1
|
Velarde M, Sempore MY, Allibert V, Montel V, Castells J, Treffel L, Chopard A, Brioche T, Cochon L, Morel J, Bastide B, Durieux AC, Stevens L, Freyssenet D. Molecular determinants of skeletal muscle force loss in response to 5 days of dry immersion in human. J Cachexia Sarcopenia Muscle 2024. [PMID: 39450600 DOI: 10.1002/jcsm.13559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/10/2024] [Accepted: 07/15/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Astronauts in Earth's orbit experience microgravity, resulting in a decline of skeletal muscle mass and function. On Earth, models simulating microgravity have shown that the extent of the loss in muscle force is greater than the loss in muscle mass. The reasons behind this disproportionate loss of muscle force are still poorly understood. In the present study, we hypothesize that alongside the loss in skeletal muscle mass, modifications in the expression profile of genes encoding critical determinants of resting membrane potential, excitation-contraction coupling and Ca2+ handling contribute to the decline in skeletal muscle force. METHODS Healthy male volunteers (n = 18) participated in a 5-day dry immersion (DI) study, an Earth-based model of simulated microgravity. Muscle force measurement and MRI analysis of the cross-sectional area of thigh muscles were performed before and after DI. Biopsies of the vastus lateralis skeletal muscle performed before and after DI were used for the determination Ca2+ properties of isolated muscle fibres, molecular and biochemical analyses. RESULTS The extent of the decline in force, measured as maximal voluntary contraction of knee extensors (-11.1%, P < 0.01) was higher than the decline in muscle mass (-2.5%, P < 0.01). The decline in muscle mass was molecularly supported by a significant repression of the anabolic IGF-1/Akt/mTOR pathway (-19.9% and -40.9% in 4E-BP1 and RPS6 phosphorylation, respectively), a transcriptional downregulation of the autophagy-lysosome pathway and a downregulation in the mRNA levels of myofibrillar protein slow isoforms. At the single fibre level, biochemical and tension-pCa curve analyses showed that the loss in force was independent of fibre type (-11% and -12.3% in slow and fast fibres, respectively) and Ca2+ activation properties. Finally, we showed a significant remodelling in the expression of critical players of resting membrane potential (aquaporin 4: -24.9%, ATP1A2: +50.4%), excitation-contraction coupling (CHRNA1: +75.1%, CACNA2D1: -23.5%, JPH2: -24.2%, TRDN: -15.6%, S100A1: +27.2%), and Ca2+ handling (ATP2A2: -32.5%, CASQ1: -15%, ORAI1: -36.2%, ATP2B1: -19.1%). CONCLUSIONS These findings provide evidence that a deregulation in the expression profile of critical molecular determinants of resting membrane potential, excitation-contraction coupling, and Ca2+ handling could be involved in the loss of muscle force induced by DI. They also provide the paradigm for the understanding of muscle force loss during prolonged bed rest periods as those encountered in intensive care unit.
Collapse
Affiliation(s)
- Mathias Velarde
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet-Saint-Etienne, Saint Etienne, France
| | - Michel-Yves Sempore
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet-Saint-Etienne, Saint Etienne, France
- Département d'Anesthésie et Réanimation, Centre Hospitalier Universitaire de Saint Etienne, Saint Etienne, France
| | - Valentine Allibert
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet-Saint-Etienne, Saint Etienne, France
| | - Valérie Montel
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Lille, France
| | - Josiane Castells
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet-Saint-Etienne, Saint Etienne, France
| | - Loïc Treffel
- Institut Toulousain d'Ostéopathie, IRF'O, Toulouse, France
- Australian Research Centre in Complementary and Integrative Medicine (ARCCIM), School of Public Health, University of Technology Sydney, Ultimo, Australia
| | - Angèle Chopard
- DMEM, INRAE, Université Montpellier, Montpellier, France
| | - Thomas Brioche
- DMEM, INRAE, Université Montpellier, Montpellier, France
| | - Laetitia Cochon
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Lille, France
| | - Jérome Morel
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet-Saint-Etienne, Saint Etienne, France
- Département d'Anesthésie et Réanimation, Centre Hospitalier Universitaire de Saint Etienne, Saint Etienne, France
| | - Bruno Bastide
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Lille, France
| | - Anne-Cécile Durieux
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet-Saint-Etienne, Saint Etienne, France
| | - Laurence Stevens
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale, ULR 7369 - URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Lille, France
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet-Saint-Etienne, Saint Etienne, France
| |
Collapse
|
2
|
Morel J, Pignard AS, Castells J, Allibert V, Hatimi L, Buhot B, Velarde M, Durieux AC, Freyssenet D. Myostatin gene invalidation does not prevent skeletal muscle mass loss during experimental sepsis in mice. J Physiol 2024; 602:2839-2854. [PMID: 38748517 DOI: 10.1113/jp284973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 04/26/2024] [Indexed: 06/15/2024] Open
Abstract
Loss of muscle mass and function induced by sepsis contributes to physical inactivity and disability in intensive care unit patients. Limiting skeletal muscle deconditioning may thus be helpful in reducing the long-term effect of muscle wasting in patients. We tested the hypothesis that invalidation of the myostatin gene, which encodes a powerful negative regulator of skeletal muscle mass, could prevent or attenuate skeletal muscle wasting and improve survival of septic mice. Sepsis was induced by caecal ligature and puncture (CLP) in 13-week-old C57BL/6J wild-type and myostatin knock-out male mice. Survival rates were similar in wild-type and myostatin knock-out mice seven days after CLP. Loss in muscle mass was also similar in wild-type and myostatin knock-out mice 4 and 7 days after CLP. The loss in muscle mass was molecularly supported by an increase in the transcript level of E3-ubiquitin ligases and autophagy-lysosome markers. This transcriptional response was blunted in myostatin knock-out mice. No change was observed in the protein level of markers of the anabolic insulin/IGF1-Akt-mTOR pathway. Muscle strength was similarly decreased in wild-type and myostatin knock-out mice 4 and 7 days after CLP. This was associated with a modified expression of genes involved in ion homeostasis and excitation-contraction coupling, suggesting that a long-term functional recovery following experimental sepsis may be impaired by a dysregulated expression of molecular determinants of ion homeostasis and excitation-contraction coupling. In conclusion, myostatin gene invalidation does not provide any benefit in preventing skeletal muscle mass loss and strength in response to experimental sepsis. KEY POINTS: Survival rates are similar in wild-type and myostatin knock-out mice seven days after the induction of sepsis. Loss in muscle mass and muscle strength are similar in wild-type and myostatin knock-out mice 4 and 7 days after the induction of an experimental sepsis. Despite evidence of a transcriptional regulation, the protein level of markers of the anabolic insulin/IGF1-Akt-mTOR pathway remained unchanged. RT-qPCR analysis of autophagy-lysosome pathway markers indicates that activity of the pathway may be altered by experimental sepsis in wild-type and myostatin knock-out mice. Experimental sepsis induces greater variations in the mRNA levels of wild-type mice than those of myostatin knock-out mice, without providing any significant catabolic resistance or functional benefits.
