert J, Gayan-Ramirez G, Riou B, Beuvin M, Similowski T, Coirault C, Demoule A. Prolonged mechanical ventilation worsens sepsis-induced diaphragmatic dysfunction in the rat. PLoS One 2018; 13:e0200429. [PMID: 30067847 PMCID: PMC6070213 DOI: 10.1371/journal.pone.0200429] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 06/26/2018] [Indexed: 12/17/2022] Open
Abstract
Background Short-term mechanical ventilation (MV) protects against sepsis-induced diaphragmatic dysfunction. Prolonged MV induces diaphragmatic dysfunction in non-septic animals, but few reports describe the effects of prolonged MV in sepsis. We hypothesized that prolonged MV is not protective but worsens the diaphragmatic dysfunction induced by a mild sepsis, because MV and sepsis share key signaling mechanisms, such as cytokine upregulation. Method We studied the impact of prolonged MV (12 h) in four groups (n = 8) of male Wistar rats: 1) endotoxemia induced by intraperitoneal injection of Escherichia coli lipopolysaccharide, 2) MV without endotoxemia, 3) combination of endotoxemia and MV and 4) sham control. Diaphragm mechanical performance, pro-inflammatory cytokine concentrations (Tumor Necrosis Factor-α, Interleukin-1β, Interleukin-6) in plasma were measured. Results Prolonged MV and sepsis independtly reduced maximum diaphragm force (-27%, P = 0.003; -37%, P<0.001; respectively). MV and sepsis acted additively to further decrease diaphragm force (-62%, P<0.001). Similar results were observed for diaphragm kinetics (maximum lengthening velocity -47%, P<0.001). Sepsis and MV reduced diaphragm cross sectional area of type I and IIx fibers, which was further increased by the combination of sepsis and MV (all P<0.05). Sepsis and MV were individually associated with the presence of a robust perimysial inflammatory infiltrate, which was more marked when sepsis and MV were both present (all P<0.05). Sepsis and, to a lesser extent, MV increased proinflammatory cytokine production in plasma and diaphragm (all P<0.05); proinflammatory cytokine expression in plasma was increased further by the combination of sepsis and MV (all P<0.05). Maximum diaphragm force correlated negatively with plasma and diaphragmatic cytokine production (all p<0.05). Conclusions Prolonged (12 h) MV exacerbated sepsis-induced decrease in diaphragm performance. Systemic and diaphragmatic overproduction of pro-inflammatory cytokines may contribute to diaphragm weakness.
Collapse
Grants
- Alexandre Demoule reports personal fees from Maquet, grants, personal fees and non-financial support from Covidien, personal fees from MSD, grants and non-financial support from Philips, non-financial support from Drager, grants and personal fees from Resmed, personal fees from fisher & Paykel, all outside the submitted work. The funder provided support in the form of salaries for AD, but did not have any additional role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. The specific roles of this author is articulated in the ‘author contributions’ section.
- Chancellerie des Universités de Paris
- Institut Fédératif de Recherche 14-INSERM
- APHP
- Société de Réanimation de Langue Française (FR)
- CARDIF – L’Assistance Respiratoire
Collapse
Affiliation(s)
- Matthieu Le Dinh
- Sorbonnes Universités UPMC Univ Paris 06, UMRS INSERM 1158, Paris, France
| | - Serge Carreira
- Department of Anesthesiology and Critical Care Groupe hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
- Sorbonnes Universités UPMC Univ Paris 06, UMRS INSERM 1166, IHU ICAN, Paris, France
| | - Julie Obert
- Sorbonnes Universités UPMC Univ Paris 06, UMRS INSERM 974, Institut de Myologie, Paris, France
| | - Ghislaine Gayan-Ramirez
- Respiratory Muscle Research Unit, Laboratory of Pneumology and Respiratory Division, Katholieke Universiteit, Leuven, Belgium
| | - Bruno Riou
- Sorbonnes Universités UPMC Univ Paris 06, UMRS INSERM 1166, IHU ICAN, Paris, France
- Department of Emergency Medicine and Surgery Groupe hospitalier Pitié-Salpêtrière, APHP, Paris, France
| | - Maud Beuvin
- Sorbonnes Universités UPMC Univ Paris 06, UMRS INSERM 974, Institut de Myologie, Paris, France
| | - Thomas Similowski
- Sorbonnes Universités UPMC Univ Paris 06, UMRS INSERM 1158, Paris, France
- Department of Pneumology and Medical Intensive Care Groupe hospitalier Pitié-Salpêtrière, APHP, Paris France
| | - Catherine Coirault
- Sorbonnes Universités UPMC Univ Paris 06, UMRS INSERM 974, Institut de Myologie, Paris, France
| | - Alexandre Demoule
- Sorbonnes Universités UPMC Univ Paris 06, UMRS INSERM 1158, Paris, France
- Department of Pneumology and Medical Intensive Care Groupe hospitalier Pitié-Salpêtrière, APHP, Paris France
- * E-mail:
| |
Collapse
|
18
|
Komatsu R, Okazaki T, Ebihara S, Kobayashi M, Tsukita Y, Nihei M, Sugiura H, Niu K, Ebihara T, Ichinose M. Aspiration pneumonia induces muscle atrophy in the respiratory, skeletal, and swallowing systems. J Cachexia Sarcopenia Muscle 2018; 9:643-653. [PMID: 29790300 PMCID: PMC6104110 DOI: 10.1002/jcsm.12297] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/31/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Repetition of the onset of aspiration pneumonia in aged patients is common and causes chronic inflammation. The inflammation induces proinflammatory cytokine production and atrophy in the muscles. The proinflammatory cytokines induce muscle proteolysis by activating calpains and caspase-3, followed by further degradation by the ubiquitin-proteasome system. Autophagy is another pathway of muscle atrophy. However, little is known about the relationship between aspiration pneumonia and muscle. For swallowing muscles, it is not clear whether they produce cytokines. The main objective of this study was to determine whether aspiration pneumonia induces muscle atrophy in the respiratory (the diaphragm), skeletal (the tibialis anterior, TA), and swallowing (the tongue) systems, and their possible mechanisms. METHODS We employed a mouse aspiration pneumonia model and computed tomography (CT) scans of aged pneumonia patients. To induce aspiration pneumonia, mice were inoculated with low dose pepsin and lipopolysaccharide solution intra-nasally 5 days a week. The diaphragm, TA, and tongue were isolated, and total RNA, proteins, and frozen sections were stored. Quantitative real-time polymerase chain reaction determined the expression levels of proinflammatory cytokines, muscle E3 ubiquitin ligases, and autophagy related genes. Western blot analysis determined the activation of the muscle proteolysis pathway. Frozen sections determined the presence of muscle atrophy. CT scans were used to evaluate the muscle atrophy in aged aspiration pneumonia patients. RESULTS The aspiration challenge enhanced the expression levels of proinflammatory cytokines in the diaphragm, TA, and tongue. Among muscle proteolysis pathways, the aspiration challenge activated caspase-3 in all the three muscles examined, whereas calpains were activated in the diaphragm and the TA but not in the tongue. Activation of the ubiquitin-proteasome system was detected in all the three muscles examined. The aspiration challenge activated autophagy in the TA and the tongue, whereas weak or little activation was detected in the diaphragm. The aspiration challenge resulted in a greater proportion of smaller myofibers than in controls in the diaphragm, TA, and tongue, suggesting muscle atrophy. CT scans clearly showed that aspiration pneumonia was followed by muscle atrophy in aged patients. CONCLUSIONS Aspiration pneumonia induced muscle atrophy in the respiratory, skeletal, and swallowing systems in a preclinical animal model and in human patients. Diaphragmatic atrophy may weaken the force of cough to expectorate sputum or mis-swallowed contents. Skeletal muscle atrophy may cause secondary sarcopenia. The atrophy of swallowing muscles may weaken the swallowing function. Thus, muscle atrophy could become a new therapeutic target of aspiration pneumonia.
