1
|
Zhang HY, Wang JY, Li JJ, Zhu J, Weng GJ, Li YL, Zhao JW. Broad-spectrum pathogenic bacteria SERS sensing with face-centered high-index facets Au CPNCs & microarray chips: A novel platform able to achieve dual-readout detection. J Colloid Interface Sci 2025; 692:137485. [PMID: 40215900 DOI: 10.1016/j.jcis.2025.137485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/06/2025] [Accepted: 03/30/2025] [Indexed: 05/02/2025]
Abstract
Non-specificity and inadequate quantitative capability are the primary challenges faced by the surface-enhanced Raman scattering (SERS) technique, especially when it comes to detecting bacteria in real samples. Herein, a novel face-centered Au Convex Polyhedral Nanocrystal (Au CPNC) with high-index facets and its assembly Au CPNCs microarray chip were designed and fabricated to address these challenges, within the process where 4-mercaptophenylboronic acid (4-MPBA) was utilized as a multifunctional element. The as-prepared Au CPNC possesses anisotropic raised edges enjoying tunable localized surface plasmon resonance modes for SERS enhancement. Then we obtained long-region ordered Au CPNCs microarrays equipping even greater "hot spots" with a SERS enhancement factor (EF) up to 5.38 × 107. The constructed SERS probes excellently leveraged the outstanding SERS performance of Au CPNC and the superior functions of 4-MPBA, which enabled the differences among the bacterial "fingerprints" to be highlighted. Through partial least squares discriminant analysis (PLS-DA), we successfully identified Escherichia coli, Staphylococcus aureus, Pseudomonas aeruginosa and Listeria monocytogenes with achieving limits of detection (LODs) in spiked whole blood samples of 3, 1, 2, and 2 cfu/mL, respectively. Notably, the LODs for all samples by SERS mapping visual readout mode were as low as 10 cfu/mL. In practical applications, our method demonstrated its efficacy by 100 % accurately classifying (20 cases) of real blood samples. Altogether, the theoretical significance and application value of this study reside in providing fundamental insights and approaches for the development of pathogenic bacteria detection field.
Collapse
Affiliation(s)
- Hao-Yu Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Jing-Yuan Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jian-Jun Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China.
| | - Jian Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Guo-Jun Weng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China
| | - Ya-Li Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jun-Wu Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, People's Republic of China.
| |
Collapse
|
2
|
Braun AC, Oliveira TC, Thomazini LCD, Argenti G, Kotzian BJ, Machado V, Conte JHM, Zanfir C, Souto ACA, Ulian B, Vidart J, Wajner SM. Induced Types 2 and 3 Deiodinase in Non-Thyroidal Illness Syndrome and the Implications to Critical Illness-Induced Myopathy-A Prospective Cohort Study. Int J Mol Sci 2025; 26:2410. [PMID: 40141055 PMCID: PMC11941936 DOI: 10.3390/ijms26062410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Loss of muscle mass and strength is a common condition associated with adverse outcomes in critically ill patients. Here, we determined the correlation between non-thyroidal illness (NTIS) and molecular alterations in the muscle of critically ill individuals. We evaluated deiodinase expression, intramuscular triiodothyronine (T3) levels, and mitochondria and sarcoplasmic reticulum components. The cellular colocalization of the enzymes and its influence on myocytes and genes regulated by T3 were shown, including those of mitochondria. A prospective cohort of 96 patients. Blood and muscular samples were collected on admission to the intensive care unit (ICU), as well as clinical data and ultrasonographic measurements. Patients with NTIS showed increased oxidative stress markers associated with critical illness in muscle biopsy, such as carbonyl content and low sulfhydryl and GSH. The distribution pattern of deiodinases in muscle and its biochemical properties showed significant pathophysiological linkage between NTIS and muscle loss, as type 3-deiodinase (D3) was highly expressed in stem cells, preventing their differentiation in mature myocytes. Despite the high type 2-deiodinase (D2) expression in muscle tissue in the acute phase of critical illness, T3 was unmeasurable in the samples. In this scenario, we also demonstrated impaired expression of glucose transporters GLUT4, IRS1, and 2, which are involved in muscle illness. Here, we provide evidence that altered thyroid hormone metabolism contributes to stem cell dysfunction and further explain the mechanisms underlying critical illness-induced myopathy.
Collapse
Affiliation(s)
- André Cardoso Braun
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Thaliane Carvalho Oliveira
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Ludmilla C. D. Thomazini
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Gustavo Argenti
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Bruno Jaskulski Kotzian
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Valentina Machado
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - João Henrique M. Conte
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Carolina Zanfir
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Amanda C. A. Souto
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Bruna Ulian
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Josi Vidart
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
| | - Simone Magagnin Wajner
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil; (A.C.B.)
- Department of Internal Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Rio Grande do Sul, Brazil
| |
Collapse
|
3
|
Fessard A, Zavoriti A, Boyer N, Guillemaud J, Rahmati M, Del Carmine P, Gobet C, Chazaud B, Gondin J. Neuromuscular electrical stimulation training induces myonuclear accretion and hypertrophy in mice without overt signs of muscle damage and regeneration. Skelet Muscle 2025; 15:3. [PMID: 39910613 PMCID: PMC11796018 DOI: 10.1186/s13395-024-00372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Skeletal muscle is a plastic tissue that adapts to increased mechanical loading/contractile activity through fusion of muscle stem cells (MuSCs) with myofibers, a physiological process referred to as myonuclear accretion. However, it is still unclear whether myonuclear accretion is driven by increased mechanical loading per se, or occurs, at least in part, in response to muscle injury/regeneration. Here, we developed a non-damaging protocol to evaluate contractile activity-induced myonuclear accretion/hypertrophy in physiological conditions. METHODS Contractile activity was generated by applying repeated electrical stimuli over the mouse plantar flexor muscles. This method is commonly referred to as NeuroMuscular Electrical Simulation (NMES) in Human. Each NMES training session consisted of 80 isometric contractions delivered at ∼15% of maximal tetanic force to avoid muscle damage. C57BL/6J male mice were submitted to either a short (i.e., 6 sessions) or long (i.e., 12 sessions) individualized NMES training program while unstimulated mice were used as controls. Histological investigations were performed to assess the impact of NMES on MuSC number and status, myonuclei content and muscle tissue integrity, typology and size. RESULTS NMES led to a robust proliferation of MuSCs and myonuclear accretion in the absence of overt signs of muscle damage/regeneration. NMES-induced myonuclear accretion was specific to type IIB myofibers and was an early event preceding muscle hypertrophy inasmuch as a mild increase in myofiber cross-sectional area was only observed in response to the long-term NMES training protocol. CONCLUSION We conclude that NMES-induced myonuclear accretion and muscle hypertrophy are driven by a mild increase in mechanical loading in the absence of overt signs of muscle injury.
Collapse
Affiliation(s)
- Aurélie Fessard
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Aliki Zavoriti
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Natacha Boyer
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Jules Guillemaud
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Masoud Rahmati
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
- Department of Exercise Physiology, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad, Iran
| | - Peggy Del Carmine
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Christelle Gobet
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène (INMG), Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, 8 Avenue Rockefeller, Lyon, France.
| |
Collapse
|
4
|
Mart MF, Gordon JI, González-Seguel F, Mayer KP, Brummel N. Muscle Dysfunction and Physical Recovery After Critical Illness. J Intensive Care Med 2025:8850666251317467. [PMID: 39905778 DOI: 10.1177/08850666251317467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
During critical illness, patients experience significant and rapid onsets of muscle wasting and dysfunction with loss of strength, mass, and power. These deficits often persist long after the ICU, leading to impairments in physical function including reduced exercise capacity and increased frailty and disability. While there are numerous studies describing the epidemiology of impaired muscle and physical function in the ICU, there are significantly fewer data investigating mechanisms of prolonged and persistent impairments in ICU survivors. Additionally, while several potential clinical risk factors associated with poor physical recovery have been identified, there remains a dearth of interventions that have effectively improved outcomes long-term among survivors. In this article, we aim to provide a thorough, evidence-based review of the current state of knowledge regarding muscle dysfunction and physical function after critical illness with a focus on post-ICU and post-hospitalization phase of recovery.
Collapse
Affiliation(s)
- Matthew F Mart
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Nashville, TN, USA
- Geriatric Research, Education and Clinical Center (GRECC) Service, Department of Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Joshua I Gordon
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), The Ohio State University College of Medicine, Columbus, OH, USA
| | - Felipe González-Seguel
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA
- Faculty of Medicine, School of Physical Therapy, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Kirby P Mayer
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Nathan Brummel
- Critical Illness, Brain Dysfunction, and Survivorship (CIBS) Center, Nashville, TN, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Center for the Advancement of Team Science, Analytics, and Systems Thinking in Health Services and Implementation Science Research (CATALYST), The Ohio State University College of Medicine, Columbus, OH, USA
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
5
|
Xue Q, Zhang D, Zou J, Wang H, Shi R, Dong L. Treatment advances of sepsis‑induced myopathy (Review). Biomed Rep 2025; 22:19. [PMID: 39651403 PMCID: PMC11621912 DOI: 10.3892/br.2024.1897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/04/2024] [Indexed: 12/11/2024] Open
Abstract
Sepsis-induced myopathy (SIM) is a muscle disease caused by multiple pathological and physiological mechanisms associated with sepsis. The pathogenesis of SIM is extremely complex and still unclear, making treatment challenging. At present, clinical treatment includes early functional exercise, respiratory muscle strength training, regulation of nutritional structure and functional electrical stimulation. Drugs targeting the regulation of the ubiquitin-proteasome system, autophagy-lysosome system, calpain and caspase activation pathways, have provided potential therapeutic targets for the treatment of muscle atrophy. Stem cell transplantation therapy brings new hope for the treatment of SIM.
Collapse
Affiliation(s)
- Qiuli Xue
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Deyou Zhang
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiarui Zou
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haitao Wang
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ruiyuan Shi
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lihua Dong
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
6
|
Lei L, He L, Zou T, Qiu J, Li Y, Zhou R, Qin Y, Yin W. Predicting early diagnosis of intensive care unit-acquired weakness in septic patients using critical ultrasound and biological markers. BMC Anesthesiol 2025; 25:39. [PMID: 39863865 PMCID: PMC11761801 DOI: 10.1186/s12871-025-02911-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE Early diagnosis of intensive care unit-acquired weakness (ICUAW) is crucial for improving the outcomes of critically ill patients. Hence, this study was designed to identify predisposing factors for ICUAW and establish a predictive model for the early diagnosis of ICUAW. METHODS This prospective observational multicenter study included septic patients from the comprehensive ICUs of West China Hospital of Sichuan University and 10 other hospitals between September and November 2023. Inclusion criteria were as follows: age over 18 years; expected ICU stay longer than 3 days; and voluntary informed consent. Patients were classified into ICUAW (MRC score < 48) and non-ICUAW (MRC score ≥ 48) groups based on muscle strength assessments. The analyzed key predictive factors encompassed demographic data, SOFA and APACHE II scores, inflammatory markers (PCT, IL-6, and CRP), and ultrasound measurements of muscle thickness and cross-sectional area. Logistic regression analysis was conducted for variable selection and nomogram model construction. RESULTS A total of 116 septic patients were included, comprising 77 males and 39 females (mean age: 56.94 ± 19.90 years). A nomogram model predicting ICUAW probability was developed, which involved vastus intermedius diameter, rectus femoris cross-sectional area, IL-6, and CRP. The AUC of the composite diagnostic ROC curve was 0.966 (95%CI: 0.936 - 0.996), with a sensitivity of 88% and a specificity of 95.8%. CONCLUSIONS Conclusively, a nomogram model is constructed for diagnosing ICUAW in septic patients, which is simple and rapid and allows for visual representation, with excellent diagnostic capability.
Collapse
Affiliation(s)
- Ling Lei
- Department of Critical Care Medicine, West China Hospital, Sichuan University, 37 Guo Xue Xiang St, Chengdu, 610041, Sichuan, China
| | - Liang He
- Department of Respiratory and Critical Care Medicine, Xindu District People's Hospital, 199 Yuying Road South, Chengdu, 610500, Sichuan, China
| | - Tongjuan Zou
- Department of Critical Care Medicine, West China Hospital, Sichuan University, 37 Guo Xue Xiang St, Chengdu, 610041, Sichuan, China
| | - Jun Qiu
- Department of Critical Care Medicine, West China Hospital, Sichuan University, 37 Guo Xue Xiang St, Chengdu, 610041, Sichuan, China
| | - Yi Li
- Department of Critical Care Medicine, West China Hospital, Sichuan University, 37 Guo Xue Xiang St, Chengdu, 610041, Sichuan, China
| | - Ran Zhou
- Department of Critical Care Medicine, West China Hospital, Sichuan University, 37 Guo Xue Xiang St, Chengdu, 610041, Sichuan, China
| | - Yao Qin
- Department of Critical Care Medicine, West China Hospital, Sichuan University, 37 Guo Xue Xiang St, Chengdu, 610041, Sichuan, China
| | - Wanhong Yin
- Department of Critical Care Medicine, West China Hospital, Sichuan University, 37 Guo Xue Xiang St, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Zhang L, Wang J, Rong S, Dong H. Elucidating novel mechanism of action of spiperone for drug repurposing to prevent and treat murine colitis and sepsis. Life Sci 2025; 361:123268. [PMID: 39580139 DOI: 10.1016/j.lfs.2024.123268] [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: 08/23/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
AIMS While Ca2+ signaling plays a vital role in maintaining normal endothelial function and vascular activity, aberrant Ca2+ signaling in endothelial dysfunction is involved in the pathogenesis of inflammation. As a safe anti-psychotic drug to mobilize Ca2+ signaling, we repurposed spiperone as a potential drug for two intestinal epithelial injury related diseases, colitis and sepsis. MATERIALS AND METHODS Spiperone-induced vasorelaxation of human submucosal arterioles and mesenteric arterioles from wide-type and TRPV4 KO mice was determined by Mulvany-style wire myograph. The action of spiperone in HUVEC was tested by Ca2+ imaging and patch clamp, and its action on murine mesenteric arterioles was measured in vivo. LPS- and CLP-induced septic mice and DSS-induced colitic mice were used to examine the anti-inflammatory effects of spiperone. KEY FINDINGS Spiperone induced endothelium-dependent hyperpolarization (EDH)-mediated vasorelaxation of healthy arterioles with EC50 of ∼50 nM predominately via PLC/IP3/IP3R pathway to induce endoplasmic reticulum (ER) Ca2+ release and further to promote Ca2+ entry via TRPV4-constituted SOCE. In both LPS- and CLP-induced septic mice, spiperone effectively prevented and treated sepsis by reducing serum proinflammatory factors, alleviating multiple organ dysfunction, rescuing the impaired EDH-mediated vasorelaxation and improving murine survival rate. Similarly, spiperone could also protect against murine colitis. SIGNIFICANCE We reveal new action mode and mechanism of spiperone to induce EDH-mediated vasorelaxation of both human and murine arterioles to protect against colitis and sepsis by innovatively inducing PLC/IP3R/Ca2+ signaling rather than canonically antagonizing GPCR. Spiperone could be repurposed as a potential new drug for the prevention/treatment of colitis and sepsis.