Collapse
Affiliation(s)
- Jérome Morel
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
- Département d'anesthésie et réanimation, Centre Hospitalier Universitaire de Saint Etienne, Saint Etienne, France
| | - Anne Sophie Pignard
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
- Département d'anesthésie et réanimation, Centre Hospitalier Universitaire de Saint Etienne, Saint Etienne, France
| | - Josiane Castells
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Valentine Allibert
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Lahcène Hatimi
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Benjamin Buhot
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Mathias Velarde
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Anne Cécile Durieux
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| |
Collapse
|
3
|
Leduc-Gaudet JP, Miguez K, Cefis M, Faitg J, Moamer A, Chaffer TJ, Reynaud O, Broering FE, Shams A, Mayaki D, Huck L, Sandri M, Gouspillou G, Hussain SN. Autophagy ablation in skeletal muscles worsens sepsis-induced muscle wasting, impairs whole-body metabolism, and decreases survival. iScience 2023; 26:107475. [PMID: 37588163 PMCID: PMC10425945 DOI: 10.1016/j.isci.2023.107475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 05/18/2023] [Accepted: 07/21/2023] [Indexed: 08/18/2023] Open
Abstract
Septic patients frequently develop skeletal muscle wasting and weakness, resulting in severe clinical consequences and adverse outcomes. Sepsis triggers sustained induction of autophagy, a key cellular degradative pathway, in skeletal muscles. However, the impact of enhanced autophagy on sepsis-induced muscle dysfunction remains unclear. Using an inducible and muscle-specific Atg7 knockout mouse model (Atg7iSkM-KO), we investigated the functional importance of skeletal muscle autophagy in sepsis using the cecal ligation and puncture model. Atg7iSkM-KO mice exhibited a more severe phenotype in response to sepsis, marked by severe muscle wasting, hypoglycemia, higher ketone levels, and a decreased in survival as compared to mice with intact Atg7. Sepsis and Atg7 deletion resulted in the accumulation of mitochondrial dysfunction, although sepsis did not further worsen mitochondrial dysfunction in Atg7iSkM-KO mice. Overall, our study demonstrates that autophagy inactivation in skeletal muscles triggers significant worsening of sepsis-induced muscle and metabolic dysfunctions and negatively impacts survival.
Collapse
Affiliation(s)
- Jean-Philippe Leduc-Gaudet
- Research Group in Cellular Signaling, Department of Medical Biology, Université du Québec À Trois-Rivières, Trois-Rivières, QC G9A 5H7, Canada
- Department of Critical Care and Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre (MUHC), Montréal, QC H3H 2R9, Canada
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Département des sciences de l’activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC H2X 1Y4, Canada
| | - Kayla Miguez
- Department of Critical Care and Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre (MUHC), Montréal, QC H3H 2R9, Canada
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Marina Cefis
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Département des sciences de l’activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC H2X 1Y4, Canada
| | - Julie Faitg
- Département des sciences de l’activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC H2X 1Y4, Canada
- Amazentis SA, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Alaa Moamer
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Tomer Jordi Chaffer
- Department of Critical Care and Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre (MUHC), Montréal, QC H3H 2R9, Canada
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Olivier Reynaud
- Département des sciences de l’activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC H2X 1Y4, Canada
| | - Felipe E. Broering
- Department of Critical Care and Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre (MUHC), Montréal, QC H3H 2R9, Canada
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Anwar Shams
- Department of Pharmacology, Faculty of Medicine, Taif University, P.O.BOX 11099, Taif 21944, Saudi Arabia
| | - Dominique Mayaki
- Department of Critical Care and Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre (MUHC), Montréal, QC H3H 2R9, Canada
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Laurent Huck
- Department of Critical Care and Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre (MUHC), Montréal, QC H3H 2R9, Canada
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Marco Sandri
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Veneto Institute of Molecular Medicine (VIMM) and Department of Biomedical Science, Università di Padova, 35129 Padova, Italy
| | - Gilles Gouspillou
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Département des sciences de l’activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC H2X 1Y4, Canada
| | - Sabah N.A. Hussain
- Department of Critical Care and Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre (MUHC), Montréal, QC H3H 2R9, Canada
- Meakins-Christie Laboratories, Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| |
Collapse
|
4
|
Osuru HP, Ikeda K, Atluri N, Thiele RH. Moderate exercise-induced dynamics on key sepsis-associated signaling pathways in the liver. Crit Care 2023; 27:266. [PMID: 37407986 DOI: 10.1186/s13054-023-04551-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/25/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND There is a clear relationship between quantitative measures of fitness (e.g., VO2 max) and outcomes after surgical procedures. Whether or not fitness is a modifiable risk factor and what underlying biological processes drive these changes are not known. The purpose of this study was to evaluate the moderate exercise training effect on sepsis outcomes (survival) as well as the hepatic biological response. We chose to study the liver because it plays a central role in the regulation of immune defense during systemic infection and receives blood flow directly from the origin of infection (gut) in the cecal ligation and puncture (CLP) model. METHODS We randomized 50 male (♂) and female (♀) Sprague-Dawley rats (10 weeks, 340 g) to 3 weeks of treadmill exercise training, performed CLP to induce polymicrobial "sepsis," and monitored survival for five days (Part I). In parallel (Part II), we randomized 60 rats to control/sedentary (G1), exercise (G2), exercise + sham surgery (G3), CLP/sepsis (G4), exercise + CLP [12 h (G5) and 24 h (G6)], euthanized at 12 or 24 h, and explored molecular pathways related to exercise and sepsis survival in hepatic tissue and serum. RESULTS Three weeks of exercise training significantly increased rat survival following CLP (polymicrobial sepsis). CLP increased inflammatory markers (e.g., TNF-a, IL-6), which were attenuated by exercise. Sepsis suppressed the SOD and Nrf2 expression, and exercise before sepsis restored SOD and Nrf2 levels near the baseline. CLP led to increased HIF1a expression and oxidative and nitrosative stress, the latter of which were attenuated by exercise. Haptoglobin expression levels were increased in CLP animals, which was significantly amplified in exercise + CLP (24 h) rats. CONCLUSIONS Moderate exercise training (3 weeks) increased the survival in rats exposed to CLP, which was associated with less inflammation, less oxidative and nitrosative stress, and activation of antioxidant defense pathways.
