Bioenergetic Failure Drives Functional Exhaustion of Monocytes in Acute-on-Chronic Liver Failure

Deepanshu Maheshwari, Dhananjay Kumar, Rakesh Kumar Jagdish, Nidhi Nautiyal, Ashinikumar Hidam, Rekha Kumari, Rashi Sehgal, Nirupama Trehanpati, Sukriti Baweja, Guresh Kumar, Swati Sinha, Meenu Bajpai, Viniyendra Pamecha, Chhagan Bihari, Rakhi Maiwall, Shiv Kumar Sarin, Anupam Kumar, Deepanshu Maheshwari, Dhananjay Kumar, Rakesh Kumar Jagdish, Nidhi Nautiyal, Ashinikumar Hidam, Rekha Kumari, Rashi Sehgal, Nirupama Trehanpati, Sukriti Baweja, Guresh Kumar, Swati Sinha, Meenu Bajpai, Viniyendra Pamecha, Chhagan Bihari, Rakhi Maiwall, Shiv Kumar Sarin, Anupam Kumar

Abstract

Objective: The monocyte-macrophage system is central to the host's innate immune defense and in resolving injury. It is reported to be dysfunctional in acute-on-chronic liver failure (ACLF). The disease-associated alterations in ACLF monocytes are not fully understood. We investigated the mechanism of monocytes' functional exhaustion and the role of umbilical cord mesenchymal stem cells (ucMSCs) in re-energizing monocytes in ACLF.

Design: Monocytes were isolated from the peripheral blood of ACLF patients (n = 34) and matched healthy controls (n = 7) and patients with compensated cirrhosis (n = 7); phagocytic function, oxidative burst, and bioenergetics were analyzed. In the ACLF mouse model, ucMSCs were infused intravenously, and animals were sacrificed at 24 h and day 11 to assess changes in monocyte function, liver injury, and regeneration.

Results: Patients with ACLF (alcohol 64%) compared with healthy controls and those with compensated cirrhosis had an increased number of peripheral blood monocytes (p < 0.0001) which displayed significant defects in phagocytic (p < 0.0001) and oxidative burst capacity (p < 0.0001). ACLF patients also showed a significant increase in the number of liver macrophages as compared with healthy controls (p < 0.001). Bioenergetic analysis showed markedly reduced oxidative phosphorylation (p < 0.0001) and glycolysis (p < 0.001) in ACLF monocytes. Patients with monocytes having maximum mitochondrial respiration of <37.9 pmol/min [AUC = 0.822, hazard ratio (HR) = 4.5] and baseline glycolysis of ≤42.7 mpH/min (AUC = 0.901, HR = 9.1) showed increased 28-day mortality (p < 0.001). Co-culturing ACLF monocytes with ucMSC showed improved mitochondrial respiration (p < 0.01) and phagocytosis (p < 0.0001). Furthermore, ucMSC therapy increased monocyte energy (p < 0.01) and phagocytosis (p < 0.001), reduced hepatic injury, and enhanced hepatocyte regeneration in ACLF animals.

Conclusion: Bioenergetic failure drives the functional exhaustion of monocytes in ACLF. ucMSCs resuscitate monocyte energy and prevent its exhaustion. Restoring monocyte function can ameliorate hepatic injury and promote liver regeneration in the animal model of ACLF.

Keywords: acute-on-chronic liver failure (ACLF); bioenergetics; monocyte; regeneration; ucMSC therapy.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Maheshwari, Kumar, Jagdish, Nautiyal, Hidam, Kumari, Sehgal, Trehanpati, Baweja, Kumar, Sinha, Bajpai, Pamecha, Bihari, Maiwall, Sarin and Kumar.