Collapse
Affiliation(s)
- Riyo Komatsu
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuma Okazaki
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoru Ebihara
- Department of Rehabilitation Medicine, Toho University Graduate School of Medicine, Tokyo, Japan
| | - Makoto Kobayashi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoko Tsukita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mayumi Nihei
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kaijun Niu
- Department of Nutrition and Food Science, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
| | - Takae Ebihara
- Department of Geriatric Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | - Masakazu Ichinose
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
19
|
Regulation of Akt-mTOR, ubiquitin-proteasome and autophagy-lysosome pathways in locomotor and respiratory muscles during experimental sepsis in mice. Sci Rep 2017; 7:10866. [PMID: 28883493 PMCID: PMC5589872 DOI: 10.1038/s41598-017-11440-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/24/2017] [Indexed: 11/17/2022] Open
Abstract
Sepsis induced loss of muscle mass and function contributes to promote physical inactivity and disability in patients. In this experimental study, mice were sacrificed 1, 4, or 7 days after cecal ligation and puncture (CLP) or sham surgery. When compared with diaphragm, locomotor muscles were more prone to sepsis-induced muscle mass loss. This could be attributed to a greater activation of ubiquitin-proteasome system and an increased myostatin expression. Thus, this study strongly suggests that the contractile activity pattern of diaphragm muscle confers resistance to atrophy compared to the locomotor gastrocnemius muscle. These data also suggest that a strategy aimed at preventing the activation of catabolic pathways and preserving spontaneous activity would be of interest for the treatment of patients with sepsis-induced neuromyopathy.
Collapse
|
20
|
Demoule A, Molinari N, Jung B, Prodanovic H, Chanques G, Matecki S, Mayaux J, Similowski T, Jaber S. Patterns of diaphragm function in critically ill patients receiving prolonged mechanical ventilation: a prospective longitudinal study. Ann Intensive Care 2016; 6:75. [PMID: 27492005 PMCID: PMC4974210 DOI: 10.1186/s13613-016-0179-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 07/18/2016] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND In intensive care unit (ICU) patients, diaphragmatic dysfunction (DD) can occur on admission or during the subsequent stay. The respective incidence of these two phenomena has not been previously studied in humans. The study was designed to describe temporal trends in diaphragm function in mechanically ventilated (MV) patients. METHODS Ancillary study of a prospective, 6-month, observational cohort study conducted in two ICUs. MV patients were studied within 24 h following intubation (day-1) and every 48-72 h thereafter. Diaphragm function was assessed by twitch tracheal pressure (Ptr,stim) in response to bilateral anterior magnetic phrenic nerve stimulation. Diaphragm dysfunction was defined as Ptr,stim < 11 cmH2O. Patients who received MV for at least 5 days were retained, and the first and the last measures were analysed. RESULTS Forty-three patients were included. Overall, 79 % of patients developed DD at some point during their ICU stay: 23 (53 %) patients presented DD on initiation of mechanical ventilation, 14 (33 %) of whom had persistent DD, while diaphragm function improved in 9 (21 %). Among the remaining 20 (47 %) patients who did not present DD on initiation of MV, 11 (26 %) developed DD during the ICU stay, while 9 (21 %) did not. Mortality was higher in patients with DD either on initiation of mechanical ventilation or during the subsequent ICU stay than in those who never developed DD (35 vs. 0 %, p = 0.04). Duration of MV was higher in patients with DD on initiation of MV that subsequently persisted than in patients who never exhibited diaphragm dysfunction (18 vs. 5 days, p = 0.04). Factors associated with a change in Ptr,stim were: age [linear coefficient regression (Coeff.) -0.097, standard error (SD) 0.047, p = 0.046], PaO2/FiO2 ratio (Coeff. 0.014, SD 0.006, p = 0.0211) and the proportion of the time under MV with sedation (per 10 %, Coeff. -5.359, SD 2.451, p = 0.035). CONCLUSIONS DD is observed in a large majority of MV patients ≥5 days at some point of their ICU stay. Various patterns of DD are observed, including DD on initiation of mechanical ventilation and ICU-acquired DD. Trial registration clinicaltrials.gov Identifier # NCT00786526.
Collapse
Affiliation(s)
- Alexandre Demoule
- INSERM, UMRS1158 Neurophysiologie respiratoire expérimentale et clinique, Sorbonne Universités, UPMC Univ Paris 06, Paris, France
- Intensive Care Unit and Respiratory Division (Département “R3S”), Groupe Hospitalier Pitié-Salpêtrière Charles Foix, AP-HP, 75013 Paris, France
- Service de Pneumologie et Réanimation Médicale, Groupe Hospitalier Pitié-Salpêtrière, 47-83 boulevard de l’Hôpital, 75651 Paris Cedex 13, France
| | - Nicolas Molinari
- Department of Medical Information, Hôpital Arnaud de Villeneuve, IMAG U5149, University of Montpellier, Montpellier, France
| | - Boris Jung
- INSERM U1046, CNRS UMR 9214, Montpellier School of Medicine, University of Montpellier, Montpellier, France
- Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| | - Hélène Prodanovic
- Intensive Care Unit and Respiratory Division (Département “R3S”), Groupe Hospitalier Pitié-Salpêtrière Charles Foix, AP-HP, 75013 Paris, France
| | - Gerald Chanques
- INSERM U1046, CNRS UMR 9214, Montpellier School of Medicine, University of Montpellier, Montpellier, France
- Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| | - Stefan Matecki
- Physiology and Experimental Medecine, Heart-Muscle UMR CNRS 9214 – INSERM U1046, Montpellier University, Montpellier, France
| | - Julien Mayaux
- Intensive Care Unit and Respiratory Division (Département “R3S”), Groupe Hospitalier Pitié-Salpêtrière Charles Foix, AP-HP, 75013 Paris, France
| | - Thomas Similowski
- INSERM, UMRS1158 Neurophysiologie respiratoire expérimentale et clinique, Sorbonne Universités, UPMC Univ Paris 06, Paris, France
- Intensive Care Unit and Respiratory Division (Département “R3S”), Groupe Hospitalier Pitié-Salpêtrière Charles Foix, AP-HP, 75013 Paris, France
| | - Samir Jaber
- INSERM U1046, CNRS UMR 9214, Montpellier School of Medicine, University of Montpellier, Montpellier, France
- Intensive Care and Anesthesiology Department, Saint Eloi Hospital, Montpellier, France
| |
Collapse
|
21
|
Kim MJ, Yoo YC, Sung NY, Lee J, Park SR, Shon EJ, Lee BD, Kim MR. Anti-Inflammatory Effects of Liriope platyphylla in LPS-Stimulated Macrophages and Endotoxemic Mice. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:1127-1143. [DOI: 10.1142/s0192415x16500634] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In the present study, the anti-inflammatory and antisepticemic activities of a water extract of Liriope platyphylla (LP) were investigated. We first estimated the scavenging activity of DPPH and the hydroxyl radical and total phenolic contents of LP. Results indicated that LP, a rich source of phenolic compounds, showed a remarkable radical scavenging capacity. A MTT assay showed that LP treatment did not affect the toxicity against the RAW 264.7 macrophage cells, up to the concentration of 500[Formula: see text][Formula: see text]g/mL. Treatment of LP significantly attenuated the production of inflammatory mediators, such as nitric oxide (NO), interleukin-6 (IL-6), tumor-necrosis factor (TNF)-[Formula: see text] and prostaglandin (PG)E2 in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages cells. Moreover, LP contributed to the down-regulation of inducible NO synthase (iNOS) and TNF-[Formula: see text] mRNA expression, as well as cyclooxygenase-2 (COX-2) protein expression. A western blotting assay further showed that LP inhibited activation of mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-[Formula: see text]B. In an animal experiment using an LPS-induced septicemia model in C57BL/6 mice, oral administration of LP (40[Formula: see text]mg/kg body weight) markedly reduced the level of TNF-[Formula: see text] and IL-6 in serum and protected against LPS-induced lethal shock in mice. Taken together, the results of treatments of LP on inhibited LPS-induced inflammatory responses in both in vitro and in vivo models and indicate it may be a promising neutraceutical or medicinal agent to prevent or cure inflammation-related disease.