Collapse
Affiliation(s)
- Hari Prasad Osuru
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710-0710, Charlottesville, VA, 22908-0710, USA.
| | - Keita Ikeda
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710-0710, Charlottesville, VA, 22908-0710, USA
| | - Navya Atluri
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710-0710, Charlottesville, VA, 22908-0710, USA
| | - Robert H Thiele
- Department of Anesthesiology, University of Virginia School of Medicine, P.O. Box 800710-0710, Charlottesville, VA, 22908-0710, USA.
| |
Collapse
|
5
|
Martin A, Gallot YS, Freyssenet D. Molecular mechanisms of cancer cachexia-related loss of skeletal muscle mass: data analysis from preclinical and clinical studies. J Cachexia Sarcopenia Muscle 2023; 14:1150-1167. [PMID: 36864755 PMCID: PMC10235899 DOI: 10.1002/jcsm.13073] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/15/2022] [Accepted: 08/14/2022] [Indexed: 03/04/2023] Open
Abstract
Cancer cachexia is a systemic hypoanabolic and catabolic syndrome that diminishes the quality of life of cancer patients, decreases the efficiency of therapeutic strategies and ultimately contributes to decrease their lifespan. The depletion of skeletal muscle compartment, which represents the primary site of protein loss during cancer cachexia, is of very poor prognostic in cancer patients. In this review, we provide an extensive and comparative analysis of the molecular mechanisms involved in the regulation of skeletal muscle mass in human cachectic cancer patients and in animal models of cancer cachexia. We summarize data from preclinical and clinical studies investigating how the protein turnover is regulated in cachectic skeletal muscle and question to what extent the transcriptional and translational capacities, as well as the proteolytic capacity (ubiquitin-proteasome system, autophagy-lysosome system and calpains) of skeletal muscle are involved in the cachectic syndrome in human and animals. We also wonder how regulatory mechanisms such as insulin/IGF1-AKT-mTOR pathway, endoplasmic reticulum stress and unfolded protein response, oxidative stress, inflammation (cytokines and downstream IL1ß/TNFα-NF-κB and IL6-JAK-STAT3 pathways), TGF-ß signalling pathways (myostatin/activin A-SMAD2/3 and BMP-SMAD1/5/8 pathways), as well as glucocorticoid signalling, modulate skeletal muscle proteostasis in cachectic cancer patients and animals. Finally, a brief description of the effects of various therapeutic strategies in preclinical models is also provided. Differences in the molecular and biochemical responses of skeletal muscle to cancer cachexia between human and animals (protein turnover rates, regulation of ubiquitin-proteasome system and myostatin/activin A-SMAD2/3 signalling pathways) are highlighted and discussed. Identifying the various and intertwined mechanisms that are deregulated during cancer cachexia and understanding why they are decontrolled will provide therapeutic targets for the treatment of skeletal muscle wasting in cancer patients.
Collapse
Affiliation(s)
- Agnès Martin
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| | - Yann S. Gallot
- LBEPS, Univ Evry, IRBA, Université Paris SaclayEvryFrance
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| |
Collapse
|
6
|
Pierre A, Bourel C, Favory R, Brassart B, Wallet F, Daussin FN, Normandin S, Howsam M, Romien R, Lemaire J, Grolaux G, Durand A, Frimat M, Bastide B, Amouyel P, Boulanger E, Preau S, Lancel S. Sepsis-like Energy Deficit Is Not Sufficient to Induce Early Muscle Fiber Atrophy and Mitochondrial Dysfunction in a Murine Sepsis Model. BIOLOGY 2023; 12:529. [PMID: 37106730 PMCID: PMC10136327 DOI: 10.3390/biology12040529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Sepsis-induced myopathy is characterized by muscle fiber atrophy, mitochondrial dysfunction, and worsened outcomes. Whether whole-body energy deficit participates in the early alteration of skeletal muscle metabolism has never been investigated. Three groups were studied: "Sepsis" mice, fed ad libitum with a spontaneous decrease in caloric intake (n = 17), and "Sham" mice fed ad libitum (Sham fed (SF), n = 13) or subjected to pair-feeding (Sham pair fed (SPF), n = 12). Sepsis was induced by the intraperitoneal injection of cecal slurry in resuscitated C57BL6/J mice. The feeding of the SPF mice was restricted according to the food intake of the Sepsis mice. Energy balance was evaluated by indirect calorimetry over 24 h. The tibialis anterior cross-sectional area (TA CSA), mitochondrial function (high-resolution respirometry), and mitochondrial quality control pathways (RTqPCR and Western blot) were assessed 24 h after sepsis induction. The energy balance was positive in the SF group and negative in both the SPF and Sepsis groups. The TA CSA did not differ between the SF and SPF groups, but was reduced by 17% in the Sepsis group compared with the SPF group (p < 0.05). The complex-I-linked respiration in permeabilized soleus fibers was higher in the SPF group than the SF group (p < 0.05) and lower in the Sepsis group than the SPF group (p < 0.01). Pgc1α protein expression increased 3.9-fold in the SPF mice compared with the SF mice (p < 0.05) and remained unchanged in the Sepsis mice compared with the SPF mice; the Pgc1α mRNA expression decreased in the Sepsis compared with the SPF mice (p < 0.05). Thus, the sepsis-like energy deficit did not explain the early sepsis-induced muscle fiber atrophy and mitochondrial dysfunction, but led to specific metabolic adaptations not observed in sepsis.