Collapse
Affiliation(s)
- Luyun Zhang
- Department of Intensive Critical Care, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610000, Sichuan, China
| | - Jianxin Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Shaoya Rong
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China
| | - Hui Dong
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, #1 Ningde Road, Qingdao 266073, China.
| |
Collapse
|
8
|
Dalle S, Schouten M, Vanderbeke K, Van Parys E, Ramaekers M, Thomis M, Costamagna D, Koppo K. The CB1 antagonist Rimonabant improves muscle regeneration and remodels the inflammatory and endocannabinoid profile upon injury in male mice. Life Sci 2025; 361:123296. [PMID: 39645163 DOI: 10.1016/j.lfs.2024.123296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 11/12/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Skeletal muscle regeneration upon injury requires timely activation of inflammatory, myogenic, fibrotic, apoptotic and anabolic systems. Optimization of these features might improve the recovery process. Whereas recent data indicate that the endocannabinoid system, and more particularly cannabinoid receptor 1 (CB1) antagonism, is involved in the regulation of inflammatory, myogenic, fibrotic, apoptotic and anabolic pathways, it was never studied whether CB1 antagonism can improve muscle regeneration. The present study investigated the effect of the CB1 antagonist Rimonabant (10 mg/kg/d) on functional (5 days post-cardiotoxin injury; 5DPI) and molecular muscle responses (3DPI and 7DPI) in mice. Rimonabant prevented cardiotoxin-induced muscle strength loss 5DPI, increased myofiber growth (7DPI) and improved the muscle molecular profile 3DPI and 7DPI. In general, inflammation (e.g. p-p65NF-κB, CD80) and apoptosis (e.g. cleaved caspase-3, cleaved PARP) were downregulated by Rimonabant, whereas it upregulated the expression of Pax7 but other myogenic factors remained unaffected by rimonabant. In addition, Rimonabant restored the injury-induced (inflammatory) lipid profile to a large extent, including oxygenated fatty acids, unsaturated fatty acids and endocannabinoids such as 2-arachidonoyl glycerol and palmitoylethanolamide. Altogether, these data show that the endocannabinoid system might be a novel therapeutic target to improve muscle regeneration, which is relevant for age- and disease-related muscle degeneration.
Collapse
Affiliation(s)
- Sebastiaan Dalle
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium; MOVANT Research Group, Department of Rehabilitation Sciences and Physiotherapy, University of Antwerp, Antwerp, Belgium.
| | - Moniek Schouten
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium.
| | - Kaat Vanderbeke
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium.
| | - Evy Van Parys
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium
| | - Monique Ramaekers
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium.
| | - Martine Thomis
- Physical Activity, Sports & Health Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium
| | - Domiziana Costamagna
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium.
| | - Katrien Koppo
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Tervuursevest 101, 3001 Leuven, Belgium.
| |
Collapse
|
9
|
Yao H, Xie Q, Yang Y, Zhou C, Zeng Z, Zhang W. Melatonin attenuates sepsis-induced muscle atrophy by regulating the PI3K/Akt signaling pathway. Int Immunopharmacol 2025; 144:113619. [PMID: 39602954 DOI: 10.1016/j.intimp.2024.113619] [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: 08/26/2024] [Revised: 10/27/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND In intensive care units, sepsis-related muscle atrophy is a severe complication of numerous diseases, yet the underlying mechanism and potential therapeutic options remain elusive. Recent research has identified melatonin as a promising candidate for attenuating organ dysfunction triggered by sepsis. METHODS We used in vitro and in vivo models to simulate sepsis, C2C12 myotubes were treated with LPS, and the mice underwent cecal ligation and puncture (CLP) surgery. Following a pretreatment regimen involving melatonin and the AKT inhibitor MK-2206 2HCl, we analyzed changes in p-Akt and MuRF1 protein levels, fiber cross-sectional areas, and myotube diameters. The analyses included RNA sequencing, Western blotting, qRT-PCR, and immunofluorescence staining. RESULTS Activation of the PI3K/Akt pathway in skeletal muscle occurred 24 h post-CLP surgery in mice. This was accompanied by upregulated MuRF1 expression and reduced muscle fiber cross-sectional area, which culminated in muscle atrophy. However, these detrimental effects were attenuated when the mice were pretreated with melatonin via intraperitoneal injection for seven consecutive days. Similarly, LPS treatment of C2C12 myotubes activated the PI3K/Akt pathway, elevated MuRF1 expression, and markedly reduced myotube diameter after 48 h, leading to muscle atrophy. Pretreatment of C2C12 myotubes with melatonin 24 h in advance mitigated these adverse effects. However, cotreatment of C2C12 myotubes with melatonin and MK-2206 2HCl attenuated the beneficial effects of melatonin. CONCLUSION Melatonin can attenuate sepsis-induced muscle atrophy by regulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Huiming Yao
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Qian Xie
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yuting Yang
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Chaoqi Zhou
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Zhenguo Zeng
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
10
|
Li N, Lan J, Yang J, Ding H. Whole milk protein powder separated by low-temperature nanofiltration membrane administration alleviates sepsis-induced myopathy. Nutr Metab (Lond) 2024; 21:85. [PMID: 39456082 PMCID: PMC11515193 DOI: 10.1186/s12986-024-00862-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Sepsis-induced myopathy (SIM) has been recognized as a critical risk factor for the development of acquired muscle weakness among patients in the intensive care unit. These individuals frequently encounter inadequate dietary intake and malnutrition. With the aggravation of the severity of the person's condition, leading to increased skeletal muscle protein breakdown and reduced synthesis, which is an urgent problem to be solved in clinical nutritional treatment. Whole milk protein powder (WMPP) has promising bioactive nutrients and holds promising potential for enhancing skeletal muscle mass. The study was designed to delve into the potential effects and mechanisms of WMPP intervention for increaseing skeletal muscle mass on SIM mice. Our results clearly show that the intervention with WMPP can significantly improve the exercise capacity and skeletal muscle mass in SIM mice. It significantly increases the diameter and cross-sectional area (CSA) of skeletal muscle fibers, while effectively reducing the excessive aggregation of collagen fibers and the abnormal accumulation of adipose tissue in the skeletal muscle of SIM mice. Moreover, WMPP intervention also significantly alleviated the oxidative stress status of mitochondria, which subsequently enhanced the expression of mitochondrial metabolic enzymes. The mechanism may be associated with decreased AMPK phosphorylation in skeletal muscle tissue and simultaneously increased phosphorylation of mTOR, p70S6K1, and 4EBP-1 in SIM mice. In summary, the WMPP intervention significantly enhances exercise capacity and skeletal muscle mass while mitigating the oxidative stress status of mitochondria. Furthermore, it regulates skeletal muscle anabolism via the AMPK/mTOR signaling pathway in SIM mice.
Collapse
Affiliation(s)
- Na Li
- School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, Ningxia, 750004, China
- General Hospital of Ningxia Medical University, 804 Shengli Street, Xingqing District, Yinchuan, Ningxia, 750003, China
| | - Junyu Lan
- School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, Ningxia, 750004, China
| | - Jianjun Yang
- School of Public Health, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, Ningxia, 750004, China.
| | - Huan Ding
- General Hospital of Ningxia Medical University, 804 Shengli Street, Xingqing District, Yinchuan, Ningxia, 750003, China.
| |
Collapse
|
11
|
Yu Q, Song J, Yang L, Miao Y, Xie L, Ma X, Xie P, Chen S. A scoping review of preclinical intensive care unit-acquired weakness models. Front Physiol 2024; 15:1423567. [PMID: 39416383 PMCID: PMC11480018 DOI: 10.3389/fphys.2024.1423567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Background Animal models focusing on neuromuscular outcomes are crucial for understanding the mechanisms of intensive care unit-acquired weakness (ICU-AW) and exploring potential innovative prevention and treatment strategies. Aim To analyse and evaluate preclinical ICU-AW models. Methods We manually searched five English and four Chinese databases from 1 January 2002, to 1 February 2024, and reviewed related study references. Full-text publications describing animal models of muscle weakness and atrophy in critical illness were included. Detailed information about model types, animal species, sex, age, induction methods, outcome measures, drawbacks and strengths was extracted from each included study. Results A total of 3,451 citations were initially retrieved, with 84 studies included in the final analysis. The most frequently studied animal model included rodents (86.9%), 64.3% of which were male animals. ICU-AW animal models were mostly induced by comprehensive intensive care unit (ICU) interventions (38.1%) and sepsis (51.2%). Most studies focused on limb muscles (66.7%), diaphragm muscles (21.4%) or both (9.5%). Reported outcomes primarily included muscular pathological changes (83.3%), electrophysiological examinations of muscles (57.1%) and animal grip strength (16.6%). However, details such as animal age, mortality data, experimental design, randomisation, blinding, sample size and interventions for the experimental group and/or control group were inadequately reported. Conclusion Many preclinical models are used to study ICU-AW, but the reporting of methodological details is often incomplete. Although current ICU animal models can mimic the characteristics of human ICU-AW, there is no standard model. Future preclinical studies should develop a standard ICU-AW animal model to enhance reproducibility and improve scientific rigor in exploring the mechanisms and potential treatment of ICU-AW.
Collapse
Affiliation(s)
- Qingmei Yu
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- School of Nursing, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiamei Song
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- School of Nursing, Zunyi Medical University, Zunyi, Guizhou, China
| | - Luying Yang
- School of Nursing, Zunyi Medical University, Zunyi, Guizhou, China
- Department of Critical Care Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People’s Hospital of Zunyi City, Zunyi, Guizhou, China
| | - Yanmei Miao
- School of Nursing, Zunyi Medical University, Zunyi, Guizhou, China
- Department of Critical Care Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People’s Hospital of Zunyi City, Zunyi, Guizhou, China
| | - Leiyu Xie
- Department of Critical Care Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People’s Hospital of Zunyi City, Zunyi, Guizhou, China
| | - Xinglong Ma
- Department of Critical Care Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People’s Hospital of Zunyi City, Zunyi, Guizhou, China
| | - Peng Xie
- Department of Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaolin Chen
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- School of Nursing, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
12
|
Li H, Yu R, Zhou Q, Song J, Zhou Q, Ma W, Wang J, Hu Z, Yang Q, Chen K. TRENDS AND OUTCOMES IN SEPSIS HOSPITALIZATIONS WITH AND WITHOUT ACUTE KIDNEY INJURY: A NATIONWIDE INPATIENT ANALYSIS. Shock 2024; 62:470-479. [PMID: 38888575 DOI: 10.1097/shk.0000000000002386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
ABSTRACT Background: Despite rapid advances in treatment, sepsis currently remains a major public health challenge worldwide. Over the past several years, there has been an increase in the clinical incidence of sepsis, as well as an increase in hospitalization rates, which bear the majority of the economic burden associated with sepsis. Sepsis is a public health burden due to the high fatality rates and accompanying morbidity. However, the sepsis-related mortality rates have fallen steadily over the years. One of the most common organs to fail in patients with sepsis is the kidney, and acute kidney injury (AKI) is associated with high mortality rates. This study's primary goal was to assess the impact of AKI on the evolution and outcome of hospitalization of patients with sepsis. Methods: Adults (≥18 years) hospitalized for sepsis in the United States between 2010 and 2019 were retrospectively analyzed using the nationally representative Nationwide Inpatient Sample database. Sepsis and AKI were defined using the codes of the International Classification of Diseases, Ninth Revision, Clinical Modification and the International Classification of Diseases, Tenth Revision, Clinical Modification. Results: Of the 4,258,360 outcomes, 3,946,048 met the inclusion criteria. The prevalence of AKI among sepsis inpatients increased from 39.10% in 2010 to 41% in 2019, but the impact of AKI on mortality declined over time, with in-hospital mortality from AKI among sepsis inpatients decreasing from 26.30% in 2010 to 16.30% in 2019. Hospitalizations linked to AKI were substantially more likely to involve infection sites such as the urinary tract, gastrointestinal tract, and endocarditis. Numerous pathogenic floras, including Escherichia coli , Staphylococcus aureus , Streptococcal , Enterococcus , and Pseudomonas , had greater rates among sepsis-related contacts with AKI. Furthermore, compared to hospitalization without comorbid AKI, the median total hospital charges and length of stay days for sepsis hospitalization with comorbid AKI were greater. Conclusion: With time, patients with sepsis have a higher frequency of AKI and a corresponding decline in mortality.