Figures

Figure 1
Figure 1
Distribution and function of peripheral blood monocytes and liver macrophages in healthy controls and acute-on-chronic liver failure (ACLF) patients. (A) Dot plot showing changes in the percentage of peripheral blood CD14+ monocytes in ACLF compared with healthy controls and those with cirrhosis. (B) Dot plot showing changes in the percentage of peripheral blood CD14+ monocytes between subgroups of ACLF (ACLF, ACLF-SIRS, and ACLF-sepsis). (C) Dot plot showing the percentage changes in phagocytosis (left) and oxidative burst (right) of peripheral blood CD14+ monocytes in ACLF compared with healthy controls and those with cirrhosis. (D) Dot plot showing the percentage changes in phagocytosis (left) and oxidative burst (right) of peripheral blood CD14+ monocytes between subgroups of ACLF (ACLF, ACLF-SIRS, and ACLF-sepsis). (E) Dot plot showing the percentage changes in the distribution of CD68+ macrophage (left) and CD14+ monocyte (right) in ACLF compared with healthy controls. (F) Dot plot showing the percentage changes in phagocytosis of CD68+ liver macrophages in ACLF compared with healthy controls (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001). n.s. stands for non-significant.
Figure 2
Figure 2
Energy metabolism of healthy and ACLF monocytes. (A, B) Bar graph showing changes in median fluorescence intensity of (A) phagocytosis and (B) cellular ROS of healthy CD14+ monocytes in the presence of inhibitors of mitochondria and glycolysis. (C) Real-time changes in oxygen consumption rate (OCR) with subsequent treatment with oligomycin (Oligo.) FCCP and rotenone and antimycin A (Rot. + AA.) in ACLF, cirrhosis, and healthy monocytes (left). Dot plot showing changes in basal mitochondrial respiration (middle) and glycolysis (right) in ACLF monocytes compared with healthy controls and those with cirrhosis. (D) Real-time changes in OCR and dot plot showing changes in the given mitochondrial respiratory parameter of monocytes in the ACLF subgroups (ACLF, ACLF-SIRS, and ACLF-sepsis). (E) Dot plot showing changes in glycolysis (extracellular acidification rate) of monocytes in the ACLF subgroups (ACLF, ACLF-SIRS, and ACLF-sepsis). (F) MitoTracker Green staining and (G) mitochondrial functionality based on MitoTracker Red vs. MitoTracker Green percentage in ACLF monocytes compared with healthy controls (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001).
Figure 3
Figure 3
Association of systemic inflammation and endotoxemia with monocyte mitochondrial respiration defects in patients with ACLF. (A) Dot plot showing changes in plasma level of pro-inflammatory and anti-inflammatory cytokines [tumor necrosis factor-alpha (TNF-α); interleukins (IL), 6, 12, 1β, 10, 1RA, 13, and 5] in the ACLF subgroups (ACLF, ACLF-SIRS, and ACLF-sepsis). (B) Dot plot showing changes in plasma endotoxin levels in the ACLF subgroups (ACLF, ACLF-SIRS, and ACLF-sepsis). (C) Real-time changes in oxygen consumption rate (OCR) with subsequent treatment with oligomycin (Oligo.) FCCP and rotenone and antimycin A (Rot. + AA.) in healthy monocytes treated with healthy plasma (hP), ACLF plasma (aP), and ACLF plasma with LPS (aP + LPS) (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001). n.s. stands for non-significant.
Figure 4
Figure 4