Collapse
Affiliation(s)
- Alexandre Pierre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Claire Bourel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Raphael Favory
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Benoit Brassart
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Frederic Wallet
- Division of Bacteriology, Biology Pathology Institute of Lille, CHU de Lille, F-59000 Lille, France
| | - Frederic N. Daussin
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d’Opale, ULR 7369-URePSSS-Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Sylvain Normandin
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Michael Howsam
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Raphael Romien
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Jeremy Lemaire
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Gaelle Grolaux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Arthur Durand
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Marie Frimat
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Nephrology, CHU de Lille, Université de Lille, F-59000 Lille, France
| | - Bruno Bastide
- Univ. Lille, Univ. Artois, Univ. Littoral Côte d’Opale, ULR 7369-URePSSS-Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000 Lille, France
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Eric Boulanger
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| | - Sebastien Preau
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
- Division of Intensive Care, Hôpital Roger Salengro, CHU de Lille, F-59000 Lille, France
| | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France
| |
Collapse
|
7
|
Sepsis-Associated Muscle Wasting: A Comprehensive Review from Bench to Bedside. Int J Mol Sci 2023; 24:ijms24055040. [PMID: 36902469 PMCID: PMC10003568 DOI: 10.3390/ijms24055040] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/21/2023] [Accepted: 03/04/2023] [Indexed: 03/08/2023] Open
Abstract
Sepsis-associated muscle wasting (SAMW) is characterized by decreased muscle mass, reduced muscle fiber size, and decreased muscle strength, resulting in persistent physical disability accompanied by sepsis. Systemic inflammatory cytokines are the main cause of SAMW, which occurs in 40-70% of patients with sepsis. The pathways associated with the ubiquitin-proteasome and autophagy systems are particularly activated in the muscle tissues during sepsis and may lead to muscle wasting. Additionally, expression of muscle atrophy-related genes Atrogin-1 and MuRF-1 are seemingly increased via the ubiquitin-proteasome pathway. In clinical settings, electrical muscular stimulation, physiotherapy, early mobilization, and nutritional support are used for patients with sepsis to prevent or treat SAMW. However, there are no pharmacological treatments for SAMW, and the underlying mechanisms are still unknown. Therefore, research is urgently required in this field.
Collapse
|
8
|
Voiriot G, Oualha M, Pierre A, Salmon-Gandonnière C, Gaudet A, Jouan Y, Kallel H, Radermacher P, Vodovar D, Sarton B, Stiel L, Bréchot N, Préau S, Joffre J. Chronic critical illness and post-intensive care syndrome: from pathophysiology to clinical challenges. Ann Intensive Care 2022; 12:58. [PMID: 35779142 PMCID: PMC9250584 DOI: 10.1186/s13613-022-01038-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/20/2022] [Indexed: 12/24/2022] Open
Abstract
Background Post‐intensive care syndrome (PICS) encompasses physical, cognition, and mental impairments persisting after intensive care unit (ICU) discharge. Ultimately it significantly impacts the long‐term prognosis, both in functional outcomes and survival. Thus, survivors often develop permanent disabilities, consume a lot of healthcare resources, and may experience prolonged suffering. This review aims to present the multiple facets of the PICS, decipher its underlying mechanisms, and highlight future research directions. Main text This review abridges the translational data underlying the multiple facets of chronic critical illness (CCI) and PICS. We focus first on ICU-acquired weakness, a syndrome characterized by impaired contractility, muscle wasting, and persisting muscle atrophy during the recovery phase, which involves anabolic resistance, impaired capacity of regeneration, mitochondrial dysfunction, and abnormalities in calcium homeostasis. Second, we discuss the clinical relevance of post-ICU cognitive impairment and neuropsychological disability, its association with delirium during the ICU stay, and the putative role of low-grade long-lasting inflammation. Third, we describe the profound and persistent qualitative and quantitative alteration of the innate and adaptive response. Fourth, we discuss the biological mechanisms of the progression from acute to chronic kidney injury, opening the field for renoprotective strategies. Fifth, we report long-lasting pulmonary consequences of ARDS and prolonged mechanical ventilation. Finally, we discuss several specificities in children, including the influence of the child’s pre-ICU condition, development, and maturation. Conclusions Recent understandings of the biological substratum of the PICS’ distinct features highlight the need to rethink our patient trajectories in the long term. A better knowledge of this syndrome and precipitating factors is necessary to develop protocols and strategies to alleviate the CCI and PICS and ultimately improve patient recovery.