Collapse
Affiliation(s)
- Min-Jee Kim
- Department of Food and Nutrition, Chungnam National University, Daejeon 305-764, Korea
| | - Yung-Choon Yoo
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 302-632, Korea
| | - Nak-Yun Sung
- Department of Food and Nutrition, Chungnam National University, Daejeon 305-764, Korea
| | - Julim Lee
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 302-632, Korea
| | - Seok-Rae Park
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 302-632, Korea
| | - Eun-Jung Shon
- Department of Food and Nutrition, Chungnam National University, Daejeon 305-764, Korea
| | - Bo Dam Lee
- Department of Food and Nutrition, Chungnam National University, Daejeon 305-764, Korea
| | - Mee Ree Kim
- Department of Food and Nutrition, Chungnam National University, Daejeon 305-764, Korea
| |
Collapse
|
22
|
Endotoxemia accelerates diaphragm dysfunction in ventilated rabbits. J Surg Res 2016; 206:507-516. [PMID: 27884349 DOI: 10.1016/j.jss.2016.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/13/2016] [Accepted: 08/04/2016] [Indexed: 01/30/2023]
Abstract
BACKGROUND Ventilators may induce diaphragm dysfunction, and most of the septic population who are admitted to the intensive care unit require mechanical ventilation. However, there is no evidence that sepsis accelerates the onset of ventilator-induced diaphragm dysfunction or affects the microcirculation. Our study investigated whether lipopolysaccharide (LPS)-induced endotoxemia accelerated diaphragm dysfunction in ventilated rabbits by evaluating microcirculation, lipid accumulation, and diaphragm contractility. METHODS After anesthesia and tracheostomy, 25 invasively monitored and mechanically ventilated New Zealand white rabbits were randomized to control (n = 5), controlled mechanical ventilation (CMV) (n = 5), pressure support ventilation (PSV; n = 5), CMV or PSV with LPS-induced endotoxemia (CMV-LPS and PSV-LPS, respectively; n = 5 for each). Rabbits were anesthetized and ventilated for 24 h, except the control rabbits (30 min). Diaphragmatic contractility was evaluated using neuromechanical and neuroventilatory efficiency. We evaluated the following at the end of the protocol: (1) diaphragm microcirculation; (2) lipid accumulation; and (3) diaphragm muscular fibers structure. RESULTS Diaphragm contractility, microcirculation, lipid accumulation, and fiber structures were severely compromised in endotoxemic animals after 24 h compared to nonendotoxemic rabbits. Moreover, a slight but significant increase in lipid accumulation was observed in CMV and PSV groups compared with controls (P < 0.05). CONCLUSIONS Endotoxemia accelerates the diaphragm dysfunction process in ventilated rabbits, affects the microcirculation, and results in diaphragmatic lipid accumulation and contractility impairment.
Collapse
|
23
|
Wu J, Li ST. Dexmedetomidine May Produce Extra Protective Effects on Sepsis-induced Diaphragm Injury. Chin Med J (Engl) 2016; 128:1407-11. [PMID: 25963365 PMCID: PMC4830324 DOI: 10.4103/0366-6999.156808] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE The objective was to evaluate the protective effects of dexmedetomidine (DEX), a selective agonist of α2-adrenergic receptor, on sepsis-induced diaphragm injury and the underlying molecular mechanisms. DATA SOURCES The data used in this review were mainly from PubMed articles published in English from 1990 to 2015. STUDY SELECTION Clinical or basic research articles were selected mainly according to their level of relevance to this topic. RESULTS Sepsis could induce severe diaphragm dysfunction and exacerbate respiratory weakness. The mechanism of sepsis-induced diaphragm injury includes the increased inflammatory cytokines and excessive oxidative stress and superfluous production of nitric oxide (NO). DEX can reduce inflammatory cytokines, inhibit nuclear factor-kappaB signaling pathways, suppress the activation of caspase-3, furthermore decrease oxidative stress and inhibit NO synthase. On the basis of these mechanisms, DEX may result in a shorter period of mechanical ventilation in septic patients in clinical practice. CONCLUSIONS Based on this current available evidence, DEX may produce extra protective effects on sepsis-induced diaphragm injury. Further direct evidence and more specific studies are still required to confirm these beneficial effects.
Collapse
Affiliation(s)
| | - Shi-Tong Li
- Department of Anesthesiology, First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China
| |
Collapse
|
24
|
O'Halloran KD. Piling on the pressure to combat acute respiratory distress syndrome: a PEEP into the future? Exp Physiol 2015; 100:879-80. [DOI: 10.1113/ep085363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 11/08/2022]
|
25
|
Schellekens WJM, van Hees HWH, Linkels M, Dekhuijzen PNR, Scheffer GJ, van der Hoeven JG, Heunks LMA. Levosimendan affects oxidative and inflammatory pathways in the diaphragm of ventilated endotoxemic mice. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:69. [PMID: 25888356 PMCID: PMC4355991 DOI: 10.1186/s13054-015-0798-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 02/11/2015] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Controlled mechanical ventilation and endotoxemia are associated with diaphragm muscle atrophy and dysfunction. Oxidative stress and activation of inflammatory pathways are involved in the pathogenesis of diaphragmatic dysfunction. Levosimendan, a cardiac inotrope, has been reported to possess anti-oxidative and anti-inflammatory properties. The aim of the present study was to investigate the effects of levosimendan on markers for diaphragm nitrosative and oxidative stress, inflammation and proteolysis in a mouse model of endotoxemia and mechanical ventilation. METHODS Three groups were studied: (1) unventilated mice (CON, n =8), (2) mechanically ventilated endotoxemic mice (MV LPS, n =17) and (3) mechanically ventilated endotoxemic mice treated with levosimendan (MV LPS + L, n =17). Immediately after anesthesia (CON) or after 8 hours of mechanical ventilation, blood and diaphragm muscle were harvested for biochemical analysis. RESULTS Mechanical ventilation and endotoxemia increased expression of inducible nitric oxide synthase (iNOS) mRNA and cytokine levels of interleukin (IL)-1β, IL-6 and keratinocyte-derived chemokine, and decreased IL-10, in the diaphragm; however, they had no effect on protein nitrosylation and 4-hydroxy-2-nonenal protein concentrations. Levosimendan decreased nitrosylated proteins by 10% (P <0.05) and 4-hydroxy-2-nonenal protein concentrations by 13% (P <0.05), but it augmented the rise of iNOS mRNA by 47% (P <0.05). Levosimendan did not affect the inflammatory response in the diaphragm induced by mechanical ventilation and endotoxemia. CONCLUSIONS Mechanical ventilation in combination with endotoxemia results in systemic and diaphragmatic inflammation. Levosimendan partly decreased markers of nitrosative and oxidative stress, but did not affect the inflammatory response.
Collapse
Affiliation(s)
- Willem-Jan M Schellekens
- Department of Anesthesiology, Radboud University Medical Centre, Postbox 9101, Nijmegen, 6500 HB, the Netherlands.
| | - Hieronymus W H van Hees
- Department of Pulmonary Diseases, Radboud University Medical Centre, Postbox 9101, Nijmegen, 6500 HB, the Netherlands.
| | - Marianne Linkels
- Department of Pulmonary Diseases, Radboud University Medical Centre, Postbox 9101, Nijmegen, 6500 HB, the Netherlands.
| | - P N Richard Dekhuijzen
- Department of Pulmonary Diseases, Radboud University Medical Centre, Postbox 9101, Nijmegen, 6500 HB, the Netherlands.
| | - Gert Jan Scheffer
- Department of Anesthesiology, Radboud University Medical Centre, Postbox 9101, Nijmegen, 6500 HB, the Netherlands.
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud University Medical Centre, Postbox 9101, Nijmegen, 6500 HB, the Netherlands.
| | - Leo M A Heunks
- Department of Intensive Care Medicine, Radboud University Medical Centre, Postbox 9101, Nijmegen, 6500 HB, the Netherlands.
| |
Collapse
|
26
|
Jiang J, Yang B, Han G, Yang M, Li S. Early administration of cisatracurium attenuates sepsis-induced diaphragm dysfunction in rats. Inflammation 2015; 38:305-11. [PMID: 25266883 DOI: 10.1007/s10753-014-0034-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sepsis can often induce diaphragm dysfunction, which is associated with localized elaboration of cytokines within the diaphragm. The administration of cisatracurium has been shown to decrease the inflammatory response and to facilitate mechanical ventilation. In this study, we explored whether cisatracurium could attenuate sepsis-induced diaphragm dysfunction in rats. Animals were divided into three groups: (1) the control group: rats underwent a sham surgical procedure with cecal exposure, but the cecum was neither ligated nor punctured; (2) the CLP group: rats underwent cecal ligation and puncture (CLP) and received a continuous infusion of NaCl 0.9 %; and (3) the Cis + CLP group: rats underwent CLP and received a continuous infusion of cisatracurium. After the surgical procedure, all animals underwent controlled mechanical ventilation for 18 h. Plasma concentrations of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and high-mobility group box 1 (HMGB1) were measured using an enzyme-linked immunosorbent assay. Upon completion of the experimental protocol, diaphragm contractility and HMGB1 protein expression were analyzed. Impaired diaphragm contractile function, including both force-related properties and force-frequency responses, was pronounced after CLP in comparison with that observed in the control rats. Furthermore, CLP elevated serum levels of IL-6, TNF-α, and HMGB1, and induced HMGB1 protein expression in the diaphragm. In contrast, cisatracurium counteracted the sepsis-induced inflammation reaction in the diaphragm and serum and maintained diaphragm function. These data suggest that early infusion of cisatracurium attenuates sepsis-induced diaphragm dysfunction; this may be attributable to its anti-inflammatory action.