Collapse
Affiliation(s)
- Haibo Li
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, Inner Mongolia, China
| | - Ran Yu
- Department of Anesthesiology, Chifeng Clinical Medical College of Inner Mongolia Medical University, Chifeng, Inner Mongolia, China
| | - Qi Zhou
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiannan Song
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, Inner Mongolia, China
| | - Qi Zhou
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, Inner Mongolia, China
| | - Wanli Ma
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, Inner Mongolia, China
| | - Jian Wang
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhanfei Hu
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, Inner Mongolia, China
| | - Qinfeng Yang
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Keyuan Chen
- Division of Spine Surgery, Department of Orthopaedics, Guangzhou First People's Hospital, School of Medicine South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Hu Y, Tang J, Xu Q, Fang Z, Li R, Yang M, Zhao J, Chen X. Role of pyruvate kinase M2 in regulating sepsis (Review). Mol Med Rep 2024; 30:185. [PMID: 39155878 DOI: 10.3892/mmr.2024.13309] [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: 06/11/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024] Open
Abstract
Glycolysis occurs in all living organisms as a form of energy supply. Pyruvate kinase M2 (PKM2) is one of the rate‑limiting enzymes in the glycolytic process. PKM2 is considered to serve an important role in several terminal diseases, including sepsis. However, to the best of our knowledge, the specific mechanistic role of PKM2 in sepsis remains to be systematically summarised. Therefore, the present review aims to summarise the roles of PKM2 in sepsis progression. In addition, potential treatment strategies for patients with sepsis are discussed. The present review hopes to lay the groundwork for studying the role of PKM2 and developing therapeutic strategies against metabolic disorders that occur during sepsis.
Collapse
Affiliation(s)
- Yifei Hu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Jing Tang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Qiao Xu
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Zenghui Fang
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| | - Rongqing Li
- Department of Clinical Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Mengxuan Yang
- Department of Clinical Laboratory, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310000, P.R. China
| | - Jie Zhao
- Department of Clinical Medicine, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225000, P.R. China
| | - Xin Chen
- Department of Clinical Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang 321000, P.R. China
| |
Collapse
|
14
|
Fang L, Hu F, Li H, Chang W, Liu L. Efficacy and safety of mesenchymal stem cell therapy for acute respiratory distress syndrome-a systematic review and meta-analysis. J Thorac Dis 2024; 16:5802-5814. [PMID: 39444918 PMCID: PMC11494583 DOI: 10.21037/jtd-24-281] [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: 02/21/2024] [Accepted: 07/26/2024] [Indexed: 10/25/2024]
Abstract
Background Mesenchymal stem cells (MSC) therapy for acute respiratory distress syndrome (ARDS) represents a burgeoning treatment approach, supported by numerous preclinical studies confirming its efficacy. Our study aims to provide a comprehensive evaluation of both the safety and effectiveness of MSC. Methods We conducted searches across three databases (PubMed, Embase, Cochrane) for randomized controlled studies up to June 23, 2024. A meta-analysis was performed on variables including adverse events, mortality, changes in the PaO2/FiO2 ratio, intensive care unit (ICU), length of stay, ventilation-free days, and changes in pro-inflammatory and anti-inflammatory cytokines. Relative risk (RR) values were employed for dichotomous variables, while mean difference (MD) and standard mean difference (SMD) were used for continuous variables. Risk bias was assessed using risk of bias 2 (ROB2). Results The meta-analysis encompassed 17 experiments involving 796 patients, with 410 undergoing MSC treatment and 386 in the control group. Primary outcomes indicated that MSC treatment did not escalate adverse events [RR =1.04; 95% confidence interval (CI): 0.90, 1.19; P=0.59; I2=0%]. On the contrary, it significantly diminishes the mortality (RR =0.79; 95% CI: 0.64, 0.97; P=0.02; I2=0%). Regarding secondary outcomes, MSCs led to a significant improvement in the PaO2/FiO2 ratio for ARDS patients (SMD =0.53; 95% CI: 0.15, 0.92; P=0.007; I2=0%). However, there were no significant differences in ICU length of stay (MD =-1.77; 95% CI: -6.97, 3.43; P=0.50; I2=63%) and ventilation-free days (MD =-1.29; 95% CI: -4.09, 1.51; P=0.37; I2=0%). MSCs significantly lowered C-reactive protein (CRP) (SMD =-0.65; 95% CI: -1.18, -0.13; P=0.01; I2=56%) and interleukin-6 (IL-6) levels compared to the control group (SMD =-0.76; 95% CI: -1.34, -0.17; P=0.01; I2=74%). However, changes in interleukin-10 (AIL-10) (SMD =-0.46; 95% CI: -1.51, 0.58; P=0.38; I2=77%), and changes in tumor necrosis factor-alpha (ATNF-α) (SMD =-1.5; 95% CI: -3.39, 0.40; P=0.12; I2=92%) levels showed no significant changes. Conclusions MSC therapy demonstrates reliable safety, with a significant impact on reducing mortality and improving certain clinical symptoms. Moreover, in certain aspects, it may alleviate the inflammatory response in ARDS. Nonetheless, these findings necessitate validation through additional high-quality randomized controlled trials.
Collapse
Affiliation(s)
- Lingyan Fang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Fangyuan Hu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Han Li
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Wei Chang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ling Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
15
|
Su J, Chen W, Zhou F, Li R, Tong Z, Wu S, Ye Z, Zhang Y, Lin B, Yu X, Guan B, Feng Z, Chen K, Chen Q, Chen L. Inhibitory mechanisms of decoy receptor 3 in cecal ligation and puncture-induced sepsis. mBio 2024; 15:e0052124. [PMID: 38700314 PMCID: PMC11237498 DOI: 10.1128/mbio.00521-24] [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: 02/21/2024] [Accepted: 04/02/2024] [Indexed: 05/05/2024] Open
Abstract
Despite its high mortality, specific and effective drugs for sepsis are lacking. Decoy receptor 3 (DcR3) is a potential biomarker for the progression of inflammatory diseases. The recombinant human DcR3-Fc chimera protein (DcR3.Fc) suppresses inflammatory responses in mice with sepsis, which is critical for improving survival. The Fc region can exert detrimental effects on the patient, and endogenous peptides are highly conducive to clinical application. However, the mechanisms underlying the effects of DcR3 on sepsis are unknown. Herein, we aimed to demonstrate that DcR3 may be beneficial in treating sepsis and investigated its mechanism of action. Recombinant DcR3 was obtained in vitro. Postoperative DcR3 treatment was performed in mouse models of lipopolysaccharide- and cecal ligation and puncture (CLP)-induced sepsis, and their underlying molecular mechanisms were explored. DcR3 inhibited sustained excessive inflammation in vitro, increased the survival rate, reduced the proinflammatory cytokine levels, changed the circulating immune cell composition, regulated the gut microbiota, and induced short-chain fatty acid synthesis in vivo. Thus, DcR3 protects against CLP-induced sepsis by inhibiting the inflammatory response and apoptosis. Our study provides valuable insights into the molecular mechanisms associated with the protective effects of DcR3 against sepsis, paving the way for future clinical studies. IMPORTANCE Sepsis affects millions of hospitalized patients worldwide each year, but there are no sepsis-specific drugs, which makes sepsis therapies urgently needed. Suppression of excessive inflammatory responses is important for improving the survival of patients with sepsis. Our results demonstrate that DcR3 ameliorates sepsis in mice by attenuating systematic inflammation and modulating gut microbiota, and unveil the molecular mechanism underlying its anti-inflammatory effect.
Collapse
Affiliation(s)
- Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Wenzhi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
- Institute of Edible Fungi, Fujian Academy of Agricultural Sciences, Fuzhou, Fujian, China
| | - Fen Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Rui Li
- Department of Neurosurgery & Neurocritical Care, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiyong Tong
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Shun Wu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Zhen Ye
- Department of Neurosurgery & Neurocritical Care, Huashan Hospital, Fudan University, Shanghai, China
| | - Yichao Zhang
- Department of Neurosurgery & Neurocritical Care, Huashan Hospital, Fudan University, Shanghai, China
| | - Ben Lin
- Department of Neurosurgery & Neurocritical Care, Huashan Hospital, Fudan University, Shanghai, China
| | - Xing Yu
- Department of Gastroenterology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Biyun Guan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Zhihua Feng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Kunsen Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou, China
| | - Long Chen
- Department of Neurosurgery & Neurocritical Care, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Peng M, Grootaert C, Vercauteren M, Boon N, Janssen C, Rajkovic A, Asselman J. Probing Long-Term Impacts: Low-Dose Polystyrene Nanoplastics Exacerbate Mitochondrial Health and Evoke Secondary Glycolysis via Repeated and Single Dosing. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:9967-9979. [PMID: 38814788 DOI: 10.1021/acs.est.3c10868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Nanoplastics (NPs) are omnipresent in the environment and contribute to human exposure. However, little is known regarding the long-term effects of NPs on human health. In this study, human intestinal Caco-2 cells were exposed to polystyrene nanoplastics (nanoPS) in an environmentally relevant concentration range (102-109 particles/mL) under two realistic exposure scenarios. In the first scenario, cells were repeatedly exposed to nanoPS every 2 days for 12 days to study the long-term effects. In the second scenario, only nanoPS was added once and Caco-2 cells were cultured for 12 days to study the duration of the initial effects of NPs. Under repeated dosing, initial subtle effects on mitochondria induced by low concentrations would accrue over consistent exposure to nanoPS and finally lead to significant impairment of mitochondrial respiration, mitochondrial mass, and cell differentiation process at the end of prolonged exposure, accompanied by significantly increased glycolysis over the whole exposure period. Single dosing of nanoPS elicited transient effects on mitochondrial and glycolytic functions, as well as increased reactive oxygen species (ROS) production in the early phase of exposure, but the self-recovery capacity of cells mitigated these effects at intermediate culture times. Notably, secondary effects on glycolysis and ROS production were observed during the late culture period, while the cell differentiation process and mitochondrial mass were not affected at the end. These long-term effects are of crucial importance for comprehensively evaluating the health hazards arising from lifetime exposure to NPs, complementing the extensively observed acute effects associated with prevalent short-term exposure to high concentrations. Our study underlines the need to study the toxicity of NPs in realistic long-term exposure scenarios such as repeated dosing.
Collapse
Affiliation(s)
- Miao Peng
- Laboratory of Environmental Toxicology and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
- Blue Growth Research Lab, Ghent University, Wetenschapspark 1, 8400 Oostende Belgium
| | - Charlotte Grootaert
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Maaike Vercauteren
- Laboratory of Environmental Toxicology and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
- Blue Growth Research Lab, Ghent University, Wetenschapspark 1, 8400 Oostende Belgium
| | - Nico Boon
- Center for Microbial Technology and Ecology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Colin Janssen
- Laboratory of Environmental Toxicology and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
- Blue Growth Research Lab, Ghent University, Wetenschapspark 1, 8400 Oostende Belgium
| | - Andreja Rajkovic
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Jana Asselman
- Laboratory of Environmental Toxicology and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
- Blue Growth Research Lab, Ghent University, Wetenschapspark 1, 8400 Oostende Belgium
| |
Collapse
|
17
|
Genserová L, Duška F, Krajčová A. β-hydroxybutyrate exposure restores mitochondrial function in skeletal muscle satellite cells of critically ill patients. Clin Nutr 2024; 43:1250-1260. [PMID: 38653008 DOI: 10.1016/j.clnu.2024.04.009] [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: 09/15/2023] [Revised: 03/17/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND & AIM Dysfunction of skeletal muscle satellite cells might impair muscle regeneration and prolong ICU-acquired weakness, a condition associated with disability and delayed death. This study aimed to elucidate the distinct metabolic effects of critical illness and β-OH-butyrate on satellite cells isolated from these patients. METHODS Satellite cells were extracted from vastus lateralis muscle biopsies of patients with ICU-acquired weakness (n = 10) and control group of healthy volunteers or patients undergoing elective hip replacement surgery (n = 10). The cells were exposed to standard culture media supplemented with β-OH-butyrate to assess its influence on cell proliferation by ELISA, mitochondrial functions by extracellular flux analysis, electron transport chain complexes by high resolution respirometry, and ROS production by confocal microscopy. RESULTS Critical illness led to a decline in maximal respiratory capacity, ATP production and glycolytic capacity and increased ROS production in ICU patients' cells. Notably, the function of complex II was impaired due to critical illness but restored to normal levels upon exposure to β-OH-butyrate. While β-OH-butyrate significantly reduced ROS production in both control and ICU groups, it had no significant impact on global mitochondrial functions. CONCLUSION Critical illness induces measurable bioenergetic dysfunction of skeletal muscle satellite cells. β-OH-butyrate displayed a potential in rectifying complex II dysfunction caused by critical illness and this warrants further exploration.
Collapse
Affiliation(s)
- Lucie Genserová
- Department of Internal Medicine of the Third Faculty of Medicine, Královské Vinohrady University Hospital, Charles University, Prague, Czech Republic; Department of Anaesthesia and Intensive Care of the Third Faculty of Medicine, Královské Vinohrady University Hospital, OXYLAB-Laboratory for Mitochondrial Physiology, Charles University, Prague, Czech Republic
| | - František Duška
- Department of Anaesthesia and Intensive Care of the Third Faculty of Medicine, Královské Vinohrady University Hospital, OXYLAB-Laboratory for Mitochondrial Physiology, Charles University, Prague, Czech Republic
| | - Adéla Krajčová
- Department of Anaesthesia and Intensive Care of the Third Faculty of Medicine, Královské Vinohrady University Hospital, OXYLAB-Laboratory for Mitochondrial Physiology, Charles University, Prague, Czech Republic.
| |
Collapse
|
18
|
Garvey M. Hospital Acquired Sepsis, Disease Prevalence, and Recent Advances in Sepsis Mitigation. Pathogens 2024; 13:461. [PMID: 38921759 PMCID: PMC11206921 DOI: 10.3390/pathogens13060461] [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: 05/04/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection, commonly associated with nosocomial transmission. Gram-negative bacterial species are particularly problematic due to the release of the lipopolysaccharide toxins upon cell death. The lipopolysaccharide toxin of E. coli has a greater immunogenic potential than that of other Gram-negative bacteria. The resultant dysregulation of the immune system is associated with organ failure and mortality, with pregnant women, ICU patients, and neonates being particularly vulnerable. Additionally, sepsis recovery patients have an increased risk of re-hospitalisation, chronic illness, co-morbidities, organ damage/failure, and a reduced life expectancy. The emergence and increasing prevalence of antimicrobial resistance in bacterial and fungal species has impacted the treatment of sepsis patients, leading to increasing mortality rates. Multidrug resistant pathogens including vancomycin-resistant Enterococcus, beta lactam-resistant Klebsiella, and carbapenem-resistant Acinetobacter species are associated with an increased risk of mortality. To improve the prognosis of sepsis patients, predominantly high-risk neonates, advances must be made in the early diagnosis, triage, and control of sepsis. The identification of suitable biomarkers and biomarker combinations, coupled with machine learning and artificial intelligence, show promise in early detection protocols. Rapid diagnosis of sepsis in patients is essential to inform on clinical treatment, especially with resistant infectious agents. This timely review aims to discuss sepsis prevalence, aetiology, and recent advances towards disease mitigation and control.