Collapse
Affiliation(s)
- Guillaume Voiriot
- Service de Médecine Intensive Réanimation, Hôpital Tenon, Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Mehdi Oualha
- Pediatric Intensive Care Unit, Necker Hospital, APHP, Centre - Paris University, Paris, France
| | - Alexandre Pierre
- Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University Lille, Inserm, CHU Lille, 59000, Lille, France.,Department of Intensive Care Medicine, Critical Care Center, CHU Lille, 59000, Lille, France.,Faculté de Médecine de Tours, Centre d'Etudes des Pathologies Respiratoires, INSERM U1100, University Lille, Tours, France
| | - Charlotte Salmon-Gandonnière
- Service de Médecine Intensive Réanimation, CHRU de Tours, Réseau CRICS-TRIGGERSEP F-CRIN Research Network, Tours, France
| | - Alexandre Gaudet
- Department of Intensive Care Medicine, Critical Care Center, CHU Lille, 59000, Lille, France.,Faculté de Médecine de Tours, Centre d'Etudes des Pathologies Respiratoires, INSERM U1100, University Lille, Tours, France.,Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, 59000, Lille, France
| | - Youenn Jouan
- Service de Médecine Intensive Réanimation, CHRU de Tours, Réseau CRICS-TRIGGERSEP F-CRIN Research Network, Tours, France
| | - Hatem Kallel
- Service de Réanimation, Centre Hospitalier de Cayenne, French Guiana, Cayenne, France
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, 89070, Ulm, Germany
| | - 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
| | - Benjamine Sarton
- Critical Care Unit, University Hospital of Purpan, Toulouse, France.,Toulouse NeuroImaging Center, ToNIC, Inserm 1214, Paul Sabatier University, Toulouse, France
| | - Laure Stiel
- Service de Réanimation Médicale, Groupe Hospitalier de la Région Mulhouse Sud Alsace, Mulhouse, France.,INSERM, LNC UMR 1231, FCS Bourgogne Franche Comté LipSTIC LabEx, Dijon, France
| | - Nicolas Bréchot
- Service de Médecine Intensive Réanimation, Sorbonne Université, Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,College de France, Center for Interdisciplinary Research in Biology (CIRB)-UMRS INSERM U1050 - CNRS 7241, Paris, France
| | - Sébastien Préau
- Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, University Lille, Inserm, CHU Lille, 59000, Lille, France.,Service de Médecine Intensive Réanimation, CHRU de Tours, Réseau CRICS-TRIGGERSEP F-CRIN Research Network, Tours, France
| | - Jérémie Joffre
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, 94143, USA. .,Medical Intensive Care Unit, Saint Antoine University Hospital, APHP, Sorbonne University, 75012, Paris, France. .,Sorbonne University, Centre de Recherche Saint-Antoine INSERM U938, 75012, Paris, France.
| | | |
Collapse
|
9
|
Role of Glucocorticoid Signaling and HDAC4 Activation in Diaphragm and Gastrocnemius Proteolytic Activity in Septic Rats. Int J Mol Sci 2022; 23:ijms23073641. [PMID: 35408999 PMCID: PMC8998191 DOI: 10.3390/ijms23073641] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis increases glucocorticoid and decreases IGF-1, leading to skeletal muscle wasting and cachexia. Muscle atrophy mainly takes place in locomotor muscles rather than in respiratory ones. Our study aimed to elucidate the mechanism responsible for this difference in muscle proteolysis, focusing on local inflammation and IGF-1 as well as on their glucocorticoid response and HDAC4-myogenin activation. Sepsis was induced in adult male rats by lipopolysaccharide (LPS) injection (10 mg/kg), and 24 h afterwards, rats were euthanized. LPS increased TNFα and IL-10 expression in both muscles studied, the diaphragm and gastrocnemius, whereas IL-6 and SOCS3 mRNA increased only in diaphragm. In comparison with gastrocnemius, diaphragm showed a lower increase in proteolytic marker expression (atrogin-1 and LC3b) and in LC3b protein lipidation after LPS administration. LPS increased the expression of glucocorticoid induced factors, KLF15 and REDD1, and decreased that of IGF-1 in gastrocnemius but not in the diaphragm. In addition, an increase in HDAC4 and myogenin expression was induced by LPS in gastrocnemius, but not in the diaphragm. In conclusion, the lower activation of both glucocorticoid signaling and HDAC4-myogenin pathways by sepsis can be one of the causes of lower sepsis-induced proteolysis in the diaphragm compared to gastrocnemius.
Collapse
|
10
|
Yin D, Lin D, Xie Y, Gong A, Jiang P, Wu J. Neuregulin-1β Alleviates Sepsis-Induced Skeletal Muscle Atrophy by Inhibiting Autophagy via AKT/mTOR Signaling Pathway in Rats. Shock 2022; 57:397-407. [PMID: 34559744 DOI: 10.1097/shk.0000000000001860] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Several studies have shown that excessive protein degradation is a major cause of skeletal muscle atrophy induced by sepsis, and autophagy is the main pathway participating in protein degradation. However, the role of autophagy in sepsis is still controversial. Previously, we found that neuregulin-1β (NRG-1β) alleviated sepsis-induced diaphragm atrophy through the phosphatidylinositol-3 kinase signaling pathway. Akt/mechanistic target of rapamycin (mTOR) is a classic signaling pathway to regulate autophagy, which maintains intracellular homeostasis. This study aimed to investigate whether NRG-1β could alleviate sepsis-induced skeletal muscle atrophy by regulating autophagy. METHODS L6 rat myoblast cells were differentiated using 2% fetal bovine serum into myotubes, which were divided into four groups: Con group treated with normal serum; Sep group treated with septic serum to form a sepsis cell model; septic serum + NRG-1β (SN) group treated with septic serum for 24 h followed by injection with NRG-1β and incubation for another 48 h; and serum+NRG-1β+LY294002 group, in which the PI3K inhibitor LY294002 was added 30 min before NRG-1β, and other treatments were similar to those in SN group. Effects