Collapse
Affiliation(s)
- Jihong Jiang
- Department of Anesthesiology, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, 100 Haining Road, Shanghai, 200080, People's Republic of China,
| | | | | | | | | |
Collapse
|
27
|
Kim MJ, Yoo YC, Kim HJ, Shin SK, Sohn EJ, Min AY, Sung NY, Kim MR. Aged Black Garlic Exerts Anti-Inflammatory Effects by Decreasing NO and Proinflammatory Cytokine Production with Less Cytoxicity in LPS-Stimulated RAW 264.7 Macrophages and LPS-Induced Septicemia Mice. J Med Food 2014; 17:1057-63. [DOI: 10.1089/jmf.2013.3043] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Min Jee Kim
- Department of Food and Nutrition, Chungnam National University, Yuseong-Gu, Daejeon, Korea
| | - Yung Choon Yoo
- Department of Microbiology, College of Medicine, Konyang University, Seo-Ku, Daejeon, Korea
| | - Hyun Jung Kim
- Department of Food and Nutrition, Chungnam National University, Yuseong-Gu, Daejeon, Korea
| | - Suk Kyung Shin
- Department of Food and Nutrition, Chungnam National University, Yuseong-Gu, Daejeon, Korea
| | - Eun Jeong Sohn
- Department of Food and Nutrition, Chungnam National University, Yuseong-Gu, Daejeon, Korea
| | - A Young Min
- Department of Food and Nutrition, Chungnam National University, Yuseong-Gu, Daejeon, Korea
| | - Nak Yun Sung
- Department of Microbiology, College of Medicine, Konyang University, Seo-Ku, Daejeon, Korea
| | - Mee Ree Kim
- Department of Food and Nutrition, Chungnam National University, Yuseong-Gu, Daejeon, Korea
| |
Collapse
|
28
|
Kwak JH, He Y, Yoon B, Koo S, Yang Z, Kang EJ, Lee BH, Han SY, Yoo YC, Lee KB, Kim JS. Synthesis of rhodamine-labelled dieckol: its unique intracellular localization and potent anti-inflammatory activity. Chem Commun (Camb) 2014; 50:13045-8. [DOI: 10.1039/c4cc04270k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Muscle-Specific Inhibition of the Classical Nuclear Factor-κB Pathway Is Protective Against Diaphragmatic Weakness in Murine Endotoxemia. Crit Care Med 2014; 42:e501-9. [DOI: 10.1097/ccm.0000000000000407] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
30
|
Abstract
Abstract
Background:
Diaphragm and psoas are affected during sepsis in animal models. Whether diaphragm or limb muscle is preferentially affected during sepsis in the critically ill remains unclear.
Methods:
Retrospective secondary analysis study including 40 patients, comparing control (n = 17) and critically ill patients, with (n = 14) or without sepsis (n = 9). Diaphragm volume, psoas volume, and cross-sectional area of the skeletal muscles at the third lumbar vertebra were measured during intensive care unit (ICU) stay using tridimensional computed tomography scan volumetry. Diaphragm strength was evaluated using magnetic phrenic nerve stimulation. The primary endpoint was the comparison between diaphragm and peripheral muscle volume kinetics during the ICU stay among critically ill patients, with or without sepsis.
Results:
Upon ICU admission, neither diaphragm nor psoas muscle volumes were significantly different between critically ill and control patients (163 ± 53 cm3vs. 197 ± 82 cm3 for the diaphragm, P = 0.36, and 272 ± 116 cm3vs. to 329 ± 166 cm3 for the psoas, P = 0.31). Twenty-five (15 to 36) days after admission, diaphragm volume decreased by 11 ± 13% in nonseptic and by 27 ± 12% in septic patients, P = 0.01. Psoas volume decreased by 11 ± 10% in nonseptic and by 19 ± 13% in septic patients, P = 0.09. Upon ICU admission, diaphragm strength was correlated with diaphragm volume and was lower in septic (6.2 cm H2O [5.6 to 9.3]) than that in nonseptic patients (13.2 cm H2O [12.3 to 15.6]), P = 0.01.
Conclusions:
During the ICU stay, both diaphragm and psoas volumes decreased. In septic patients, the authors report for the first time in humans preferential diaphragm atrophy compared with peripheral muscles.
Collapse
|
31
|
Abstract
BACKGROUND Acute diaphragmatic dysfunction has been reported in septic and cardiogenic shock, but few data are available concerning the effect of hemorrhagic shock on diaphragmatic function. The authors examined the impact of a hemorrhagic shock on the diaphragm. METHODS Four parallel groups of adult rats were submitted to hemorrhagic shock induced by controlled exsanguination targeting a mean arterial blood pressure of 30 mmHg for 1 h, followed by a 1-h fluid resuscitation with either saline or shed blood targeting a mean arterial blood pressure of 80 mmHg. Diaphragm and soleus strip contractility was measured in vitro. Blood flow in the muscle microcirculation was measured in vivo using a Laser Doppler technique. Muscle proinflammatory cytokine concentrations were also measured. RESULTS Hemorrhagic shock was characterized by a decrease in mean arterial blood pressure to 34 ± 5 mmHg (-77 ± 4%; P< 0.05) and high plasma lactate levels (7.6 ± 0.9 mM; P < 0.05). Although tetanic tension of the diaphragm was not altered, hemorrhagic shock induced dramatic impairment of tetanic tension of the soleus (-40 ± 19%; P < 0.01), whereas proinflammatory cytokine levels were low and not different between the two muscles. Resuscitation with either blood or saline did not further modify either diaphragm or soleus performance and proinflammatory cytokine levels. The shock-induced decrease in blood flow was much more pronounced in the soleus than in the diaphragm (-75 ± 13% vs. -17 ± 10%; P = 0.02), and a significant interaction was observed between shock and muscle (P < 0.001). CONCLUSION Diaphragm performance is preserved during hemorrhagic shock, whereas soleus performance is impaired, with no further impact of either blood or saline fluid resuscitation.
Collapse
|
32
|
Demoule A, Jung B, Prodanovic H, Molinari N, Chanques G, Coirault C, Matecki S, Duguet A, Similowski T, Jaber S. Diaphragm dysfunction on admission to the intensive care unit. Prevalence, risk factors, and prognostic impact-a prospective study. Am J Respir Crit Care Med 2013; 188:213-9. [PMID: 23641946 DOI: 10.1164/rccm.201209-1668oc] [Citation(s) in RCA: 267] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Diaphragmatic insults occurring during intensive care unit (ICU) stays have become the focus of intense research. However, diaphragmatic abnormalities at the initial phase of critical illness remain poorly documented in humans. OBJECTIVES To determine the incidence, risk factors, and prognostic impact of diaphragmatic impairment on ICU admission. METHODS Prospective, 6-month, observational cohort study in two ICUs. Mechanically ventilated patients were studied within 24 hours after intubation (Day 1) and 48 hours later (Day 3). Seventeen anesthetized intubated control anesthesia patients were also studied. The diaphragm was assessed by twitch tracheal pressure in response to bilateral anterior magnetic phrenic nerve stimulation (Ptr,stim). MEASUREMENTS AND MAIN RESULTS Eighty-five consecutive patients aged 62 (54-75) (median [interquartile range]) were evaluated (medical admission, 79%; Simplified Acute Physiology Score II, 54 [44-68]). On Day 1, Ptr,stim was 8.2 (5.9-12.3) cm H2O and 64% of patients had Ptr,stim less than 11 cm H2O. Independent predictors of low Ptr,stim were sepsis (linear regression coefficient, -3.74; standard error, 1.16; P = 0.002) and Simplified Acute Physiology Score II (linear regression coefficient, -0.07; standard error, 1.69; P = 0.03). Compared with nonsurvivors, ICU survivors had higher Ptr,stim (9.7 [6.3-13.8] vs. 7.3 [5.5-9.7] cm H2O; P = 0.004). This was also true for hospital survivors versus nonsurvivors (9.7 [6.3-13.5] vs. 7.8 [5.5-10.1] cm H2O; P = 0.004). Day 1 and Day 3 Ptr,stim were similar. CONCLUSIONS A reduced capacity of the diaphragm to produce inspiratory pressure (diaphragm dysfunction) is frequent on ICU admission. It is associated with sepsis and disease severity, suggesting that it may represent another form of organ failure. It is associated with a poor prognosis. Clinical trial registered with www.clinicaltrials.gov (NCT 00786526).