Collapse
Affiliation(s)
- Mary Garvey
- Department of Life Science, Atlantic Technological University, F91 YW50 Sligo, Ireland; ; Tel.: +353-0719-305-529
- Centre for Precision Engineering, Materials and Manufacturing Research (PEM), Atlantic Technological University, F91 YW50 Sligo, Ireland
| |
Collapse
|
19
|
Lessard L, Saugues A, Gondin J, Mounier R, Kneppers A. Measurement of Myonuclear Accretion In Vitro and In Vivo. Methods Mol Biol 2024. [PMID: 38647863 DOI: 10.1007/7651_2024_540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Adult skeletal muscle stem cells (MuSC) are the regenerative precursors of myofibers and also have an important role in myofiber growth, adaptation, and maintenance by fusing to the myofibers-a process referred to as "myonuclear accretion." Due to a focus on MuSC function during regeneration, myofibers remain a largely overlooked component of the MuSC niche influencing MuSC fate. Here, we describe a method to directly measure the rate of myonuclear accretion in vitro and in vivo using ethynyl-2'-deoxyuridine (EdU)-based tracing of MuSC progeny. This method supports the dissection of MuSC intrinsic and myofiber-derived factors influencing myonuclear accretion as an alternative fate of MuSCs supporting myofiber homeostasis and plasticity.
Collapse
Affiliation(s)
- Lola Lessard
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Audrey Saugues
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France
| | - Anita Kneppers
- Institut NeuroMyoGène, Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR5261, INSERM U1315, Lyon, France.
| |
Collapse
|
20
|
Le Stang V, Latronico N, Dres M, Bertoni M. Critical illness-associated limb and diaphragmatic weakness. Curr Opin Crit Care 2024; 30:121-130. [PMID: 38441088 PMCID: PMC10919276 DOI: 10.1097/mcc.0000000000001135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW In the current review, we aim to highlight the evolving evidence on the diagnosis, prevention and treatment of critical illness weakness (CIW) and critical illness associated diaphragmatic weakness (CIDW). RECENT FINDINGS In the ICU, several risk factors can lead to CIW and CIDW. Recent evidence suggests that they have different pathophysiological mechanisms and impact on outcomes, although they share common risk factors and may overlap in several patients. Their diagnosis is challenging, because CIW diagnosis is primarily clinical and, therefore, difficult to obtain in the ICU population, and CIDW diagnosis is complex and not easily performed at the bedside. All of these issues lead to underdiagnosis of CIW and CIDW, which significantly increases the risk of complications and the impact on both short and long term outcomes. Moreover, recent studies have explored promising diagnostic techniques that are may be easily implemented in daily clinical practice. In addition, this review summarizes the latest research aimed at improving how to prevent and treat CIW and CIDW. SUMMARY This review aims to clarify some uncertain aspects and provide helpful information on developing monitoring techniques and therapeutic interventions for managing CIW and CIDW.
Collapse
Affiliation(s)
- Valentine Le Stang
- Sorbonne Université, INSERM, UMRS1158 Neurophysiologie respiratoire expérimentale et clinique
- AP-HP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Médecine Intensive –Réanimation (Département ‘R3S’), Paris, France
| | - Nicola Latronico
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia
- Department of Emergency, ASST Spedali Civili University Hospital, Piazzale Ospedali Civili, 1, 25123 Brescia, Italy
- ‘Alessandra BONO’ Interdepartmental University Research Center on LOng Term Outcome (LOTO) in Critical Illness Survivors, University of Brescia, Brescia, Italy
| | - Martin Dres
- Sorbonne Université, INSERM, UMRS1158 Neurophysiologie respiratoire expérimentale et clinique
- AP-HP. Sorbonne Université, Hôpital Pitié-Salpêtrière, Service de Médecine Intensive –Réanimation (Département ‘R3S’), Paris, France
| | - Michele Bertoni
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia
- Department of Emergency, ASST Spedali Civili University Hospital, Piazzale Ospedali Civili, 1, 25123 Brescia, Italy
| |
Collapse
|
21
|
Sheng Z, Yu Z, Wang M, Zhou R, Chen S, Yu X, Li F. Targeting STAT6 to mitigate sepsis-induced muscle atrophy and weakness: Modulation of mitochondrial dysfunction, ferroptosis, and CHI3L1-Mediated satellite cell loss. Biochem Biophys Rep 2024; 37:101608. [PMID: 38188367 PMCID: PMC10770525 DOI: 10.1016/j.bbrep.2023.101608] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/18/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Sepsis-induced muscle weakness is a debilitating consequence of prolonged critical illness, often associated with a poor prognosis. While recent research has shown that STAT6 functions as an inhibitor of myogenesis, its role in sepsis-induced muscle weakness remains unclear. In this study, we hypothesized that inhibiting STAT6 could attenuate sepsis-induced muscle atrophy and weakness, and we explored the underlying mechanisms. Leveraging a microarray dataset from sepsis patients, we identified significant enrichment of genes related to muscle function, ferroptosis, and the p53 signalling pathway in muscle tissue from sepsis patients. Using a murine sepsis model induced by cecum ligation and puncture (CLP), we explore the multifaceted role of STAT6 inhibition. Our findings demonstrate that STAT6 inhibition effectively attenuates muscle atrophy, enhances grip strength, preserves mitochondrial integrity, and modulates ferroptosis in septic mice. Additionally, we identify elevated levels of CHI3L1 in septic muscle tissue, which are significantly reduced by STAT6 inhibition. In-depth analysis of primary muscle satellite cells reveals that CHI3L1 overexpression is associated with increased expression of key regulators of satellite cell myogenicity, while negatively impacting cell viability. Silencing CHI3L1 expression mitigates satellite cell injury and loss, highlighting its pivotal role in sepsis-induced muscle damage. In summary, this study unveils the potential of STAT6 as a therapeutic target for mitigating sepsis-induced muscle atrophy and weakness. Our findings underscore the regulation of mitochondrial dysfunction, ferroptosis, and CHI3L1-mediated satellite cell damage by STAT6, offering promising avenues for therapeutic intervention in the management of sepsis-induced muscle weakness.
Collapse
Affiliation(s)
- Zhiyong Sheng
- Department of Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- Department of Neurological Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhihong Yu
- Department of Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Meng Wang
- Department of Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Rui Zhou
- Department of Neurological Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shenjian Chen
- Department of Neurological Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xin Yu
- Department of Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Fuxing Li
- Department of Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
22
|
Huang W, Wang B, Ou Q, Zhang X, He Y, Mao X, Wei X, Kou X. ASC-expressing pyroptotic extracellular vesicles alleviate sepsis by protecting B cells. Mol Ther 2024; 32:395-410. [PMID: 38093517 PMCID: PMC10861962 DOI: 10.1016/j.ymthe.2023.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Pyroptosis is an inflammatory programmed cell death process characterized by membrane rupture. Interestingly, pyroptotic cells can generate plenty of nanosized vesicles. Non-inflammatory apoptotic cell death-derived apoptotic vesicles (apoVs) were systemically characterized and displayed multiple physiological functions and therapeutic potentials. However, the characteristics of pyroptotic cell-generated extracellular vesicles (EVs) are largely unknown. Here, we identified a group of pyroptotic EVs (pyroEVs) from in vitro cultured pyroptotic mesenchymal stem cells (MSCs), as well as from septic mouse blood. Compared with apoVs, pyroEVs express similar levels of annexin V, calreticulin, and common EV markers, but express a decreased level of apoptotic marker cleave caspase-3. PyroEVs, but not apoVs and exosomes, specifically express pyroptotic maker apoptosis-associated speck-like protein containing CARD (ASC). More importantly, MSC-derived pyroEVs protect B cells in the spleen and bone marrow to relieve inflammatory responses and enhance the survival rate of the septic mice. Mechanistically, pyroEV membrane-expressed ASC binds to B cells to repress cell death by repressing Toll-like receptor 4. This study uncovered the characteristics of pyroEVs and their therapeutic role in sepsis and B cell-mediated immune response.
Collapse
Affiliation(s)
- Weiying Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Ben Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Qianmin Ou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xiao Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Department of Prosthodontics, Peking University School and Hospital of Stomatology and National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Yifan He
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, China.
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China.
| |
Collapse
|
23
|
Murray KO, Brant JO, Spradlin RA, Thome T, Laitano O, Ryan TE, Riva A, Kladde MP, Clanton TL. Exertional heat stroke causes long-term skeletal muscle epigenetic reprogramming, altered gene expression, and impaired satellite cell function in mice. Am J Physiol Regul Integr Comp Physiol 2024; 326:R160-R175. [PMID: 38047316 PMCID: PMC11283893 DOI: 10.1152/ajpregu.00226.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/26/2023] [Accepted: 11/26/2023] [Indexed: 12/05/2023]
Abstract
The effect of exertional heat stroke (EHS) exposure on skeletal muscles is incompletely understood. Muscle weakness is an early symptom of EHS but is not considered a major target of multiorgan injury. Previously, in a preclinical mouse model of EHS, we observed the vulnerability of limb muscles to a second EHS exposure, suggesting hidden processes contributing to declines in muscle resilience. Here, we evaluated the possible molecular origins of EHS-induced declines in muscle resilience. Female C57BL/6 mice [total n = 56; 28/condition, i.e., EHS and exercise control (EXC)] underwent forced wheel running at 37.5°C/40% relative humidity until symptom limitation (unconsciousness). EXC mice exercised identically at room temperature (22-23°C). After 1 mo of recovery, the following were assessed: 1) specific force and caffeine-induced contracture in soleus (SOL) and extensor digitorum longus (EDL) muscles; 2) transcriptome and DNA methylome responses in gastrocnemius (GAST); and 3) primary satellite cell function (proliferation and differentiation). There were no differences in specific force in either SOL or EDL from EXC. Only EHS solei exhibited lower caffeine sensitivity. EHS GAST exhibited higher RNA expression of genes encoding structural proteins of slow fibers, heat shock proteins, and myogenesis. A total of ∼2,500 differentially methylated regions of DNA that could potentially affect many cell functions were identified. Primary satellite cells exhibited suppressed proliferation rates but normal differentiation responses. Results demonstrate long-term changes in skeletal muscles 1 mo after EHS that could contribute to declines in muscle resilience. Skeletal muscle may join other, more recognized tissues considered vulnerable to long-term effects of EHS.NEW & NOTEWORTHY Exertional heat stroke (EHS) in mice induces long-term molecular and functional changes in limb muscle that could reflect a loss of "resilience" to further stress. The phenotype was characterized by altered caffeine sensitivity and suppressed satellite cell proliferative potential. This was accompanied by changes in gene expression and DNA methylation consistent with ongoing muscle remodeling and stress adaptation. We propose that EHS may induce a prolonged vulnerability of skeletal muscle to further stress or injury.
Collapse
Affiliation(s)
- Kevin O Murray
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, United States
| | - Jason O Brant
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, United States
| | - Ray A Spradlin
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, United States
| | - Trace Thome
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, United States
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, United States
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, United States
| | - Alberto Riva
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, United States
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, United States
| | - Michael P Kladde
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida, United States
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, United States
| | - Thomas L Clanton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, Florida, United States
| |
Collapse
|
24
|
Chen J, Huang M. Intensive care unit-acquired weakness: Recent insights. JOURNAL OF INTENSIVE MEDICINE 2024; 4:73-80. [PMID: 38263973 PMCID: PMC10800771 DOI: 10.1016/j.jointm.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/16/2023] [Accepted: 07/07/2023] [Indexed: 01/25/2024]
Abstract
Intensive care unit-acquired weakness (ICU-AW) is a common complication in critically ill patients and is associated with a variety of adverse outcomes. These include the need for prolonged mechanical ventilation and ICU stay; higher ICU, in-hospital, and 1-year mortality; and increased in-hospital costs. ICU-AW is associated with multiple risk factors including age, underlying disease, severity of illness, organ failure, sepsis, immobilization, receipt of mechanical ventilation, and other factors related to critical care. The pathological mechanism of ICU-AW remains unclear and may be considerably varied. This review aimed to evaluate recent insights into ICU-AW from several aspects including risk factors, pathophysiology, diagnosis, and treatment strategies; this provides new perspectives for future research.
Collapse
Affiliation(s)
- Juan Chen
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Man Huang
- Department of General Intensive Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| |
Collapse
|
25
|
Chen J, Chen W, Wu L, Wang RH, Xiang JJ, Zheng FK, Huang QM. Causal relationships between plasma lipids and sepsis: A Mendelian randomization study. Medicine (Baltimore) 2023; 102:e36288. [PMID: 38065901 PMCID: PMC10713144 DOI: 10.1097/md.0000000000036288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Although observational studies have indicated that plasma lipids are associated with an increased risk of sepsis, due to confounders and reverse causality, the causal relationship remains unclear. This study was designed to assess the causal effects of plasma lipid levels on sepsis. We used a 2-sample Mendelian randomization (MR) method to evaluate the causal effect of plasma lipids on sepsis. MR analysis employs methods such as inverse variance weighted, MR-Egger regression, weighted median regression (WME), simple mode and weighted mode. The inverse variance weighted (IVW) method was predominantly utilized to assess causality. Heterogeneity was affirmed by Cochran Q test, while pleiotropy was corroborated by MR-Egger regression analysis. The robustness and reliability of the results were demonstrated through "leave-one-out" sensitivity analysis. Instrumental variables included 226 single-nucleotide polymorphisms (SNPs), comprising of 7 for triglyceride (TG), 169 for high-density lipoprotein cholesterol (HDL-C), and 50 for low-density lipoprotein cholesterol (LDL-C). The risk of sepsis appeared to increase with rising LDL-C levels, as indicated by the inverse variance weighted analysis (OR 1.11, 95% CI from0.99 to1.24, P = 0.068). However, no causality existed between LDL-C, HDL-C, TG and sepsis. Two-sample MR analysis indicated that increased LDL-C level is a risk factor for sepsis, while TG and HDL-C levels have protective effects against sepsis. However, no significant causal relationship was found between TG, HDL-C, and LDL-C levels and sepsis.