of NRG-1β were also evaluated in vivo using Sprague-Dawley (SD) rats, in which sepsis was induced by cecal ligation and puncture (CLP). RESULTS In L6 myotubes treated with septic serum, the expression of autophagy-related proteins UNC-51 like kinase 1, p-Beclin-1, and Beclin-1, and the ratio of LC3B II/I were highly increased, while protein p62 expression was decreased, indicating that autophagy was excessively activated. Moreover, NRG-1 expression was decreased, as detected by confocal immunofluorescence and western blotting. Upon exogenous addition of NRG-1β, autophagy was inhibited by the activation of Akt/mTOR signaling pathway, and cell viability was also increased. These effects disappeared in the presence of LY294002. In SD rats, sepsis was induced by CLP. NRG-1β was shown to inhibit autophagy in these rats via the Akt/mTOR pathway, leading to increased body weight of the septic SD rats and alleviation of atrophy of the tibialis anterior muscle. CONCLUSION NRG-1β could alleviate sepsis-induced skeletal muscle atrophy by inhibiting autophagy via the AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Dandan Yin
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Dawei Lin
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Yunbin Xie
- Department of Anesthesiology, The First People's Hospital of Changzhou, Changzhou City, Jiangsu Province, China
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Peng Jiang
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Jin Wu
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Zhenjiang City, Jiangsu Province, China
| |
Collapse
|
11
|
Liu Q, Gao J, Deng J, Xiao J. Current Studies and Future Directions of Exercise Therapy for Muscle Atrophy Induced by Heart Failure. Front Cardiovasc Med 2020; 7:593429. [PMID: 33195482 PMCID: PMC7644508 DOI: 10.3389/fcvm.2020.593429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Muscle atrophy is a common complication of heart failure. At present, there is no specific treatment to reverse the course of muscle atrophy. Exercise training, due to the safety and easy operation, is a recommended therapy for muscle atrophy induced by heart failure. However, the patients with muscle atrophy are weak in mobility and may not be able to train for a long time. Therefore, it is necessary to explore novel targets of exercise protection for muscle atrophy, so as to improve the quality of life and survival rate of patients with muscular atrophy induced by heart failure. This article aims to review latest studies, summarize the evidence and limitations, and provide a glimpse into the future of exercise for the treatment of muscle atrophy induced by heart failure. We wish to highlight some important findings about the essential roles of exercise sensors in muscle atrophy induced by heart failure, which might be helpful for searching potential therapeutic targets for muscle wasting induced by heart failure.
Collapse
Affiliation(s)
- Qi Liu
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Juan Gao
- School of Medicine, Shanghai University, Shanghai, China
| | - Jiali Deng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
12
|
Is Mitochondrial Oxidative Stress the Key Contributor to Diaphragm Atrophy and Dysfunction in Critically Ill Patients? Crit Care Res Pract 2020; 2020:8672939. [PMID: 32377432 PMCID: PMC7191397 DOI: 10.1155/2020/8672939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 02/08/2023] Open
Abstract
Diaphragm dysfunction is prevalent in the progress of respiratory dysfunction in various critical illnesses. Respiratory muscle weakness may result in insufficient ventilation, coughing reflection suppression, pulmonary infection, and difficulty in weaning off respirators. All of these further induce respiratory dysfunction and even threaten the patients' survival. The potential mechanisms of diaphragm atrophy and dysfunction include impairment of myofiber protein anabolism, enhancement of myofiber protein degradation, release of inflammatory mediators, imbalance of metabolic hormones, myonuclear apoptosis, autophagy, and oxidative stress. Among these contributors, mitochondrial oxidative stress is strongly implicated to play a key role in the process as it modulates diaphragm protein synthesis and degradation, induces protein oxidation and functional alteration, enhances apoptosis and autophagy, reduces mitochondrial energy supply, and is regulated by inflammatory cytokines via related signaling molecules. This review aims to provide a concise overview of pathological mechanisms of diaphragmatic dysfunction in critically ill patients, with special emphasis on the role and modulating mechanisms of mitochondrial oxidative stress.
Collapse
|
13
|
Cavaillon J, Singer M, Skirecki T. Sepsis therapies: learning from 30 years of failure of translational research to propose new leads. EMBO Mol Med 2020; 12:e10128. [PMID: 32176432 PMCID: PMC7136965 DOI: 10.15252/emmm.201810128] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/13/2022] Open
Abstract
Sepsis has been identified by the World Health Organization (WHO) as a global health priority. There has been a tremendous effort to decipher underlying mechanisms responsible for organ failure and death, and to develop new treatments. Despite saving thousands of animals over the last three decades in multiple preclinical studies, no new effective drug has emerged that has clearly improved patient outcomes. In the present review, we analyze the reasons for this failure, focusing on the inclusion of inappropriate patients and the use of irrelevant animal models. We advocate against repeating the same mistakes and propose changes to the research paradigm. We discuss the long-term consequences of surviving sepsis and, finally, list some putative approaches-both old and new-that could help save lives and improve survivorship.