Collapse
|
33
|
Pinniger GJ, Lavin T, Bakker AJ. Skeletal muscle weakness caused by carrageenan-induced inflammation. Muscle Nerve 2012; 46:413-20. [PMID: 22907233 DOI: 10.1002/mus.23318] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The skeletal muscle weakness associated with many chronic diseases has been attributed to the catabolic effect of pro-inflammatory cytokines. We aimed to determine if local muscle inflammation has direct affects on contractile function and contributes to muscle weakness independent of muscle atrophy or mechanical injury. METHODS Local muscle inflammation was induced by injecting an algal-derived polysaccharide, carrageenan (10 mg/kg), into the right tibialis anterior muscle in healthy ARC mice. The contralateral muscle was injected with sterile isotonic saline, and the muscles were removed after 24 h for measurement of contractile function and cytokine concentration. RESULTS Carrageenan significantly reduced maximum specific force, decreased the maximum rate of force development, altered the force-frequency relationship, and increased intramuscular levels of pro-inflammatory cytokines and chemokines. CONCLUSIONS These results indicate that carrageenan directly affects contractile function and causes skeletal muscle weakness. Local muscle inflammation may contribute to the weakness observed in inflammatory related disorders.
Collapse
Affiliation(s)
- Gavin J Pinniger
- School of Biomedical, Biomolecular and Chemical Sciences, The University of Western Australia, Crawley, Western Australia, 6009 Australia.
| | | | | |
Collapse
|
34
|
Alamdari N, Toraldo G, Aversa Z, Smith I, Castillero E, Renaud G, Qaisar R, Larsson L, Jasuja R, Hasselgren PO. Loss of muscle strength during sepsis is in part regulated by glucocorticoids and is associated with reduced muscle fiber stiffness. Am J Physiol Regul Integr Comp Physiol 2012; 303:R1090-9. [PMID: 23019215 DOI: 10.1152/ajpregu.00636.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Sepsis is associated with impaired muscle function but the role of glucocorticoids in sepsis-induced muscle weakness is not known. We tested the role of glucocorticoids in sepsis-induced muscle weakness by treating septic rats with the glucocorticoid receptor antagonist RU38486. In addition, normal rats were treated with dexamethasone to further examine the role of glucocorticoids in the regulation of muscle strength. Sepsis was induced in rats by cecal ligation and puncture, and muscle force generation (peak twitch and tetanic tension) was determined in lower extremity muscles. In other experiments, absolute and specific force as well as stiffness (reflecting the function of actomyosin cross bridges) were determined in isolated skinned muscle fibers from control and septic rats. Sepsis and treatment with dexamethasone resulted in reduced maximal twitch and tetanic force in intact isolated extensor digitorum longus muscles. The absolute and specific maximal force in isolated muscle fibers was reduced during sepsis together with decreased fiber stiffness. These effects of sepsis were blunted (but not abolished) by RU38486. The results suggest that muscle weakness during sepsis is at least in part regulated by glucocorticoids and reflects loss of contractility at the cellular (individual muscle fiber) level. In addition, the results suggest that reduced function of the cross bridges between actin and myosin (documented as reduced muscle fiber stiffness) may be involved in sepsis-induced muscle weakness. An increased understanding of mechanisms involved in loss of muscle strength will be important for the development of new treatment strategies in patients with this debilitating consequence of sepsis.
Collapse
Affiliation(s)
- Nima Alamdari
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Haegens A, Schols AM, Gorissen SH, van Essen AL, Snepvangers F, Gray DA, Shoelson SE, Langen RC. NF-κB activation and polyubiquitin conjugation are required for pulmonary inflammation-induced diaphragm atrophy. Am J Physiol Lung Cell Mol Physiol 2011; 302:L103-10. [PMID: 22003096 DOI: 10.1152/ajplung.00084.2011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Loss of diaphragm muscle strength in inflammatory lung disease contributes to mortality and is associated with diaphragm fiber atrophy. Ubiquitin (Ub) 26S-proteasome system (UPS)-dependent protein breakdown, which mediates muscle atrophy in a number of physiological and pathological conditions, is elevated in diaphragm muscle of patients with chronic obstructive pulmonary disease. Nuclear factor kappa B (NF-κB), an essential regulator of many inflammatory processes, has been implicated in the regulation of poly-Ub conjugation of muscle proteins targeted for proteolysis by the UPS. Here, we test if NF-κB activation in diaphragm muscle and subsequent protein degradation by the UPS are required for pulmonary inflammation-induced diaphragm atrophy. Acute pulmonary inflammation was induced in mice by intratracheal lipopolysaccharide instillation. Fiber cross-sectional area, ex vivo tyrosine release, protein poly-Ub conjugation, and inflammatory signaling were determined in diaphragm muscle. The contribution of NF-κB or the UPS to diaphragm atrophy was assessed in mice with intact or genetically repressed NF-κB signaling or attenuated poly-Ub conjugation, respectively. Acute pulmonary inflammation resulted in diaphragm atrophy measured by reduced muscle fiber cross-sectional area. This was accompanied by diaphragm NF-κB activation, and proteolysis, measured by tyrosine release from the diaphragm. Poly-Ub conjugation was increased in diaphragm, as was the expression of muscle-specific E3 Ub ligases. Genetic suppression of poly-Ub conjugation prevented inflammation-induced diaphragm muscle atrophy, as did muscle-specific inhibition of NF-κB signaling. In conclusion, the present study is the first to demonstrate that diaphragm muscle atrophy, resulting from acute pulmonary inflammation, requires NF-κB activation and UPS-mediated protein degradation.
Collapse
Affiliation(s)
- Astrid Haegens
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
36
|
van Hees HWH, Schellekens WJM, Linkels M, Leenders F, Zoll J, Donders R, Dekhuijzen PNR, van der Hoeven JG, Heunks LMA. Plasma from septic shock patients induces loss of muscle protein. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:R233. [PMID: 21958504 PMCID: PMC3334781 DOI: 10.1186/cc10475] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 08/19/2011] [Accepted: 09/29/2011] [Indexed: 11/13/2022]
Abstract
Introduction ICU-acquired muscle weakness commonly occurs in patients with septic shock and is associated with poor outcome. Although atrophy is known to be involved, it is unclear whether ligands in plasma from these patients are responsible for initiating degradation of muscle proteins. The aim of the present study was to investigate if plasma from septic shock patients induces skeletal muscle atrophy and to examine the time course of plasma-induced muscle atrophy during ICU stay. Methods Plasma was derived from septic shock patients within 24 hours after hospital admission (n = 21) and healthy controls (n = 12). From nine patients with septic shock plasma was additionally derived at two, five and seven days after ICU admission. These plasma samples were added to skeletal myotubes, cultured from murine myoblasts. After incubation for 24 hours, myotubes were harvested and analyzed on myosin content, mRNA expression of E3-ligase and Nuclear Factor Kappa B (NFκB) activity. Plasma samples were analyzed on cytokine concentrations. Results Myosin content was approximately 25% lower in myotubes exposed to plasma from septic shock patients than in myotubes exposed to plasma from controls (P < 0.01). Furthermore, patient plasma increased expression of E3-ligases Muscle RING Finger protein-1 (MuRF-1) and Muscle Atrophy F-box protein (MAFbx) (P < 0.01), enhanced NFκB activity (P < 0.05) and elevated levels of ubiquitinated myosin in myotubes. Myosin loss was significantly associated with elevated plasma levels of interleukin (IL)-6 in septic shock patients (P < 0.001). Addition of antiIL-6 to septic shock plasma diminished the loss of myosin in exposed myotubes by approximately 25% (P < 0.05). Patient plasma obtained later during ICU stay did not significantly reduce myosin content compared to controls. Conclusions Plasma from patients with septic shock induces loss of myosin and activates key regulators of proteolysis in skeletal myotubes. IL-6 is an important player in sepsis-induced muscle atrophy in this model. The potential to induce atrophy is strongest in plasma obtained during the early phase of human sepsis.