Collapse
Affiliation(s)
- Jing Chen
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Wei Chen
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Lin Wu
- Guangxi University of Chinese Medicine, Nanning, China
| | - Rong Hui Wang
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Jun Jun Xiang
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Fu Kui Zheng
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| | - Qiao Ming Huang
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, China
| |
Collapse
|
26
|
Zhang W, Jiang H, Wu G, Huang P, Wang H, An H, Liu S, Zhang W. The pathogenesis and potential therapeutic targets in sepsis. MedComm (Beijing) 2023; 4:e418. [PMID: 38020710 PMCID: PMC10661353 DOI: 10.1002/mco2.418] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as "a life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection." At present, sepsis continues to pose a grave healthcare concern worldwide. Despite the use of supportive measures in treating traditional sepsis, such as intravenous fluids, vasoactive substances, and oxygen plus antibiotics to eradicate harmful pathogens, there is an ongoing increase in both the morbidity and mortality associated with sepsis during clinical interventions. Therefore, it is urgent to design specific pharmacologic agents for the treatment of sepsis and convert them into a novel targeted treatment strategy. Herein, we provide an overview of the molecular mechanisms that may be involved in sepsis, such as the inflammatory response, immune dysfunction, complement deactivation, mitochondrial damage, and endoplasmic reticulum stress. Additionally, we highlight important targets involved in sepsis-related regulatory mechanisms, including GSDMD, HMGB1, STING, and SQSTM1, among others. We summarize the latest advancements in potential therapeutic drugs that specifically target these signaling pathways and paramount targets, covering both preclinical studies and clinical trials. In addition, this review provides a detailed description of the crosstalk and function between signaling pathways and vital targets, which provides more opportunities for the clinical development of new treatments for sepsis.
Collapse
Affiliation(s)
- Wendan Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Honghong Jiang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Gaosong Wu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Pengli Huang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Haonan Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huazhasng An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghaiChina
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosecurityShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
27
|
Elkalawy H, Sekhar P, Abosena W. Early detection and assessment of intensive care unit-acquired weakness: a comprehensive review. Acute Crit Care 2023; 38:409-424. [PMID: 38052508 DOI: 10.4266/acc.2023.00703] [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: 05/14/2023] [Accepted: 10/17/2023] [Indexed: 12/07/2023] Open
Abstract
Intensive care unit-acquired weakness (ICU-AW) is a serious complication in critically ill patients. Therefore, timely and accurate diagnosis and monitoring of ICU-AW are crucial for effectively preventing its associated morbidity and mortality. This article provides a comprehensive review of ICU-AW, focusing on the different methods used for its diagnosis and monitoring. Additionally, it highlights the role of bedside ultrasound in muscle assessment and early detection of ICU-AW. Furthermore, the article explores potential strategies for preventing ICU-AW. Healthcare providers who manage critically ill patients utilize diagnostic approaches such as physical exams, imaging, and assessment tools to identify ICU-AW. However, each method has its own limitations. The diagnosis of ICU-AW needs improvement due to the lack of a consensus on the appropriate approach for its detection. Nevertheless, bedside ultrasound has proven to be the most reliable and cost-effective tool for muscle assessment in the ICU. Combining the Sequential Organ Failure Assessment (SOFA) score, Acute Physiology and Chronic Health Evaluation (APACHE) II score assessment, and ultrasound can be a convenient approach for the early detection of ICU-AW. This approach can facilitate timely intervention and prevent catastrophic consequences. However, further studies are needed to strengthen the evidence.
Collapse
Affiliation(s)
- Hanan Elkalawy
- Department of Anesthesiology and Perioperative Medicine, Tufts Medical Center, Boston, MA, USA
| | - Pavan Sekhar
- Department of Anesthesiology and Perioperative Medicine, Tufts Medical Center, Boston, MA, USA
| | - Wael Abosena
- Department of Surgery, Faculty of Medicine, Tanta University, Gharbeya, Egypt
| |
Collapse
|
28
|
Liu H, Pan D, Li P, Wang D, Xia B, Zhang R, Lu J, Xing X, Du J, Zhang X, Jin L, Jiang L, Yao L, Li M, Wu J. Loss of ZBED6 Protects Against Sepsis-Induced Muscle Atrophy by Upregulating DOCK3-Mediated RAC1/PI3K/AKT Signaling Pathway in Pigs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302298. [PMID: 37551034 PMCID: PMC10582467 DOI: 10.1002/advs.202302298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/12/2023] [Indexed: 08/09/2023]
Abstract
Sepsis-induced muscle atrophy often increases morbidity and mortality in intensive care unit (ICU) patients, yet neither therapeutic target nor optimal animal model is available for this disease. Here, by modifying the surgical strategy of cecal ligation and puncture (CLP), a novel sepsis pig model is created that for the first time recapitulates the whole course of sepsis in humans. With this model and sepsis patients, increased levels of the transcription factor zinc finger BED-type containing 6 (ZBED6) in skeletal muscle are shown. Protection against sepsis-induced muscle wasting in ZBED6-deficient pigs is further demonstrated. Mechanistically, integrated analysis of RNA-seq and ChIP-seq reveals dedicator of cytokinesis 3 (DOCK3) as the direct target of ZBED6. In septic ZBED6-deficient pigs, DOCK3 expression is increased in skeletal muscle and myocytes, activating the RAC1/PI3K/AKT pathway and protecting against sepsis-induced muscle wasting. Conversely, opposite gene expression patterns and exacerbated muscle wasting are observed in septic ZBED6-overexpressing myotubes. Notably, sepsis patients show increased ZBED6 expression along with reduced DOCK3 and downregulated RAC1/PI3K/AKT pathway. These findings suggest that ZBED6 is a potential therapeutic target for sepsis-induced muscle atrophy, and the established sepsis pig model is a valuable tool for understanding sepsis pathogenesis and developing its therapeutics.
Collapse
Affiliation(s)
- Huan Liu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Dengke Pan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalChengduSichuan610072China
| | - Pu Li
- Department of Critical Care Medicinethe Second Affiliated Hospital of Air Force Medical UniversityNo.569, Xinsi RoadXi'anShaanxi710038China
| | - Dandan Wang
- Laboratory of Animal (Poultry) Genetics Breeding and ReproductionMinistry of AgricultureInstitute of Animal SciencesChinese Academy of Agricultural Sciences (CAAS)Beijing100193China
| | - Bo Xia
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Ruixin Zhang
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Junfeng Lu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Xiangyang Xing
- Chengdu Clonorgan Biotechnology Co. LTDChengduSichuan610041China
| | - Jiaxiang Du
- Chengdu Clonorgan Biotechnology Co. LTDChengduSichuan610041China
| | - Xiao Zhang
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Long Jin
- Institute of Animal Genetics and BreedingCollege of Animal Science and TechnologySichuan Agricultural UniversityChengduSichuan611130China
| | - Lin Jiang
- Laboratory of Animal (Poultry) Genetics Breeding and ReproductionMinistry of AgricultureInstitute of Animal SciencesChinese Academy of Agricultural Sciences (CAAS)Beijing100193China
| | - Linong Yao
- Department of Critical Care Medicinethe Second Affiliated Hospital of Air Force Medical UniversityNo.569, Xinsi RoadXi'anShaanxi710038China
| | - Mingzhou Li
- Institute of Animal Genetics and BreedingCollege of Animal Science and TechnologySichuan Agricultural UniversityChengduSichuan611130China
| | - Jiangwei Wu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| |
Collapse
|
29
|
van der Slikke EC, Beumeler LFE, Holmqvist M, Linder A, Mankowski RT, Bouma HR. Understanding Post-Sepsis Syndrome: How Can Clinicians Help? Infect Drug Resist 2023; 16:6493-6511. [PMID: 37795206 PMCID: PMC10546999 DOI: 10.2147/idr.s390947] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/21/2023] [Indexed: 10/06/2023] Open
Abstract
Sepsis is a global health challenge, with over 49 million cases annually. Recent medical advancements have increased in-hospital survival rates to approximately 80%, but the escalating incidence of sepsis, owing to an ageing population, rise in chronic diseases, and antibiotic resistance, have also increased the number of sepsis survivors. Subsequently, there is a growing prevalence of "post-sepsis syndrome" (PSS). This syndrome includes long-term physical, medical, cognitive, and psychological issues after recovering from sepsis. PSS puts survivors at risk for hospital readmission and is associated with a reduction in health- and life span, both at short and long term, after hospital discharge. Comprehensive understanding of PSS symptoms and causative factors is vital for developing optimal care for sepsis survivors, a task of prime importance for clinicians. This review aims to elucidate our current knowledge of PSS and its relevance in enhancing post-sepsis care provided by clinicians.
Collapse
Affiliation(s)
- Elisabeth C van der Slikke
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, 9713GZ, the Netherlands
| | - Lise F E Beumeler
- Department of Intensive Care, Medical Centre Leeuwarden, Leeuwarden, 8934AD, the Netherlands
- Department of Sustainable Health, Campus Fryslân, University of Groningen, Groningen, 8911 CE, the Netherlands
| | - Madlene Holmqvist
- Department of Infection Medicine, Skåne University Hospital Lund, Lund, 221 84, Sweden
| | - Adam Linder
- Department of Infection Medicine, Skåne University Hospital Lund, Lund, 221 84, Sweden
| | - Robert T Mankowski
- Department of Physiology and Aging, University of Florida, Gainesville, FL, 32610, USA
| | - Hjalmar R Bouma
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, 9713GZ, the Netherlands
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, 9713GZ, the Netherlands
| |
Collapse
|
30
|
Sikora JP, Karawani J, Sobczak J. Neutrophils and the Systemic Inflammatory Response Syndrome (SIRS). Int J Mol Sci 2023; 24:13469. [PMID: 37686271 PMCID: PMC10488036 DOI: 10.3390/ijms241713469] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
We are not entirely able to understand, assess, and modulate the functioning of the immune system in clinical situations that lead to a systemic inflammatory response. In the search for diagnostic and treatment strategies (which are still far from perfect), it became very important to study the pathogenesis and participation of endogenous inflammation mediators. This study attempts to more precisely establish the role of neutrophils in individual phenomena occurring during an inflammatory and anti-inflammatory reaction, taking into account their cidal, immunoregulatory, and reparative abilities. Pro- and anticoagulatory properties of endothelium in systemic inflammatory response syndrome (SIRS) are emphasised, along with the resulting clinical implications (the application of immunotherapy using mesenchymal stem/stromal cells (MSCs) or IL-6 antagonists in sepsis and COVID-19 treatment, among others). Special attention is paid to reactive oxygen species (ROS), produced by neutrophils activated during "respiratory burst" in the course of SIRS; the protective and pathogenic role of these endogenous mediators is highlighted. Moreover, clinically useful biomarkers of SIRS (neutrophil extracellular traps, cell-free DNA, DAMP, TREMs, NGAL, miRNA, selected cytokines, ROS, and recognised markers of endothelial damage from the group of adhesins by means of immunohistochemical techniques) related to the neutrophils are presented, and their role in the diagnosing and forecasting of sepsis, burn disease, and COVID-19 is emphasised. Finally, examples of immunomodulation of sepsis and antioxidative thermal injury therapy are presented.
Collapse
Affiliation(s)
- Janusz P. Sikora
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
| | - Jakub Karawani
- Faculty of Medicine, Lazarski University, ul. Świeradowska 43, 02-662 Warsaw, Poland;
| | - Jarosław Sobczak
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
- Department of Management and Logistics in Healthcare, Medical University of Łódź, ul. Lindleya 6, 90-131 Łódź, Poland
| |
Collapse
|
31
|
Su J, Chen S, Xiao J, Feng Z, Hu S, Su Q, Chen Q, Chen D. Aloe-Emodin Ameliorates Cecal Ligation and Puncture-Induced Sepsis. Int J Mol Sci 2023; 24:11972. [PMID: 37569344 PMCID: PMC10418438 DOI: 10.3390/ijms241511972] [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: 06/25/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Sepsis remains a major challenge owing to its severe adverse effects and high mortality, against which specific pharmacological interventions with high efficacy are limited. Mitigation of hyperactive inflammatory responses is a key factor in enhancing the likelihood of survival in patients with sepsis. The Aloe genus has several health benefits, including anti-inflammatory properties. The toxicological implications of aloe-emodin (AE), extracted from various Aloe species, remain uncertain in clinical contexts. However, AE has been shown to inhibit inflammatory responses in lipopolysaccharide-induced mice, indicating its potential as a therapeutic approach for sepsis treatment. Nonetheless, there is a paucity of data regarding the therapeutic benefits of AE in the widely recognized cecal ligation and puncture (CLP)-induced sepsis model, which is commonly used as the gold standard model for sepsis research. This study demonstrates the potential benefits of AE in the treatment of CLP-induced sepsis and investigates its underlying mechanism, along with the efficacy of postoperative AE treatment in mice with CLP-induced sepsis. The results of this study suggest that AE can mitigate sepsis in mice by diminishing systemic inflammation and regulating the gut microbiota. The study provides novel insights into the molecular mechanisms underlying the anti-inflammatory effects of AE.
Collapse
Affiliation(s)
- Jingqian Su
- Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Z.F.); (S.H.); (Q.S.)
- Provincial University Key Laboratory of Microbial Pathogenesis and Interventions, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Siyuan Chen
- The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Products of the State Oceanic Administration, Fujian Key Laboratory of Special Marine Bioresource Sustainable Utilization, Southern Institute of Oceanography, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (S.C.); (J.X.)
| | - Jianbin Xiao
- The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Products of the State Oceanic Administration, Fujian Key Laboratory of Special Marine Bioresource Sustainable Utilization, Southern Institute of Oceanography, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (S.C.); (J.X.)
| | - Zhihua Feng
- Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Z.F.); (S.H.); (Q.S.)
| | - Shan Hu
- Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Z.F.); (S.H.); (Q.S.)