Collapse
Affiliation(s)
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care MedicineUniversity College LondonLondonUK
| | - Tomasz Skirecki
- Laboratory of Flow Cytometry and Department of Anesthesiology and Intensive Care MedicineCentre of Postgraduate Medical EducationWarsawPoland
| |
Collapse
|
14
|
Pseudomonas aeruginosa Quorum Sensing Molecule Alters Skeletal Muscle Protein Homeostasis by Perturbing the Antioxidant Defense System. mBio 2019; 10:mBio.02211-19. [PMID: 31575771 PMCID: PMC6775459 DOI: 10.1128/mbio.02211-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle function is compromised in many illnesses, including chronic infections. The Pseudomonas aeruginosa quorum sensing (QS) signal, 2-amino acetophenone (2-AA), is produced during acute and chronic infections and excreted in human tissues, including the lungs of cystic fibrosis patients. We have shown that 2-AA facilitates pathogen persistence, likely via its ability to promote the formation of bacterial persister cells, and that it acts as an interkingdom immunomodulatory signal that epigenetically reprograms innate immune functions. Moreover, 2-AA compromises muscle contractility and impacts the expression of genes involved in reactive oxygen species (ROS) homeostasis in skeletal muscle and in mitochondrial functions. Here, we elucidate the molecular mechanisms of 2-AA's impairment of skeletal muscle function and ROS homeostasis. Murine in vivo and differentiated C2C12 myotube cell studies showed that 2-AA promotes ROS generation in skeletal muscle via the modulation of xanthine oxidase (XO) activity, NAD(P)H oxidase2 (NOX2) protein level, and the activity of antioxidant enzymes. ROS accumulation triggers the activity of AMP-activated protein kinase (AMPK), likely upstream of the observed locations of induction of ubiquitin ligases Muscle RING Finger 1 (MuRF1) and Muscle Atrophy F-box (MAFbx), and induces autophagy-related proteins. The protein-level perturbation in skeletal muscle of silent mating type information regulation 2 homolog 1 (SIRT1), peroxisome proliferator-activated receptor gamma coactivator 1 (PGC-1), and uncoupling protein 3 (UCP3) is rescued by the antioxidant N-acetyl-l-cysteine (NAC). Together, these results unveil a novel form of action of a QS bacterial molecule and provide molecular insights into the 2-AA-mediated skeletal muscle dysfunction caused by P. aeruginosa IMPORTANCE Pseudomonas aeruginosa, a bacterium that is resistant to treatment, causes serious acute, persistent, and relapsing infections in humans. There is increasing evidence that bacterial excreted small molecules play a critical role during infection. We have shown that a quorum sensing (QS)-regulated excreted small molecule, 2-AA, which is abundantly produced by P. aeruginosa, promotes persistent infections, dampens host inflammation, and triggers mitochondrial dysfunction in skeletal muscle. QS is a cell-to-cell communication system utilized by bacteria to promote collective behaviors. The significance of our study in identifying a mechanism that leads to skeletal muscle dysfunction, via the action of a QS molecule, is that it may open new avenues in the control of muscle loss as a result of infection and sepsis. Given that QS is a common characteristic of prokaryotes, it is possible that 2-AA-like molecules promoting similar effects may exist in other pathogens.
Collapse
|
15
|
Moarbes V, Mayaki D, Huck L, Leblanc P, Vassilakopoulos T, Petrof BJ, Hussain SNA. Differential regulation of myofibrillar proteins in skeletal muscles of septic mice. Physiol Rep 2019; 7:e14248. [PMID: 31660704 PMCID: PMC6817996 DOI: 10.14814/phy2.14248] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/06/2019] [Accepted: 09/07/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis elicits skeletal muscle atrophy as a result of decreased total protein synthesis and/or increased total protein degradation. It is unknown how and whether sepsis differentially affects the expression of specific myofibrillar proteins in respiratory and limb muscles. In this study, we measured the effects of sepsis myofibrillar mRNAs and their corresponding protein levels in the diaphragm (DIA) and tibialis anterior (TA) muscles in a murine cecal ligation and perforation (CLP) model of sepsis. Male mice (C57/BL6j) underwent CLP-induced sepsis. Sham-operated mice were subjected to the same surgical procedures, except for CLP. Mice were euthanized 24, 48, or 96 h postsurgery. Transcript and protein levels of autophagy-related genes, ubiquitin E3 ligases, and several myofibrillar genes were quantified. Sepsis elicited transient fiber atrophy in the DIA and prolonged atrophy in the TA. Atrophy was coincident with increased autophagy and ubiquitin E3 ligase expression. Myosin heavy chain isoforms decreased at 24 h in the DIA and across the time-course in the TA, myosin light chain isoforms decreased across the time-course in both muscles, and troponins T and C as well as tropomyosin decreased after 24 and 48 h in both the DIA and TA. α-Actin and troponin I were unaffected by sepsis. Sepsis-induced decreases in myofibrillar protein levels coincided with decreased mRNA expressions of these proteins, suggesting that transcriptional inhibition is involved. We hypothesize that sepsis-induced muscle atrophy is mediated by decreased transcription and enhanced degradation of specific myofibrillar proteins, including myosin heavy and light chains, troponin C, troponin T, and tropomyosin.
Collapse
Affiliation(s)
- Vanessa Moarbes
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontréalQuébecCanada
- Department of Critical CareMcGill University Health CentreMontréalQuébecCanada
| | - Dominique Mayaki
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontréalQuébecCanada
- Department of Critical CareMcGill University Health CentreMontréalQuébecCanada
| | - Laurent Huck
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontréalQuébecCanada
- Department of Critical CareMcGill University Health CentreMontréalQuébecCanada
| | - Philippe Leblanc
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontréalQuébecCanada
- Department of Critical CareMcGill University Health CentreMontréalQuébecCanada
| | - Theodoros Vassilakopoulos
- Critical Care Department, National & Kapodistrian University of Athens, Medical School, Evgenideion HospitalAthensGreece
- Department of MedicineMcGill University Health CentreMontréalQuébecCanada
| | - Basil J. Petrof
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontréalQuébecCanada
- Department of Critical CareMcGill University Health CentreMontréalQuébecCanada
| | - Sabah N. A. Hussain
- Meakins‐Christie Laboratories and Translational Research in Respiratory Diseases ProgramResearch Institute of the McGill University Health CentreMontréalQuébecCanada
- Department of Critical CareMcGill University Health CentreMontréalQuébecCanada
| |
Collapse
|
16
|
Yin X, Xin H, Mao S, Wu G, Guo L. The Role of Autophagy in Sepsis: Protection and Injury to Organs. Front Physiol 2019; 10:1071. [PMID: 31507440 PMCID: PMC6716215 DOI: 10.3389/fphys.2019.01071] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a systemic inflammatory disease with infection, and autophagy has been shown to play an important role in sepsis. This review summarizes the main regulatory mechanisms of autophagy in sepsis and its latest research. Recent studies have shown that autophagy can regulate innate immune processes and acquired immune processes, and the regulation of autophagy in different immune cells is different. Mitophagy can select damaged mitochondria and remove it to deal with oxidative stress damage. The process of mitophagy is regulated by other factors. Non-coding RNA is also an important factor in the regulation of autophagy. In addition, more and more studies in recent years have shown that autophagy plays different roles in different organs. It tends to be protective in the lungs, heart, kidneys, and brain, and tends to be damaging in skeletal muscle. We also mentioned that some drugs can regulate autophagy. The process of modulating autophagy through drug intervention appears to be a new potential hope for the treatment of sepsis.