Effect of monocyte energy metabolism on ACLF patients’ outcomes. (A) Real-time changes in oxygen consumption rate (OCR) with subsequent treatment with oligomycin (Oligo.) FCCP and rotenone and antimycin A (Rot. + AA.) (left). Dot plot showing changes in basal mitochondrial respiration (middle) and maximum mitochondrial respiration (right) of monocytes in ACLF survivors and non-survivors. (B) Dot plot showing changes in glycolysis (extracellular acidification rate) of monocytes in ACLF survivors and non-survivors. (C) Kaplan–Meyer survivorship graph showing changes in monocyte maximal respiration, basal respiration, and glycolysis (extracellular acidification rate) in ACLF survivors vs. non-survivors (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001).
Figure 5
Figure 5
Effect of umbilical cord mesenchymal stem cells (ucMSCs) on ACLF monocyte bioenergy and function. (A) Representative immunofluorescence confocal image of MSC donating its mitochondria to the ACLF monocyte (×60). (B) Representative histogram (left) and dot plot (right) showing mitochondrial uptake by ACLF monocytes measured through flow cytometry. (C) Representative histograms and dot plot showing changes in MitoTracker Green (total mitochondria) and MitoTracker Red (functional mitochondria) intensity in ACLF monocytes co-cultured with ucMSCs. (D) Real-time changes in oxygen consumption rate (OCR) with subsequent treatment with oligomycin (Oligo.) FCCP and rotenone and antimycin A (Rot. + AA.) in ACLF patients’ monocytes co-cultured with ucMSC, compared with control. (E) Dot plot showing changes in phagocytic activity of ACLF monocytes co-cultured with ucMSCs compared with control (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001).
Figure 6
Figure 6
Effect of ucMSC therapy on monocyte energy and function, liver injury, and regeneration in the animal model of ACLF. (A) Schematic diagram of MSC therapy in the ACLF animal model. (B) Dot plot showing changes in CD115+ bone marrow monocyte phagocytosis at 24 h and day 11 in control and ucMSC-treated animals. (C) Real-time changes in oxygen consumption rate (OCR) with subsequent treatment with oligomycin (Oligo.) FCCP and rotenone and antimycin A (Rot. + AA.) in CD115+ bone marrow monocytes in control and ucMSC-treated animals. (D) Representative images showing F4/80+ liver macrophages in liver tissue sections (left) and bar graphs showing changes in the number of F4/80+ cells (middle) and F4/80+ liver macrophage phagocytosis (right) in control and ucMSC-treated animals at the given time points. (E) Representative images showing hematoxylin and eosin staining of liver tissue and TUNEL+ hepatocytes (left). Dot plot showing the number of ballooned hepatocytes and TUNEL+ hepatocytes (right) in control and ucMSC-treated animals at the given time points. (F) Representative images showing PCNA+ hepatocytes in liver tissue sections (left). Bar graph showing changes in the number of PCNA+ hepatocytes (right) in control and ucMSC-treated animals at the given time points (*p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001). n.s. stands for non-significant.
Figure 7
Figure 7
Diagram showing the proposed mechanism of ucMSC therapy in ACLF. Increased systemic accumulation of DAMPS, PAMPS, and inflammatory mediators in response to acute liver injury in cirrhosis (a condition called ACLF) induces mitochondrial damage and bioenergy failure in monocytes. A broad defect in energy metabolism drives the exhaustion of phagocytic and oxidative burst function of monocytes in ACLF, required for the effective clearance of pathogens and cellular debris. This may account for the poor resolution of liver injury and infection, leading to regeneration failure, sepsis, shock, and death in ACLF. ucMSCs rejuvenate monocyte energy and function, prevent liver injury, and potentiate regeneration in ACLF animals.