- Provincial University Key Laboratory of Microbial Pathogenesis and Interventions, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Qiaofen Su
- Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Z.F.); (S.H.); (Q.S.)
- Provincial University Key Laboratory of Microbial Pathogenesis and Interventions, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Qi Chen
- Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (J.S.); (Z.F.); (S.H.); (Q.S.)
| | - Duo Chen
- The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Products of the State Oceanic Administration, Fujian Key Laboratory of Special Marine Bioresource Sustainable Utilization, Southern Institute of Oceanography, College of Life Science, Fujian Normal University, Fuzhou 350117, China; (S.C.); (J.X.)
| |
Collapse
|
32
|
Qian W, Zhou J, Shou S. Exploration of m 6A methylation regulators as epigenetic targets for immunotherapy in advanced sepsis. BMC Bioinformatics 2023; 24:257. [PMID: 37330481 DOI: 10.1186/s12859-023-05379-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/06/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND This study aims to deeply explore the relationship between m6A methylation modification and peripheral immune cells in patients with advanced sepsis and mine potential epigenetic therapeutic targets by analyzing the differential expression patterns of m6A-related genes in healthy subjects and advanced sepsis patients. METHODS A single cell expression dataset of peripheral immune cells containing blood samples from 4 patients with advanced sepsis and 5 healthy subjects was obtained from the gene expression comprehensive database (GSE175453). Differential expression analysis and cluster analysis were performed on 21 m6A-related genes. The characteristic gene was identified based on random forest algorithm, and the correlation between the characteristic gene METTL16 and 23 immune cells in patients with advanced sepsis was evaluated using single-sample gene set enrichment analysis. RESULTS IGFBP1, IGFBP2, IGF2BP1, and WTAP were highly expressed in patients with advanced sepsis and m6A cluster B. IGFBP1, IGFBP2, and IGF2BP1 were positively correlated with Th17 helper T cells. The characteristic gene METTL16 exhibited a significant positive correlation with the proportion of various immune cells. CONCLUSION IGFBP1, IGFBP2, IGF2BP1, WTAP, and METTL16 may accelerate the development of advanced sepsis by regulating m6A methylation modification and promoting immune cell infiltration. The discovery of these characteristic genes related to advanced sepsis provides potential therapeutic targets for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Weiwei Qian
- Tianjin Medical University, Tianjin, 300203, China
- Department of Emergency, Shangjin Nanfu Hospital, West China Hospital, Sichuan University, Chengdu, 610044, Sichuan, China
| | - Jian Zhou
- Department of Immunology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Songtao Shou
- Department of Emergency, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
33
|
Mendelson AA, Erickson D, Villar R. The role of the microcirculation and integrative cardiovascular physiology in the pathogenesis of ICU-acquired weakness. Front Physiol 2023; 14:1170429. [PMID: 37234410 PMCID: PMC10206327 DOI: 10.3389/fphys.2023.1170429] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Skeletal muscle dysfunction after critical illness, defined as ICU-acquired weakness (ICU-AW), is a complex and multifactorial syndrome that contributes significantly to long-term morbidity and reduced quality of life for ICU survivors and caregivers. Historically, research in this field has focused on pathological changes within the muscle itself, without much consideration for their in vivo physiological environment. Skeletal muscle has the widest range of oxygen metabolism of any organ, and regulation of oxygen supply with tissue demand is a fundamental requirement for locomotion and muscle function. During exercise, this process is exquisitely controlled and coordinated by the cardiovascular, respiratory, and autonomic systems, and also within the skeletal muscle microcirculation and mitochondria as the terminal site of oxygen exchange and utilization. This review highlights the potential contribution of the microcirculation and integrative cardiovascular physiology to the pathogenesis of ICU-AW. An overview of skeletal muscle microvascular structure and function is provided, as well as our understanding of microvascular dysfunction during the acute phase of critical illness; whether microvascular dysfunction persists after ICU discharge is currently not known. Molecular mechanisms that regulate crosstalk between endothelial cells and myocytes are discussed, including the role of the microcirculation in skeletal muscle atrophy, oxidative stress, and satellite cell biology. The concept of integrated control of oxygen delivery and utilization during exercise is introduced, with evidence of physiological dysfunction throughout the oxygen delivery pathway - from mouth to mitochondria - causing reduced exercise capacity in patients with chronic disease (e.g., heart failure, COPD). We suggest that objective and perceived weakness after critical illness represents a physiological failure of oxygen supply-demand matching - both globally throughout the body and locally within skeletal muscle. Lastly, we highlight the value of standardized cardiopulmonary exercise testing protocols for evaluating fitness in ICU survivors, and the application of near-infrared spectroscopy for directly measuring skeletal muscle oxygenation, representing potential advancements in ICU-AW research and rehabilitation.
Collapse
Affiliation(s)
- Asher A. Mendelson
- Section of Critical Care Medicine, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Dustin Erickson
- Section of Critical Care Medicine, Department of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Rodrigo Villar
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
34
|
Chen J, Chen XY, Cong XX, Wang S, Xu SB, Sun YT, Zhou YT, Zheng LL, Huang M. CELLULAR SENESCENCE IMPLICATED IN SEPSIS-INDUCED MUSCLE WEAKNESS AND AMELIORATED WITH METFORMIN. Shock 2023; 59:646-656. [PMID: 36719431 DOI: 10.1097/shk.0000000000002086] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
ABSTRACT Background: Sepsis is a life-threatening medical emergency, frequently complicated with intensive care unit-acquired weakness syndrome (ICU-AW). ICU-AW patients display flaccid weakness of the limbs, especially in the proximal limb muscles. However, little is known regarding its pathogenesis. Here, we aimed to identify the potential signaling pathway involved in ICU-AW regulation and identify a potential therapeutic drug for intervention. Methods: Both in vivo and in vitro septic mice were used. For the in vivo septic mice, either cecum ligation and puncture or intraperitoneal injection of LPS was conducted in mice. The body weight and muscle mass were then measured and recorded. Muscle strength was evaluated by limb grip strength test. The expression of proteins extracted from cells and muscles was checked through Western blot analysis. Quantitative reverse transcription-polymerase chain reaction was carried out to test the transcriptional level of genes. Senescence-associated β-galactosidase (SA-β-gal) staining and Sirius red for collagen staining were conducted. Metformin, as an antiaging agent, was then tested for any attenuation of sepsis-related symptoms. For in vitro sepsis modeling, myoblasts were treated with LPS, analyzed for senescence-related protein expression, and subsequently retested upon metformin treatment. Results: We found that both the weight and strength of muscle were dramatically reduced in cecum ligation and puncture- or LPS-induced septic mice. RNA-seq analysis revealed that various cellular senescent genes were involved in sepsis. In line with this, expression of senescence-related genes, p53 and p21 were both upregulated. Both SA-β-gal and Sirius red for collagen staining were enhanced in tibialis anterior muscles. Notably, inhibition of p53 expression by siRNA prominently reduced the number of SA-β-gal-positive myoblasts upon LPS treatment. This indicated sepsis-induced cellular senescence to be dependent on p53. Consistent with the function of metformin in antiaging, metformin attenuated cellular senescence in both murine myoblasts and skeletal muscles during sepsis. Muscle strength of septic mice was improved upon metformin treatment. Metformin intervention is therefore proposed as a potential therapeutic strategy for ICU-AW. Conclusion: Taken together, we revealed a previously unappreciated linkage between cellular senescence and sepsis-induced muscle weakness and propose metformin as a potential therapeutic drug for the treatment of ICU-AW.
Collapse
Affiliation(s)
- Juan Chen
- Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Yi Chen
- Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Shen Wang
- Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shui Bo Xu
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Ting Sun
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | - Man Huang
- Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
35
|
Klawitter F, Ehler J, Bajorat R, Patejdl R. Mitochondrial Dysfunction in Intensive Care Unit-Acquired Weakness and Critical Illness Myopathy: A Narrative Review. Int J Mol Sci 2023; 24:5516. [PMID: 36982590 PMCID: PMC10052131 DOI: 10.3390/ijms24065516] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Mitochondria are key structures providing most of the energy needed to maintain homeostasis. They are the main source of adenosine triphosphate (ATP), participate in glucose, lipid and amino acid metabolism, store calcium and are integral components in various intracellular signaling cascades. However, due to their crucial role in cellular integrity, mitochondrial damage and dysregulation in the context of critical illness can severely impair organ function, leading to energetic crisis and organ failure. Skeletal muscle tissue is rich in mitochondria and, therefore, particularly vulnerable to mitochondrial dysfunction. Intensive care unit-acquired weakness (ICUAW) and critical illness myopathy (CIM) are phenomena of generalized weakness and atrophying skeletal muscle wasting, including preferential myosin breakdown in critical illness, which has also been linked to mitochondrial failure. Hence, imbalanced mitochondrial dynamics, dysregulation of the respiratory chain complexes, alterations in gene expression, disturbed signal transduction as well as impaired nutrient utilization have been proposed as underlying mechanisms. This narrative review aims to highlight the current known molecular mechanisms immanent in mitochondrial dysfunction of patients suffering from ICUAW and CIM, as well as to discuss possible implications for muscle phenotype, function and therapeutic approaches.
Collapse
Affiliation(s)
- Felix Klawitter
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Johannes Ehler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany
| | - Rika Bajorat
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Robert Patejdl
- Oscar Langendorff Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
36
|
Inhibition of DDX3X alleviates persistent inflammation, immune suppression and catabolism syndrome in a septic mice model. Int Immunopharmacol 2023; 117:109779. [PMID: 36806038 DOI: 10.1016/j.intimp.2023.109779] [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: 11/11/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/22/2023]
Abstract
OBJECTIVE DDX3X is involved in various pathological processes such as infection, immunity and cell death. This study aimed to investigate the effect of RK-33, a specific inhibitor of DDX3X, on the progression of sepsis to persistent inflammation, immune suppression and catabolism syndrome(PICS). METHODS The septic mice model was established using caecal ligation and perforation (CLP). The mice were randomly divided into four groups: sham group, sham + RK-33 group (20 mg/kg, intraperitoneal injection, once a day), CLP group and CLP + RK-33 group (20 mg/kg, intraperitoneal injection, once a day). The number of inflammatory cells in the peripheral blood, spleen and bone marrow was calculated, and inflammatory cytokines were detected using an enzyme-linked immunosorbent assay. The septic mice's body weight and skeletal muscle mass were measured, and skeletal muscle tissues were examined using eosin staining. Western blotting was performed to detect the expression levels of MuRF1, atrogin1 and NLRP3 in the skeletal muscle of septic mice. Additionally, reactive oxidative species, superoxide dismutase and malondialdehyde were measured using commercial kits. RESULTS RK-33 reduced inflammatory cell counts and cytokine levels in CLP mice, ameliorated the decline in CD4 and CD8 T cells and prevented the loss of body weight and skeletal muscle mass in septic mice. Additionally, RX-33 reduced oxidative stress in the skeletal muscle of septic mice. CONCLUSION In the established sepsis mouse model, RK-33 alleviated inflammation and oxidative stress, ameliorated CLP-induced immunosuppression and skeletal muscle atrophy and improved survival. These findings suggest that RK-33 could be a novel potential therapeutic agent for preventing the progression of sepsis to PICS.
Collapse
|
37
|
Yang CH, Chen YA, Bin PJ, Ou SM, Tarng DC. Associations of the Serum Total Carbon Dioxide Level with Long-Term Clinical Outcomes in Sepsis Survivors. Infect Dis Ther 2023; 12:687-701. [PMID: 36749474 PMCID: PMC9925627 DOI: 10.1007/s40121-023-00765-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/26/2023] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Sepsis is characterized by a dysregulated host response to infection that leads to multiple organ dysfunction and often complicated with metabolic acidosis. However, the associations between serum total carbon dioxide level (TCO2) and long-term clinical outcomes in sepsis survivors remains unknown. METHODS A total of 7212 sepsis survivors aged ≥ 20 years who were discharged from January 1, 2008 to December 31, 2018 were included in our analyses. The sepsis survivors were further divided into high TCO2 (≥ 18 mmol/L) and low TCO2 (< 18 mmol/L) groups, comprising 5023 and 2189 patients, respectively. The following outcomes of interest were included: all-cause mortality, myocardial infarction, ischemic stroke, hospitalization for heart failure, ventricular arrhythmia, and end-stage renal disease (ESRD). RESULTS After propensity score matching, the low TCO2 group was at higher risks of all-cause mortality (hazard ratio [HR] 1.28, 95% confidence interval [95% CI] 1.18-1.39), myocardial infarction (HR 1.83, 95% CI 1.39-2.43), and ESRD (HR 1.38, 95% CI 1.16-1.64) than the high TCO2 group. The results remained similar after considering death as a competing risk. CONCLUSION Patients discharged from hospitalization for sepsis have higher risks of worse long-term clinical outcomes. Physicians may need to pay more attention to sepsis survivors whose TCO2 was low.
Collapse
Affiliation(s)
- Ching Han Yang
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Yee-An Chen
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan
| | - Pin-Jie Bin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shuo-Ming Ou
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan.
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei, 11217, Taiwan.
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department and Institute of Physiology, National Yang-Ming University, Taipei, Taiwan.
- Department and Institute of Physiology, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
38
|
Impaired muscle stem cell function and abnormal myogenesis in acquired myopathies. Biosci Rep 2023; 43:232343. [PMID: 36538023 PMCID: PMC9829652 DOI: 10.1042/bsr20220284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle possesses a high plasticity and a remarkable regenerative capacity that relies mainly on muscle stem cells (MuSCs). Molecular and cellular components of the MuSC niche, such as immune cells, play key roles to coordinate MuSC function and to orchestrate muscle regeneration. An abnormal infiltration of immune cells and/or imbalance of pro- and anti-inflammatory cytokines could lead to MuSC dysfunctions that could have long lasting effects on muscle function. Different genetic variants were shown to cause muscular dystrophies that intrinsically compromise MuSC function and/or disturb their microenvironment leading to impaired muscle regeneration that contributes to disease progression. Alternatively, many acquired myopathies caused by comorbidities (e.g., cardiopulmonary or kidney diseases), chronic inflammation/infection, or side effects of different drugs can also perturb MuSC function and their microenvironment. The goal of this review is to comprehensively summarize the current knowledge on acquired myopathies and their impact on MuSC function. We further describe potential therapeutic strategies to restore MuSC regenerative capacity.