Collapse
Affiliation(s)
- Hieronymus W H van Hees
- Department of Pulmonary Diseases, Radboud University Nijmegen Medical Centre, Geert Grooteplein-Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Aversa Z, Alamdari N, Hasselgren PO. Molecules modulating gene transcription during muscle wasting in cancer, sepsis, and other critical illness. Crit Rev Clin Lab Sci 2011; 48:71-86. [DOI: 10.3109/10408363.2011.591365] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Sigala I, Zacharatos P, Toumpanakis D, Michailidou T, Noussia O, Theocharis S, Roussos C, Papapetropoulos A, Vassilakopoulos T. MAPKs and NF-κB differentially regulate cytokine expression in the diaphragm in response to resistive breathing: the role of oxidative stress. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1152-62. [PMID: 21325641 DOI: 10.1152/ajpregu.00376.2010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Inspiratory resistive breathing (IRB) induces cytokine expression in the diaphragm. The mechanism of this cytokine induction remains elusive. The roles of MAPKs and NF-κB and the impact of oxidative stress in IRB-induced cytokine upregulation in the diaphragm were studied. Wistar rats were subjected to IRB (50% of maximal inspiratory pressure) via a two-way nonrebreathing valve for 1, 3, or 6 h. Additional groups of rats subjected to IRB for 6 h were randomly assigned to receive either solvent or N-acetyl-cysteine (NAC) or inhibitors of NF-κB (BAY-11-7082), ERK1/2 (PD98059), and P38 MAPK (SB203580) to study the effect of oxidative stress, NF-κB, and MAPKs in IRB-induced cytokine upregulation in the diaphragm. Quietly breathing animals served as controls. IRB upregulated cytokine (IL-6, TNF-α, IL-10, IL-2, IL-1β) protein levels in the diaphragm and resulted in increased activation of MAPKs (P38, ERK1/2) and NF-κB. Inhibition of NF-κB and ERK1/2 blunted the upregulation of all cytokines except that of IL-6, which was further increased. P38 inhibition attenuated all cytokine (including IL-6) upregulation. Both P38 and ERK1/2 inhibition decreased NF-κB/p65 subunit phosphorylation. NAC pretreatment blunted IRB-induced cytokine upregulation in the diaphragm and resulted in decreased ERK1/2, P38, and NF-κB/p65 phosphorylation. In conclusion, IRB-induced cytokine upregulation in the diaphragm is under the regulatory control of MAPKs and NF-κB. IL-6 is regulated differently from all other cytokines through a P38-dependent and NF-κB independent pathway. Oxidative stress is a stimulus for IRB-induced cytokine upregulation in the diaphragm.
Collapse
Affiliation(s)
- Ioanna Sigala
- Dept. of Critical Care and Pulmonary Services, University of Athens Medical School, Evangelismos Hospital, 45-47 Ipsilandou Str., Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Jaber S, Petrof BJ, Jung B, Chanques G, Berthet JP, Rabuel C, Bouyabrine H, Courouble P, Koechlin-Ramonatxo C, Sebbane M, Similowski T, Scheuermann V, Mebazaa A, Capdevila X, Mornet D, Mercier J, Lacampagne A, Philips A, Matecki S. Rapidly Progressive Diaphragmatic Weakness and Injury during Mechanical Ventilation in Humans. Am J Respir Crit Care Med 2011; 183:364-71. [DOI: 10.1164/rccm.201004-0670oc] [Citation(s) in RCA: 439] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
40
|
Maalouf SW, Talhouk RS, Schanbacher FL. Inflammatory responses in epithelia: endotoxin-induced IL-6 secretion and iNOS/NO production are differentially regulated in mouse mammary epithelial cells. JOURNAL OF INFLAMMATION-LONDON 2010; 7:58. [PMID: 21118556 PMCID: PMC3009620 DOI: 10.1186/1476-9255-7-58] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Accepted: 11/30/2010] [Indexed: 11/26/2022]
Abstract
Background IL-6 is a pro-inflammatory cytokine that signals via binding to a soluble or membrane bound receptor, while nitric oxide (NO), an oxidative stress molecule, diffuses through the cell membrane without a receptor. Both mediators signal through different mechanisms, yet they are dependent on NFκB. We proposed that both mediators are co-induced and co-regulated in inflamed mammary epithelial cells. Methods SCp2 mammary epithelial cells were treated with bacterial endotoxin (ET) for different time periods and analyzed for induction of IL-6 secretion and NO production by ELISA and Griess reaction, respectively. The expression of IL-6 and induced NO synthase (iNOS) was assayed by real time PCR and/or western immunoblots, and the activation of NFκB was assayed by immunobinding assay. To investigate the role of mammary cell microenvironment (cell-substratum or interaction of mammary epithelial cell types; critical to mammary development, function, and disease) in modulation of the inflammatory response, SCp2 cells were cultured with or without extracellular matrix (EHS) or in coculture with their myoepithelial counterpart (SCg6), and assayed for ET-induced IL-6 and NO. Results Endotoxin induced NFκB activation at 1 h after ET application. IL-6 secretion and NO production were induced, but with unexpected delay in expression of mRNA for iNOS compared to IL-6. NFκB/p65 activation was transient but NFκB/p50 activation persisted longer. Selective inhibition of NFκB activation by Wedelolactone reduced ET-induced expression of IL-6 mRNA and protein but not iNOS mRNA or NO production, suggesting differences in IL-6 and iNOS regulation via NFκB. SCp2 cells in coculture with SCg6 but not in presence of EHS dramatically induced IL-6 secretion even in the absence of ET. ET-induced NO production was blunted in SCp2/SCg6 cocultures compared to that in SCp2 alone. Conclusions The differential regulation of IL-6 and iNOS together with the differential activation of different NFκB dimers suggest that IL-6 and iNOS are regulated by different NFκB dimers, and differentially regulated by the microenvironment of epithelial cells. The understanding of innate immune responses and inflammation in epithelia and linkage thereof is crucial for understanding the link between chronic inflammation and cancer in epithelial tissues such as the mammary gland.
Collapse
Affiliation(s)
- Samar W Maalouf
- Department of Animal Sciences, The Ohio State University, OARDC, Wooster, OH, USA.
| | | | | |
Collapse
|
41
|
Labbe K, Danialou G, Gvozdic D, Demoule A, Divangahi M, Boyd JH, Petrof BJ. Inhibition of monocyte chemoattractant protein-1 prevents diaphragmatic inflammation and maintains contractile function during endotoxemia. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2010; 14:R187. [PMID: 20950459 PMCID: PMC3219293 DOI: 10.1186/cc9295] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 08/05/2010] [Accepted: 10/07/2010] [Indexed: 11/10/2022]
Abstract
Introduction Respiratory muscle weakness is common in sepsis patients. Proinflammatory mediators produced during sepsis have been implicated in diaphragmatic contractile dysfunction, but the role of chemokines has not been explored. This study addressed the role of monocyte chemoattractant protein-1 (MCP-1, also known as CCL2), in the pathogenesis of diaphragmatic inflammation and weakness during endotoxemia. Methods Mice were treated as follows (n = 6 per group): (a) saline, (b) endotoxin (25 μg/g IP), (c) endotoxin + anti-MCP-1 antibody, and (d) endotoxin + isotype control antibody. Muscles were also exposed to recombinant MCP-1 in vivo and in vitro. Measurements were made of diaphragmatic force generation, leukocyte infiltration, and proinflammatory mediator (MCP-1, IL-1α, IL-1β, IL-6, NF-κB) expression/activity. Results In vivo, endotoxin-treated mice showed a large decrease in diaphragmatic force, together with upregulation of MCP-1 and other cytokines, but without an increase in intramuscular leukocytes. Antibody neutralization of MCP-1 prevented the endotoxin-induced force loss and reduced expression of MCP-1, IL-1α, IL-1β, and IL-6 in the diaphragm. MCP-1 treatment of nonseptic muscles also led to contractile weakness, and MCP-1 stimulated its own transcription independent of NF-κB activation in vitro. Conclusions These results suggest that MCP-1 plays an important role in the pathogenesis of diaphragmatic weakness during sepsis by both direct and indirect mechanisms. We speculate that its immunomodulatory properties and ability to modify skeletal muscle function make MCP-1 a potential therapeutic target in critically ill patients with sepsis and associated respiratory muscle weakness.