Collapse
Affiliation(s)
- Xin Yin
- Department of Critical Care Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huang Xin
- Department of Critical Care Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuai Mao
- Department of Critical Care Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangping Wu
- Department of Critical Care Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liheng Guo
- Department of Critical Care Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
17
|
Jensen IJ, Sjaastad FV, Griffith TS, Badovinac VP. Sepsis-Induced T Cell Immunoparalysis: The Ins and Outs of Impaired T Cell Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 200:1543-1553. [PMID: 29463691 DOI: 10.4049/jimmunol.1701618] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Sepsis results in a deluge of pro- and anti-inflammatory cytokines, leading to lymphopenia and chronic immunoparalysis. Sepsis-induced long-lasting immunoparalysis is defined, in part, by impaired CD4 and CD8 αβ T cell responses in the postseptic environment. The dysfunction in T cell immunity affects naive, effector, and memory T cells and is not restricted to classical αβ T cells. Although sepsis-induced severe and transient lymphopenia is a contributory factor to diminished T cell immunity, T cell-intrinsic and -extrinsic factors/mechanisms also contribute to impaired T cell function. In this review, we summarize the current knowledge of how sepsis quantitatively and qualitatively impairs CD4 and CD8 T cell immunity of classical and nonclassical T cell subsets and discuss current therapeutic approaches being developed to boost the recovery of T cell immunity postsepsis induction.
Collapse
Affiliation(s)
- Isaac J Jensen
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Frances V Sjaastad
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455
| | - Thomas S Griffith
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, MN 55455.,Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,Department of Urology, University of Minnesota, Minneapolis, MN 55455.,Minneapolis VA Health Care System, Minneapolis, MN 55455
| | - Vladimir P Badovinac
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242; .,Department of Pathology, University of Iowa, Iowa City, IA 52242; and.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
18
|
Wang BJ, Zheng WL, Feng NN, Wang T, Zou H, Gu JH, Yuan Y, Liu XZ, Liu ZP, Bian JC. The Effects of Autophagy and PI3K/AKT/m-TOR Signaling Pathway on the Cell-Cycle Arrest of Rats Primary Sertoli Cells Induced by Zearalenone. Toxins (Basel) 2018; 10:toxins10100398. [PMID: 30274213 PMCID: PMC6215106 DOI: 10.3390/toxins10100398] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/16/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022] Open
Abstract
A high concentration of Zearalenone (ZEA) will perturb the differentiation of germ cells, and induce a death of germ cells, but the toxic mechanism and molecular mechanism remain unclear. The Sertoli cells (SCs) play an irreplaceable role in spermatogenesis. In order to explore the potential mechanism of ZEA male reproductive toxicity, we studied the effects of ZEA on cell proliferation, cell-cycle distribution, cell-cycle-related proteins and autophagy-related pathway the PI3K/Akt/mTOR signaling in primary cultured rats SCs, and the effects of autophagy and PI3K/AKT/m TOR signaling pathway on the SCs cell-cycle arrest induced by ZEA treated with the autophagy promoter RAPA, autophagy inhibitor CQ, and the PI3K inhibitor LY294002, respectively. The data revealed that ZEA could inhibit the proliferation of SCs by arresting the cell cycle in the G2/M phase and trigger the autophagy via inhibiting the PI3K/Akt/m TOR signaling pathway. Promoting or inhibiting the level of autophagy could either augment or reverse the arrest of cell cycle. And it was regulated by PI3K/Akt/m TOR signaling pathway. Taken together, this study provides evidence that autophagy and PI3K/Akt/m TOR signaling pathway are involved in regulating rats primary SCs cell-cycle arrest due to ZEA in vitro. To some extent, ZEA-induced autophagy plays a protective role in this process.
Collapse
Affiliation(s)
- Bing-Jie Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| | - Wang-Long Zheng
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Nan-Nan Feng
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Jian-Hong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Xue-Zhong Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Zong-Ping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| | - Jian-Chun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
19
|
Zearalenone altered the cytoskeletal structure via ER stress- autophagy- oxidative stress pathway in mouse TM4 Sertoli cells. Sci Rep 2018; 8:3320. [PMID: 29463855 PMCID: PMC5820275 DOI: 10.1038/s41598-018-21567-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/07/2018] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to investigate the molecular mechanisms of the destruction of cytoskeletal structure by Zearalenone (ZEA) in mouse-derived TM4 cells. In order to investigate the role of autophagy, oxidative stress and endoplasmic reticulum(ER) stress in the process of destruction of cytoskeletal structure, the effects of ZEA on the cell viability, cytoskeletal structure, autophagy, oxidative stress, ER stress, MAPK and PI3K- AKT- mTOR signaling pathways were studied. The data demonstrated that ZEA damaged the cytoskeletal structure through the induction of autophagy that leads to the alteration of cytoskeletal structure via elevated oxidative stress. Our results further showed that the autophagy was stimulated by ZEA through PI3K-AKT-mTOR and MAPK signaling pathways in TM4 cells. In addition, ZEA also induced the ER stress which was involved in the induction of the autophagy through inhibiting the ERK signal pathway to suppress the phosphorylation of mTOR. ER stress was involved in the damage of cytoskeletal structure through induction of autophagy by producing ROS. Taken together, this study revealed that ZEA altered the cytoskeletal structure via oxidative stress - autophagy- ER stress pathway in mouse TM4 Sertoli cells.
Collapse
|