Collapse
|
39
|
Martí‐Chillón G, Muntión S, Preciado S, Osugui L, Navarro‐Bailón A, González‐Robledo J, Sagredo V, Blanco JF, Sánchez‐Guijo F. Therapeutic potential of mesenchymal stromal/stem cells in critical-care patients with systemic inflammatory response syndrome. Clin Transl Med 2023; 13:e1163. [PMID: 36588089 PMCID: PMC9806020 DOI: 10.1002/ctm2.1163] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Despite notable advances in the support and treatment of patients admitted to the intensive care unit (ICU), the management of those who develop a systemic inflammatory response syndrome (SIRS) still constitutes an unmet medical need. MAIN BODY Both the initial injury (trauma, pancreatitis, infections) and the derived uncontrolled response promote a hyperinflammatory status that leads to systemic hypotension, tissue hypoperfusion and multiple organ failure. Mesenchymal stromal/stem cells (MSCs) are emerging as a potential therapy for severe ICU patients due to their potent immunomodulatory, anti-inflammatory, regenerative and systemic homeostasis-regulating properties. MSCs have demonstrated clinical benefits in several inflammatory-based diseases, but their role in SIRS needs to be further explored. CONCLUSION In the current review, after briefly overviewing SIRS physiopathology, we explore the potential mechanisms why MSC therapy could aid in the recovery of this condition and the pre-clinical and early clinical evidence generated to date.
Collapse
Affiliation(s)
| | - Sandra Muntión
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- RICORS TERAVISCIIIMadridSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| | - Silvia Preciado
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- RICORS TERAVISCIIIMadridSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| | - Lika Osugui
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| | - Almudena Navarro‐Bailón
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- RICORS TERAVISCIIIMadridSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| | - Javier González‐Robledo
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- Department of MedicineUniversity of SalamancaSalamancaSpain
| | | | - Juan F. Blanco
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
- Department of SurgeryUniversity of SalamancaSalamancaSpain
| | - Fermín Sánchez‐Guijo
- IBSAL‐University Hospital of SalamancaSalamancaSpain
- Department of MedicineUniversity of SalamancaSalamancaSpain
- RICORS TERAVISCIIIMadridSpain
- Regenerative Medicine and Cellular Therapy Network Center of Castilla y LeónSalamancaSpain
| |
Collapse
|
40
|
Eshghi F, Tahmasebi S, Alimohammadi M, Soudi S, Khaligh SG, Khosrojerdi A, Heidari N, Hashemi SM. Study of immunomodulatory effects of mesenchymal stem cell-derived exosomes in a mouse model of LPS induced systemic inflammation. Life Sci 2022; 310:120938. [PMID: 36150466 DOI: 10.1016/j.lfs.2022.120938] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/19/2022] [Accepted: 09/04/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Sepsis is a debilitating systemic inflammation that resulted from infection or injury. Despite many advances in treatment, the resulting mortality rate has remained high due to increasing antibiotic resistance and aging communities. The present study investigated the effects of stem cell-derived exosomes in a mouse model of LPS-induced systemic inflammation. MATERIALS AND METHODS To induce sepsis, the LPS model was used. Mice were divided into three groups: normal, patient group (LPS + PBS), and treatment group (LPS + exosome). The treatment group received an intravenous exosome 1 h after induction of the model. Patient and treatment groups were sacrificed at 4, 6, 24, and 48 h after induction of the model, and their tissues were isolated. Blood samples were taken from animal hearts to perform biochemical and immunological tests. The study results were analyzed using Graph Pad Prism software version 9. RESULTS Mesenchymal stem cell-derived exosomes decreased serum levels of ALT and AST liver enzymes, decreased neutrophil to lymphocyte ratio (NLR), and improved kidney, liver, and lung tissue damage at 4, 6, and 24 h after model induction. At 24 h, the exosomes were able to reduce serum urea levels. This study revealed decreased levels of inflammatory cytokines such as IL-6, IL-1β, and TNF-α after exosome injection. CONCLUSION Our findings suggest that treating mice with stem cell-derived exosomes can ameliorate the destructive effects of inflammation caused by sepsis by reducing inflammatory factors and tissue damage.
Collapse
Affiliation(s)
- Fateme Eshghi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Arezou Khosrojerdi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Neda Heidari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Damanti S, Cristel G, Ramirez GA, Bozzolo EP, Da Prat V, Gobbi A, Centurioni C, Di Gaeta E, Del Prete A, Calabrò MG, Calvi MR, Borghi G, Zangrillo A, De Cobelli F, Landoni G, Tresoldi M. Influence of reduced muscle mass and quality on ventilator weaning and complications during intensive care unit stay in COVID-19 patients. Clin Nutr 2022; 41:2965-2972. [PMID: 34465493 PMCID: PMC8364854 DOI: 10.1016/j.clnu.2021.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND & AIMS Sarcopenia, a loss of muscle mass, quality and function, which is particularly evident in respiratory muscles, has been associated with many clinical adverse outcomes. In this study, we aimed at evaluating the role of reduced muscle mass and quality in predicting ventilation weaning, complications, length of intensive care unit (ICU) and of hospital stay and mortality in patients admitted to ICU for SARS-CoV-2-related pneumonia. METHODS This was an observational study based on a review of medical records of all adult patients admitted to the ICU of a tertiary hospital in Milan and intubated for SARS-CoV-2-related pneumonia during the first wave of the COVID-19 pandemic. Muscle mass and quality measurement were retrieved from routine thoracic CT scans, when sections passing through the first, second or third lumbar vertebra were available. RESULTS A total of 81 patients were enrolled. Muscle mass was associated with successful extubation (OR 1.02, 95% C.I. 1.00-1.03, p = 0.017), shorter ICU stay (OR 0.97, 95% C.I. 0.95-0.99, p = 0.03) and decreased hospital mortality (HR 0.98, 95% C.I. 0.96-0.99, p = 0.02). Muscle density was associated with successful extubation (OR 1.07, 95% C.I. 1.01-1.14; p = 0.02) and had an inverse association with the number of complications in ICU (Β -0.07, 95% C.I. -0.13 - -0.002, p = 0.03), length of hospitalization (Β -1.36, 95% C.I. -2.21 - -0.51, p = 0.002) and in-hospital mortality (HR 0.88, 95% C.I. 0.78-0.99, p = 0.046). CONCLUSIONS Leveraging routine CT imaging to measure muscle mass and quality might constitute a simple, inexpensive and powerful tool to predict survival and disease course in patients with COVID-19. Preserving muscle mass during hospitalisation might have an adjuvant role in facilitating remission from COVID-19.
Collapse
Affiliation(s)
- Sarah Damanti
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Scientific Institute, Italy,Corresponding author. Unit of General Medicine and Advanced Care, IRCCS San Raffaele Hospital, Via Olgettina 60, Milan, Italy
| | - Giulia Cristel
- Department of Radiology, Centre for Experimental Imaging, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Alvise Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Enrica Paola Bozzolo
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Scientific Institute, Italy
| | - Valentina Da Prat
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Scientific Institute, Italy
| | - Agnese Gobbi
- Vita-Salute San Raffaele University, Milano, Italy
| | | | - Ettore Di Gaeta
- Department of Radiology, Centre for Experimental Imaging, IRCCS San Raffaele Scientific Institute, Milan, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Andrea Del Prete
- Department of Radiology, Centre for Experimental Imaging, IRCCS San Raffaele Scientific Institute, Milan, Italy,Vita-Salute San Raffaele University, Milano, Italy
| | - Maria Grazia Calabrò
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Rosa Calvi
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Borghi
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Zangrillo
- Vita-Salute San Raffaele University, Milano, Italy,Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco De Cobelli
- Department of Radiology, Centre for Experimental Imaging, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Landoni
- Vita-Salute San Raffaele University, Milano, Italy,Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, IRCCS San Raffaele Scientific Institute, Italy
| |
Collapse
|
42
|
Gong J, Yang J, He Y, Chen X, Yang G, Sun R. Construction of m7G subtype classification on heterogeneity of sepsis. Front Genet 2022; 13:1021770. [PMID: 36506322 PMCID: PMC9729242 DOI: 10.3389/fgene.2022.1021770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022] Open
Abstract
Sepsis is a highly heterogeneous disease and a major factor in increasing mortality from infection. N7-Methylguanosine (m7G) is a widely RNA modification in eukaryotes, which involved in regulation of different biological processes. Researchers have found that m7G methylation contributes to a variety of human diseases, but its research in sepsis is still limited. Here, we aim to establish the molecular classification of m7G gene-related sepsis, reveal its heterogeneity and explore the underlying mechanism. We first identified eight m7G related prognostic genes, and identified two different molecular subtypes of sepsis through Consensus Clustering. Among them, the prognosis of C2 subtype is worse than that of C1 subtype. The signal pathways enriched by the two subtypes were analyzed by ssGSEA, and the results showed that the amino acid metabolism activity of C2 subtype was more active than that of C1 subtype. In addition, the difference of immune microenvironment among different subtypes was explored through CIBERSORT algorithm, and the results showed that the contents of macrophages M0 and NK cells activated were significantly increased in C2 subtype, while the content of NK cells resting decreased significantly in C2 subtype. We further explored the relationship between immune regulatory genes and inflammation related genes between C2 subtype and C1 subtype, and found that C2 subtype showed higher expression of immune regulatory genes and inflammation related genes. Finally, we screened the key genes in sepsis by WGCNA analysis, namely NUDT4 and PARN, and verified their expression patterns in sepsis in the datasets GSE131761 and GSE65682. The RT-PCR test further confirmed the increased expression of NUDTA4 in sepsis patients. In conclusion, sepsis clustering based on eight m7G-related genes can well distinguish the heterogeneity of sepsis patients and help guide the personalized treatment of sepsis patients.
Collapse
Affiliation(s)
- Jinru Gong
- Department of Pulmonary and Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Jiasheng Yang
- Department of Pulmonary and Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yaowei He
- Department of Pulmonary and Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaoxuan Chen
- Department of Pulmonary and Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Guangyu Yang
- Department of Pulmonary and Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ruilin Sun
- Department of Pulmonary and Critical Care Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China,*Correspondence: Ruilin Sun,
| |
Collapse
|
43
|
dos Santos AAC, Rodrigues LE, Alecrim-Zeza AL, de Araújo Ferreira L, Trettel CDS, Gimenes GM, da Silva AF, Sousa-Filho CPB, Serdan TDA, Levada-Pires AC, Hatanaka E, Borges FT, de Barros MP, Cury-Boaventura MF, Bertolini GL, Cassolla P, Marzuca-Nassr GN, Vitzel KF, Pithon-Curi TC, Masi LN, Curi R, Gorjao R, Hirabara SM. Molecular and cellular mechanisms involved in tissue-specific metabolic modulation by SARS-CoV-2. Front Microbiol 2022; 13:1037467. [PMID: 36439786 PMCID: PMC9684198 DOI: 10.3389/fmicb.2022.1037467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/26/2022] [Indexed: 09/09/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is triggered by the SARS-CoV-2, which is able to infect and cause dysfunction not only in lungs, but also in multiple organs, including central nervous system, skeletal muscle, kidneys, heart, liver, and intestine. Several metabolic disturbances are associated with cell damage or tissue injury, but the mechanisms involved are not yet fully elucidated. Some potential mechanisms involved in the COVID-19-induced tissue dysfunction are proposed, such as: (a) High expression and levels of proinflammatory cytokines, including TNF-α IL-6, IL-1β, INF-α and INF-β, increasing the systemic and tissue inflammatory state; (b) Induction of oxidative stress due to redox imbalance, resulting in cell injury or death induced by elevated production of reactive oxygen species; and (c) Deregulation of the renin-angiotensin-aldosterone system, exacerbating the inflammatory and oxidative stress responses. In this review, we discuss the main metabolic disturbances observed in different target tissues of SARS-CoV-2 and the potential mechanisms involved in these changes associated with the tissue dysfunction.