Collapse
Affiliation(s)
- Katherine Labbe
- Meakins-Christie Laboratories, McGill University, 3626 Saint Urbain, Montreal, Quebec, Canada.
| | | | | | | | | | | | | |
Collapse
|
42
|
Inhibition of the IKK/NF-κB pathway by AAV gene transfer improves muscle regeneration in older mdx mice. Gene Ther 2010; 17:1476-83. [PMID: 20720575 DOI: 10.1038/gt.2010.110] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The IκB kinase (IKKα, β and the regulatory subunit IKKγ) complex regulates nuclear factor of κB (NF-κB) transcriptional activity, which is upregulated in many chronic inflammatory diseases. NF-κB signaling promotes inflammation and limits muscle regeneration in Duchenne muscular dystrophy (DMD), resulting in fibrotic and fatty tissue replacement of muscle that exacerbates the wasting process in dystrophic muscles. Here, we examined whether dominant-negative forms of IKKα (IKKα-dn) and IKKβ (IKKβ-dn) delivered by adeno-associated viral (AAV) vectors to the gastrocnemius (GAS) and tibialis anterior (TA) muscles of 1, 2 and 11-month-old mdx mice, a murine DMD model, block NF-κB activation and increase muscle regeneration. At 1 month post-treatment, the levels of nuclear NF-κB in locally treated muscle were decreased by gene transfer with either AAV-CMV-IKKα-dn or AAV-CMV-IKKβ-dn, but not by IKK wild-type controls (IKKα and β) or phosphate-buffered saline (PBS). Although treatment with AAV-IKKα-dn or AAV-IKKβ-dn vectors had no significant effect on muscle regeneration in young mdx mice treated at 1 and 2 months of age and collected 1 month later, treatment of old (11 months) mdx with AAV-CMV-IKKα-dn or AAV-CMV-IKKβ-dn significantly increased levels of muscle regeneration. In addition, there was a significant decrease in myofiber necrosis in the AAV-IKKα-dn- and AAV-IKKβ-dn-treated mdx muscle in both young and old mice. These results demonstrate that inhibition of IKKα or IKKβ in dystrophic muscle reduces the adverse effects of NF-κB signaling, resulting in a therapeutic effect. Moreover, these results clearly demonstrate the therapeutic benefits of inhibiting NF-κB activation by AAV gene transfer in dystrophic muscle to promote regeneration, particularly in older mdx mice, and block necrosis.
Collapse
|
43
|
Iida Y, Yamada S, Nishida O, Nakamura T. Body mass index is negatively correlated with respiratory muscle weakness and interleukin-6 production after coronary artery bypass grafting. J Crit Care 2010; 25:172.e1-8. [DOI: 10.1016/j.jcrc.2009.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 04/09/2009] [Accepted: 05/15/2009] [Indexed: 01/24/2023]
|
44
|
Abstract
Sepsis is a major cause of morbidity and mortality in critically ill patients, and despite advances in management, mortality remains high. In survivors, sepsis increases the risk for the development of persistent acquired weakness syndromes affecting both the respiratory muscles and the limb muscles. This acquired weakness results in prolonged duration of mechanical ventilation, difficulty weaning, functional impairment, exercise limitation, and poor health-related quality of life. Abundant evidence indicates that sepsis induces a myopathy characterized by reductions in muscle force-generating capacity, atrophy (loss of muscle mass), and altered bioenergetics. Sepsis elicits derangements at multiple subcellular sites involved in excitation contraction coupling, such as decreasing membrane excitability, injuring sarcolemmal membranes, altering calcium homeostasis due to effects on the sarcoplasmic reticulum, and disrupting contractile protein interactions. Muscle wasting occurs later and results from increased proteolytic degradation as well as decreased protein synthesis. In addition, sepsis produces marked abnormalities in muscle mitochondrial functional capacity and when severe, these alterations correlate with increased death. The mechanisms leading to sepsis-induced changes in skeletal muscle are linked to excessive localized elaboration of proinflammatory cytokines, marked increases in free-radical generation, and activation of proteolytic pathways that are upstream of the proteasome including caspase and calpain. Emerging data suggest that targeted inhibition of these pathways may alter the evolution and progression of sepsis-induced myopathy and potentially reduce the occurrence of sepsis-mediated acquired weakness syndromes.
Collapse
|
45
|
Lack of CFTR in skeletal muscle predisposes to muscle wasting and diaphragm muscle pump failure in cystic fibrosis mice. PLoS Genet 2009; 5:e1000586. [PMID: 19649303 PMCID: PMC2709446 DOI: 10.1371/journal.pgen.1000586] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Accepted: 07/03/2009] [Indexed: 11/19/2022] Open
Abstract
Cystic fibrosis (CF) patients often have reduced mass and strength of skeletal muscles, including the diaphragm, the primary muscle of respiration. Here we show that lack of the CF transmembrane conductance regulator (CFTR) plays an intrinsic role in skeletal muscle atrophy and dysfunction. In normal murine and human skeletal muscle, CFTR is expressed and co-localized with sarcoplasmic reticulum-associated proteins. CFTR-deficient myotubes exhibit augmented levels of intracellular calcium after KCl-induced depolarization, and exposure to an inflammatory milieu induces excessive NF-kB translocation and cytokine/chemokine gene upregulation. To determine the effects of an inflammatory environment in vivo, sustained pulmonary infection with Pseudomonas aeruginosa was produced, and under these conditions diaphragmatic force-generating capacity is selectively reduced in Cftr(-/-) mice. This is associated with exaggerated pro-inflammatory cytokine expression as well as upregulation of the E3 ubiquitin ligases (MuRF1 and atrogin-1) involved in muscle atrophy. We conclude that an intrinsic alteration of function is linked to the absence of CFTR from skeletal muscle, leading to dysregulated calcium homeostasis, augmented inflammatory/atrophic gene expression signatures, and increased diaphragmatic weakness during pulmonary infection. These findings reveal a previously unrecognized role for CFTR in skeletal muscle function that may have major implications for the pathogenesis of cachexia and respiratory muscle pump failure in CF patients.
Collapse
|
46
|
Lee KH, Yeh MH, Kao ST, Hung CM, Chen BC, Liu CJ, Yeh CC. Xia-bai-san inhibits lipopolysaccharide-induced activation of intercellular adhesion molecule-1 and nuclear factor-kappa B in human lung cells. JOURNAL OF ETHNOPHARMACOLOGY 2009; 124:530-538. [PMID: 19454309 DOI: 10.1016/j.jep.2009.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2008] [Revised: 04/26/2009] [Accepted: 05/10/2009] [Indexed: 05/27/2023]
Abstract
UNLABELLED Xia-bai-san (XBS) is a traditional Chinese medicine that has been used clinically for centuries in Asian countries to treat some types of common cold and asthma-like diseases similar to infantile pneumonia and childhood bronchitis. In previous studies, XBS was found to suppress the inflammatory process induced in lungs of mice treated with lipopolysaccharide (LPS). PURPOSE The present study was undertaken to examine the effects of XBS on LPS-inducible production of inflammatory cytokines, up-regulation of intercellular cell adhesion molecule-1 (ICAM-1), and activation of nuclear factor NF-kappaB in cultured human lung cells. PRINCIPAL RESULTS Extracts of four raw herbs (Cortex mori, Cortex lycii, Radix glycyrrhizae, and Fructus oryzae) were used to prepare the decoction. XBS decreased the histological damage and up-regulation of ICAM-1 observed in lungs of mice treated with lipopolysaccharide (LPS). In cultured human pulmonary epithelial A549 cells, XBS and its components Morus alba and Glycyrrhiza uralensis suppressed the up-regulation of IL-8 and ICAM-1 in response to LPS. Production of TNF-alpha, IL-1beta, IL-6 and IL-8 by LPS-treated human THP-1 monomyelocytes was also suppressed by XBS. A549 cells expressed ICAM-1 in response to medium from LPS-treated THP-1 cells; expression was decreased by XBS. The adhesion of THP-1 cells to LPS-treated A549 cells were inhibited in the presence of XBS. Activation of NF-kappaB by LPS in A549 cells was suppressed by XBS, Morus alba, and Glycyrrhiza uralensis through inhibition of IkappaB phosphorylation; the concentrations at which suppression occurred were identical to those at which production of inflammatory cytokines and up-regulation of ICAM-1 were inhibited. MAJOR CONCLUSIONS These findings support the hypothesis that XBS, Morus alba, and Glycyrrhiza uralensis inhibit the inflammatory process in lung tissue through suppression of the IkappaB signaling pathway. XBS may prove helpful in the management of asthma, various allergic disorders, sepsis, or any other condition associated with pulmonary inflammation.