Collapse
Affiliation(s)
| | - Luiz Eduardo Rodrigues
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Amanda Lins Alecrim-Zeza
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Liliane de Araújo Ferreira
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Caio dos Santos Trettel
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Gabriela Mandú Gimenes
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Adelson Fernandes da Silva
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | | | - Tamires Duarte Afonso Serdan
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
- Department of Molecular Pathobiology, University of New York, New York, NY, United States
| | - Adriana Cristina Levada-Pires
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Elaine Hatanaka
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Fernanda Teixeira Borges
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
- Divisão de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Marcelo Paes de Barros
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Maria Fernanda Cury-Boaventura
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Gisele Lopes Bertolini
- Department of Physiological Sciences, Biological Science Center, State University of Londrina, Londrina, PR, Brazil
| | - Priscila Cassolla
- Department of Physiological Sciences, Biological Science Center, State University of Londrina, Londrina, PR, Brazil
| | | | - Kaio Fernando Vitzel
- School of Health Sciences, College of Health, Massey University, Auckland, New Zealand
| | - Tania Cristina Pithon-Curi
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Laureane Nunes Masi
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Rui Curi
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
- Instituto Butantan, São Paulo, Brazil
| | - Renata Gorjao
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Sandro Massao Hirabara
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| |
Collapse
|
44
|
Kim J, Stolarski A, Zhang Q, Wee K, Remick D. HYDROCORTISONE, ASCORBIC ACID, AND THIAMINE THERAPY DECREASE RENAL OXIDATIVE STRESS AND ACUTE KIDNEY INJURY IN MURINE SEPSIS. Shock 2022; 58:426-433. [PMID: 36445231 PMCID: PMC9713586 DOI: 10.1097/shk.0000000000001995] [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] [Indexed: 12/03/2022]
Abstract
ABSTRACT Background: Acute kidney injury (AKI) occurs frequently in septic patients and correlates with increased mortality. Because clinical studies investigating hydrocortisone, ascorbic acid, and thiamine (HAT) have demonstrated discordant results, studies were performed using mortality stratification for therapy to identify candidates for therapy and determine mechanisms of organ injury. Methods: Sepsis was induced using the cecal ligation and puncture (CLP) model of sepsis with fluid and antibiotic support. Heart rate (HR) measurements obtained 6 hours after CLP stratified mice into live predicted (P-Live) or die predicted (P-Die). Stratified mice were then randomized for treatment with HAT or vehicle given 7 hours after CLP. Physiologic measurements were taken again at 24 hours, and mice were killed to collect blood and organs. Results: The following five groups were created: (1) P-Live vehicle, (2) P-Live HAT, (3) P-Die vehicle, (4) P-Die HAT, and (5) naive mice. Comparisons were made to test the hypotheses that (1) P-Die vehicle mice will have significant deterioration compared with P-Live mice targeting the kidney and (2) HAT will correct these deleterious changes in P-Die mice. Compared with P-Live, P-Die mice had a significant decline in all measured physiologic parameters (HR, cardiac output, breath rate, and temperature), which were corrected with HAT therapy (P < 0.05 for all parameters). The P-Die mice had declines in the ascorbic acid within the blood, peritoneal lavage, and kidney homogenate compared with P-Live mice indicating consumption, and the decline was corrected with HAT. Elevated IL-6, KC, Macrophage Inflammatory Protein-2, and IL-1RA were found in P-Die mice and decreased with HAT. Markers of endothelial cell injury (glypican 1 and glypican 4) were elevated in the P-Die mice, and these values were decreased with HAT therapy. Low oxygen levels with subsequent oxidative stress (OS) in the kidney were visualized in histologic sections using hypoxyprobe and also with carbonyl proteins and 8-iso-prostaglandin F2α in kidney homogenates. The P-Die mice had significant elevations of renal OSs, which was ameliorated with HAT. Kidney injury was evident in the P-Die mice compared with P-Live mice with elevations in blood urea nitrogen and cystatin C, which were significantly reduced with HAT. There was no evidence of global hypoxia or organ injury because hepatic parameters remained normal. Conclusions: Our data show that in CLP-induced sepsis, P-Die mice have increased inflammation, OS, and kidney injury. Hydrocortisone, ascorbic acid, and thiamine therapy decreased renal OS and injury in the P-Die group when given after the onset of sepsis-induced physiologic changes.
Collapse
Affiliation(s)
- John Kim
- Departments of Pathology and Laboratory Medicine
| | - Allan Stolarski
- Surgery, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts
| | | | | | | |
Collapse
|
45
|
Cao C, Zhang L, Liu F, Shen J. Therapeutic Benefits of Mesenchymal Stem Cells in Acute Respiratory Distress Syndrome: Potential Mechanisms and Challenges. J Inflamm Res 2022; 15:5235-5246. [PMID: 36120184 PMCID: PMC9473549 DOI: 10.2147/jir.s372046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) presents as a form of acute respiratory failure resulting from non-cardiogenic pulmonary edema due to excessive alveolocapillary permeability, which may be pulmonary or systemic in origin. In the last 3 years, the coronavirus disease 2019 pandemic has resulted in an increase in ARDS cases and highlighted the challenges associated with this syndrome, as well as the unacceptably high mortality rates and lack of effective treatments. Currently, clinical treatment remains primarily supportive, including mechanical ventilation and drug-based therapy. Mesenchymal stem cell (MSC) therapies are emerging as a promising intervention in patients with ARDS and have promising therapeutic effects and safety. The therapeutic mechanisms include modifying the immune response and assisting with tissue repair. This review provides an overview of the general properties of MSCs and outlines their role in mitigating lung injury and promoting tissue repair in ARDS. Finally, we summarize the current challenges in the study of translational MSC research and identify avenues by which the discipline may progress in the coming years.
Collapse
Affiliation(s)
- Chao Cao
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, People's Republic of China.,Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai, People's Republic of China.,Shanghai Medical College Fudan University, Shanghai, People's Republic of China
| | - Lin Zhang
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, People's Republic of China.,Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai, People's Republic of China
| | - Fuli Liu
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, People's Republic of China.,Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai, People's Republic of China
| | - Jie Shen
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, People's Republic of China.,Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai, People's Republic of China.,Shanghai Medical College Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
46
|
Zhang T, Zhao J, Fu J, Chen G, Ma T. Improvement of the sepsis survival rate by adenosine 2a receptor antagonists depends on immune regulatory functions of regulatory T-cells. Front Immunol 2022; 13:996446. [PMID: 36148230 PMCID: PMC9485829 DOI: 10.3389/fimmu.2022.996446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Adenosine shows a significant immunosuppressive effect in sepsis via binding to the adenosine 2a receptor (A2aR). Both genetic deletion and pharmacological inhibition of the A2aR may improve survival in sepsis. However, available research on this protective mechanism is quite limited. We used an A2aR antagonist (ZM241385) to treat a cecal ligation and puncture model of normal mice or regulatory T-cell (Treg)-depletion mice and found that the protective effect of ZM241385 is dependent on Tregs. Mechanically, A2aR inactivation was associated with decreased frequencies and reduced function of Foxp3+ Tregs, as evidenced by Foxp3 and CTLA-4 expression and classical effector T-cell proliferative assays, suggesting Treg modulation is a potential protective mechanism against sepsis. Simultaneously, the function and quantity of abdominal neutrophils were improved with ZM241385 treatment. To see if a link exists between them, Tregs and neutrophils were co-cultured, and it was found that ZM241385 blocked the inhibitory effect of Tregs on neutrophils. According to our research, Tregs play a key role in how A2aR antagonists improve sepsis prognosis and bacterial clearance.
Collapse
Affiliation(s)
- Teng Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Zhao
- Department of Intensive Care Unit, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingnan Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Guibing Chen
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Ma
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Tao Ma,
| |
Collapse
|
47
|
Boelens YFN, Melchers M, van Zanten ARH. Poor physical recovery after critical illness: incidence, features, risk factors, pathophysiology, and evidence-based therapies. Curr Opin Crit Care 2022; 28:409-416. [PMID: 35796071 PMCID: PMC9594146 DOI: 10.1097/mcc.0000000000000955] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
PURPOSE OF REVIEW To summarize the incidence, features, pathogenesis, risk factors, and evidence-based therapies of prolonged intensive care unit (ICU) acquired weakness (ICU-AW). We aim to provide an updated overview on aspects of poor physical recovery following critical illness. RECENT FINDINGS New physical problems after ICU survival, such as muscle weakness, weakened condition, and reduced exercise capacity, are the most frequently encountered limitations of patients with postintensive care syndrome. Disabilities may persist for months to years and frequently do not fully recover. Hormonal and mitochondrial disturbances, impaired muscle regeneration due to injured satellite cells and epigenetic differences may be involved in sustained ICU-AW. Although demographics and ICU treatment factors appear essential determinants for physical recovery, pre-ICU health status is also crucial. Currently, no effective treatments are available. Early mobilization in the ICU may improve physical outcomes at ICU-discharge, but there is no evidence for benefit on long-term physical recovery. SUMMARY Impaired physical recovery is observed frequently among ICU survivors. The pre-ICU health status, demographic, and ICU treatment factors appear to be important determinants for physical convalescence during the post-ICU phase. The pathophysiological mechanisms involved are poorly understood, thereby resulting in exiguous evidence-based treatment strategies to date.
Collapse
Affiliation(s)
- Yente Florine Niké Boelens
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, The Netherlands
- Wageningen University & Research, Division of Human Nutrition and Health, Wageningen, The Netherlands
| | - Max Melchers
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, The Netherlands
- Wageningen University & Research, Division of Human Nutrition and Health, Wageningen, The Netherlands
| | - Arthur Raymond Hubert van Zanten
- Department of Intensive Care Medicine, Gelderse Vallei Hospital, Ede, The Netherlands
- Wageningen University & Research, Division of Human Nutrition and Health, Wageningen, The Netherlands
| |
Collapse
|
48
|
Damanti S, Cilla M, Cilona M, Fici A, Merolla A, Pacioni G, De Lorenzo R, Martinenghi S, Vitali G, Magnaghi C, Fumagalli A, Gennaro Mazza M, Benedetti F, Tresoldi M, Rovere Querini P. Prevalence of Long COVID-19 Symptoms After Hospital Discharge in Frail and Robust Patients. Front Med (Lausanne) 2022; 9:834887. [PMID: 35911387 PMCID: PMC9329529 DOI: 10.3389/fmed.2022.834887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
Background A motley postacute symptomatology may develop after COVID-19, irrespective of the acute disease severity, age, and comorbidities. Frail individuals have reduced physiological reserves and manifested a worse COVID-19 course, during the acute setting. However, it is still unknown, whether frailty may subtend some long COVID-19 manifestations. We explored the prevalence of long COVID-19 disturbs in COVID-19 survivals. Methods This was an observational study. Patients aged 65 years or older were followed-up 1, 3, and 6 months after hospitalization for COVID-19 pneumonia. Results A total of 382 patients were enrolled. Frail patients were more malnourished (median Mini Nutritional Assessment Short Form score 8 vs. 9, p = 0.001), at higher risk of sarcopenia [median Strength, Assistance with walking, Rising from a chair, Climbing stairs, and Falls (SARC-F) score 3 vs. 1.5, p = 0.003], and manifested a worse physical performance [median Short Physical Performance Battery (SPPB) score 10 vs. 11, p = 0.0007] than robust individuals, after hospital discharge following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pneumonia. Frailty was significantly associated with: (i) confusion, as a presenting symptom of COVID-19 [odds ratio (OR) 77.84, 95% CI 4.23–1432.49, p = 0.003]; (ii) malnutrition (MNA-SF: adjusted B –5.63, 95% CI –8.39 to –2.87, p < 0.001), risk of sarcopenia (SARC-F: adjusted B 9.11, 95% CI 3.10–15.13, p = 0.003), impaired muscle performance (SPPB: B –3.47, 95% CI –6.33 to –0.61, p = 0.02), complaints in mobility (adjusted OR 1674200.27, 95% CI 4.52–619924741831.25, p = 0.03), in self-care (adjusted OR 553305.56, 95% CI 376.37–813413358.35, p < 0.001), and in performing usual activities of daily living (OR 71.57, 95% CI 2.87–1782.53, p = 0.009) at 1-month follow-up; (iii) dyspnea [modified Medical Research Council (mMRC): B 4.83, 95% CI 1.32–8.33, p = 0.007] and risk of sarcopenia (SARC-F: B 7.12, 95% CI 2.17–12.07, p = 0.005) at 3-month follow-up; and (iv) difficulties in self-care (OR 2746.89, 95% CI 6.44–1172310.83, p = 0.01) at the 6-month follow-up. In a subgroup of patients (78 individuals), the prevalence of frailty increased at the 1-month follow-up compared to baseline (p = 0.009). Conclusion The precocious identification of frail COVID-19 survivors, who manifest more motor and respiratory complaints during the follow-up, could improve the long-term management of these COVID-19 sequelae.
Collapse
Affiliation(s)
- Sarah Damanti
- Unit of General Medicine and Advanced Care, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Institute, Milan, Italy
- *Correspondence: Sarah Damanti,
| | - Marta Cilla
- Unit of General Medicine and Advanced Care, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Institute, Milan, Italy
| | - Maria Cilona
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Aldo Fici
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Aurora Merolla
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giacomo Pacioni
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Rebecca De Lorenzo
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Sabina Martinenghi
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giordano Vitali
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cristiano Magnaghi
- Department of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Anna Fumagalli
- COVID Trial Unit, Department of Internal Medicine, IRCCS San Raffaele Institute, Milan, Italy
| | - Mario Gennaro Mazza
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milan, Italy
| | - Francesco Benedetti
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milan, Italy
| | - Moreno Tresoldi
- Unit of General Medicine and Advanced Care, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Institute, Milan, Italy
| | - Patrizia Rovere Querini
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
- Department of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
49
|
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: 72] [Impact Index Per Article: 24.0] [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
|
50
|
Oh TS, Zabalawi M, Jain S, Long D, Stacpoole PW, McCall CE, Quinn MA. Dichloroacetate improves systemic energy balance and feeding behavior during sepsis. JCI Insight 2022; 7:153944. [PMID: 35730570 PMCID: PMC9309051 DOI: 10.1172/jci.insight.153944] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by dysregulated host response to an infection. The metabolic aberrations associated with sepsis underly an acute and organism-wide hyperinflammatory response and multiple organ dysfunction; however, crosstalk between systemic metabolomic alterations and metabolic reprogramming at organ levels remains unknown. We analyzed substrate utilization by the respiratory exchange ratio, energy expenditure, metabolomic screening, and transcriptional profiling in a cecal ligation and puncture model to show that sepsis increases circulating free fatty acids and acylcarnitines but decreases levels of amino acids and carbohydrates, leading to a drastic shift in systemic fuel preference. Comparative analysis of previously published metabolomics from septic liver indicated a positive correlation with hepatic and plasma metabolites during sepsis. In particular, glycine deficiency was a common abnormality of the plasma and liver during sepsis. Interrogation of the hepatic transcriptome in septic mice suggested that the septic liver may contribute to systemic glycine deficiency by downregulating genes involved in glycine synthesis. Interestingly, intraperitoneal injection of the pyruvate dehydrogenase kinase (PDK) inhibitor dichloroacetate reversed sepsis-induced anorexia, energy imbalance, inflammation, dyslipidemia, hypoglycemia, and glycine deficiency. Collectively, our data indicated that PDK inhibition rescued systemic energy imbalance and metabolic dysfunction in sepsis partly through restoration of hepatic fuel metabolism.
Collapse
Affiliation(s)
- Tae Seok Oh
- Department of Pathology, Section on Comparative Medicine, and
| | - Manal Zabalawi
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Shalini Jain
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - David Long
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Peter W. Stacpoole
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine and Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Charles E. McCall
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Matthew A. Quinn
- Department of Pathology, Section on Comparative Medicine, and,Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|