Collapse
Affiliation(s)
- Kuo-Hua Lee
- Hsin-Chu Branch Station, Council of Agriculture-TLI, Hsin-Chu, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
47
|
Chemokine receptor and ligand upregulation in the diaphragm during endotoxemia and Pseudomonas lung infection. Mediators Inflamm 2009; 2009:860565. [PMID: 19421418 PMCID: PMC2676322 DOI: 10.1155/2009/860565] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Revised: 02/05/2009] [Accepted: 02/09/2009] [Indexed: 11/17/2022] Open
Abstract
Sepsis-induced diaphragmatic inflammation has been associated with
respiratory failure, but the role of chemokines in this process has
not been evaluated. Here we sought to study the local expression and
molecular regulation of the chemokines, regulated upon activation
normal T cell expressed and secreted (RANTES) and macrophage
inflammatory protein (MIP)-1α, in the murine diaphragm during sepsis. Constitutive
expression levels of RANTES and MIP-1α, as well as their receptors, CCR1 and CCR5, were
significantly higher in diaphragm than limb muscle. Sepsis was induced
by acute lipopolysaccharide (LPS) delivery or subacutely by
intratracheal administration of live Pseudomonas aeruginosa bacteria.
Both sepsis models triggered a marked upregulation of RANTES and MIP-1α in the diaphragm. In vitro, stimulation of diaphragmatic
muscle cells with LPS also led to RANTES upregulation. Inhibition of
the NF-kB pathway using pharmacologic or dominant negative genetic
approaches blocked the LPS-induced RANTES upregulation, while free
radical scavengers had no effect. We conclude that sepsis leads to
greatly increased expression of RANTES, MIP-1α and their cognate receptors in the diaphragm. Manipulation
of the NF-kB pathway and other regulators of chemokine expression in
the diaphragm could represent a novel method for mitigating the
skeletal muscle inflammatory response associated with sepsis-induced
diaphragmatic dysfunction.
Collapse
|
48
|
Guillen-Marti J, Diaz R, Quiles MT, Lopez-Cano M, Vilallonga R, Huguet P, Ramon-y-Cajal S, Sanchez-Niubo A, Reventós J, Armengol M, Arbos MA. MMPs/TIMPs and inflammatory signalling de-regulation in human incisional hernia tissues. J Cell Mol Med 2008; 13:4432-43. [PMID: 19397782 PMCID: PMC4515059 DOI: 10.1111/j.1582-4934.2008.00637.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Incisional hernia is a common and important complication of laparotomies. Epidemiological studies allude to an underlying biological cause, at least in a subset of population. Interest has mainly focused on abnormal collagen metabolism. However, the role played by other determinants of extracellular matrix (ECM) composition is unknown. To date, there are few laboratory studies investigating the importance of biological factors contributing to incisional hernia development. We performed a descriptive tissue-based analysis to elucidate the possible relevance of matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) in association with local cytokine induction in human incisional hernia tissues. The expression profiles of MMPs, TIMPs and pro-inflammatory cytokine signalling were investigated in aponeurosis and skeletal muscle specimens taken intraoperatively from incisional hernia (n= 10) and control (n= 10) patients. Semiquantitative RT-PCR, zymography and immunoblotting analyses were done. Incisional hernia samples displayed alterations in the microstructure and loss of ECM, as assessed by histological analyses. Moreover, incisional hernia tissues showed increased MMP/TIMP ratios and de-regulated inflammatory signalling (tumor necrosis factor [TNFA] and interleukin [IL]-6 tended to increase, whereas aponeurosis TNFA receptors decreased). The changes were tissue-specific and were detectable at the mRNA and/or protein level. Statistical analyses showed several associations between individual MMPs, TIMPs, interstitial collagens and inflammatory markers. The increment of MMPs in the absence of a counterbalance by TIMPs, together with an ongoing de-regulated inflammatory signalling, may contribute in inducing a functional defect of the ECM network by post-translational mechanisms, which may trigger abdominal wall tissue loss and eventual rupture. The notable TIMP3 protein down-regulation in incisional hernia fascia may be of pathophysiological significance. We conclude that this study may help to pinpoint novel hypotheses of pathogenesis that can lead to a better understanding of the disease and ultimately to improvement in current therapeutic approaches.
Collapse
Affiliation(s)
- Jordi Guillen-Marti
- Institut de Recerca Hospital Universitari Vall d'Hebron, Universitat Autónoma de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Jeyanathan M, Mu J, Kugathasan K, Zhang X, Damjanovic D, Small C, Divangahi M, Petrof BJ, Hogaboam CM, Xing Z. Airway Delivery of Soluble Mycobacterial Antigens Restores Protective Mucosal Immunity by Single Intramuscular Plasmid DNA Tuberculosis Vaccination: Role of Proinflammatory Signals in the Lung. THE JOURNAL OF IMMUNOLOGY 2008; 181:5618-26. [DOI: 10.4049/jimmunol.181.8.5618] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
50
|
Huey KA, Fiscus G, Richwine AF, Johnson RW, Meador BM. In vivo vitamin E administration attenuates interleukin-6 and interleukin-1beta responses to an acute inflammatory insult in mouse skeletal and cardiac muscle. Exp Physiol 2008; 93:1263-72. [PMID: 18586856 DOI: 10.1113/expphysiol.2008.043190] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Antioxidants are associated with reduced pro-inflammatory cytokine expression in immune cells and isolated tissues; however, no studies have examined whether short-term vitamin E administration is associated with reduced lipopolysaccharide (LPS)-induced cytokine expression in mouse skeletal and cardiac muscle, in vivo. These experiments tested the hypothesis that vitamin E administration attenuates nuclear factor kappaB (NF-kappaB), IL-6, IL-1beta and tumour necrosis factor alpha (TNFalpha) responses in skeletal and cardiac muscle to an inflammatory challenge induced by systemic LPS. We compared IL-6, IL-1beta and TNFalpha mRNA and protein, activated NF-kappaB and total oxidized proteins in skeletal and cardiac muscle 4 or 24 h after saline or LPS injection in mice receiving vitamin E or placebo for 3 days prior to the insult. Skeletal and cardiac IL-6 mRNA and protein were significantly elevated by LPS in both groups, but responses were significantly lower in vitamin E- compared with placebo-treated mice. In skeletal and cardiac muscle, LPS increased IL-1beta mRNA and protein in placebo- but not vitamin E-treated mice. Lipopolysaccharide-induced levels of cardiac IL-1beta mRNA and protein and skeletal IL-1beta mRNA were lower with vitamin E than placebo. Lipopolysaccharide-induced NF-kappaB activation and increases in total oxidized proteins were attenuated with vitamin E compared with placebo in both tissues. Vitamin E decreased LPS-induced increases in plasma IL-1beta but not IL-6 compared with placebo. The major results provide the first in vivo evidence that short-term vitamin E administration reduces IL-6 and IL-1beta responses to LPS in skeletal and cardiac muscle and prevents LPS-induced increases in NF-kappaB activation and total oxidized proteins.
Collapse
Affiliation(s)
- K A Huey
- Department of Kinesiology, University of Illinois at Urbana-Champaign, 120 Freer Hall, 906 South Goodwin Avenue, Urbana, IL 61801, USA.
| | | | | | | | | |
Collapse
|