References

    1. Moreau R, Jalan R, Gines P, Pavesi M, Angeli P, Cordoba J, et al. . Acute-on-Chronic Liver Failure is a Distinct Syndrome That Develops in Patients With Acute Decompensation of Cirrhosis. Gastroenterology (2013) 144(7):1426–37 37.e1-9. doi: 10.1053/j.gastro.2013.02.042
    1. Sarin SK, Choudhury A, Sharma MK, Maiwall R, Al Mahtab M, Rahman S, et al. . Acute-On-Chronic Liver Failure: Consensus Recommendations of the Asian Pacific Association for the Study of the Liver (APASL): An Update. Hepatol Int (2019) 13(4):353–90. doi: 10.1007/s12072-019-09946-3
    1. Choudhury A, Jindal A, Maiwall R, Sharma MK, Sharma BC, Pamecha V, et al. . Liver Failure Determines the Outcome in Patients of Acute-on-Chronic Liver Failure (ACLF): Comparison of APASL ACLF Research Consortium (AARC) and CLIF-SOFA Models. Hepatol Int (2017) 11(5):461–71. doi: 10.1007/s12072-017-9816-z
    1. Tandon P, Garcia-Tsao G. Bacterial Infections, Sepsis, and Multiorgan Failure in Cirrhosis. Semin Liver Dis (2008) 28(1):26–42. doi: 10.1055/s-2008-1040319
    1. Sarin SK, Choudhury A. Acute-on-Chronic Liver Failure: Terminology, Mechanisms and Management. Nat Rev Gastroenterol Hepatol (2016) 13(3):131–49. doi: 10.1038/nrgastro.2015.219
    1. Meersseman P, Langouche L, du Plessis J, Korf H, Mekeirele M, Laleman W, et al. . The Intensive Care Unit Course and Outcome in Acute-on-Chronic Liver Failure are Comparable to Other Populations. J Hepatol (2018) 69(4):803–9. doi: 10.1016/j.jhep.2018.04.025
    1. Arroyo V, Moreau R, Jalan R. Acute-On-Chronic Liver Failure. N Engl J Med (2020) 382(22):2137–45. doi: 10.1056/NEJMra1914900
    1. Shubham S, Kumar D, Rooge S, Maras JS, Maheshwari D, Nautiyal N, et al. . Cellular and Functional Loss of Liver Endothelial Cells Correlates With Poor Hepatocyte Regeneration in Acute-on-Chronic Liver Failure. Hepatol Int (2019) 13(6):777–87. doi: 10.1007/s12072-019-09983-y
    1. Choudhury A, Kumar M, Sharma BC, Maiwall R, Pamecha V, Moreau R, et al. . Systemic Inflammatory Response Syndrome in Acute-on-Chronic Liver Failure: Relevance of 'Golden Window': A Prospective Study. J Gastroenterol Hepatol (2017) 32(12):1989–97. doi: 10.1111/jgh.13799
    1. Fernandez J, Acevedo J, Wiest R, Gustot T, Amoros A, Deulofeu C, et al. . Bacterial and Fungal Infections in Acute-on-Chronic Liver Failure: Prevalence, Characteristics and Impact on Prognosis. Gut (2018) 67(10):1870–80. doi: 10.1136/gutjnl-2017-314240
    1. Wong F, Piano S, Singh V, Bartoletti M, Maiwall R, Alessandria C, et al. . Clinical Features and Evolution of Bacterial Infection-Related Acute-on-Chronic Liver Failure. J Hepatol (2021) 74(2):330–9. doi: 10.1016/j.jhep.2020.07.046
    1. Mucke MM, Rumyantseva T, Mucke VT, Schwarzkopf K, Joshi S, Kempf VAJ, et al. . Bacterial Infection-Triggered Acute-on-Chronic Liver Failure Is Associated With Increased Mortality. Liver Int (2018) 38(4):645–53. doi: 10.1111/liv.13568
    1. Zeng Z, Surewaard BG, Wong CH, Geoghegan JA, Jenne CN, Kubes P. CRIg Functions as a Macrophage Pattern Recognition Receptor to Directly Bind and Capture Blood-Borne Gram-Positive Bacteria. Cell Host Microbe (2016) 20(1):99–106. doi: 10.1016/j.chom.2016.06.002
    1. Broadley SP, Plaumann A, Coletti R, Lehmann C, Wanisch A, Seidlmeier A, et al. . Dual-Track Clearance of Circulating Bacteria Balances Rapid Restoration of Blood Sterility With Induction of Adaptive Immunity. Cell Host Microbe (2016) 20(1):36–48. doi: 10.1016/j.chom.2016.05.023
    1. You Q, Holt M, Yin H, Li G, Hu CJ, Ju C. Role of Hepatic Resident and Infiltrating Macrophages in Liver Repair After Acute Injury. Biochem Pharmacol (2013) 86(6):836–43. doi: 10.1016/j.bcp.2013.07.006
    1. Triantafyllou E, Pop OT, Possamai LA, Wilhelm A, Liaskou E, Singanayagam A, et al. . MerTK Expressing Hepatic Macrophages Promote the Resolution of Inflammation in Acute Liver Failure. Gut (2018) 67(2):333–47. doi: 10.1136/gutjnl-2016-313615
    1. Shi Y, Wu W, Yang Y, Yang Q, Song G, Wu Y, et al. . Decreased Tim-3 Expression is Associated With Functional Abnormalities of Monocytes in Decompensated Cirrhosis Without Overt Bacterial Infection. J Hepatol (2015) 63(1):60–7. doi: 10.1016/j.jhep.2015.02.020
    1. Antoniades CG, Wendon J, Vergani D. Paralysed Monocytes in Acute on Chronic Liver Disease. J Hepatol (2005) 42(2):163–5. doi: 10.1016/j.jhep.2004.12.005
    1. Bernsmeier C, Triantafyllou E, Brenig R, Lebosse FJ, Singanayagam A, Patel VC, et al. . CD14(+) CD15(-) HLA-DR(-) Myeloid-Derived Suppressor Cells Impair Antimicrobial Responses in Patients With Acute-on-Chronic Liver Failure. Gut (2018) 67(6):1155–67. doi: 10.1136/gutjnl-2017-314184
    1. O'Brien AJ, Fullerton JN, Massey KA, Auld G, Sewell G, James S, et al. . Immunosuppression in Acutely Decompensated Cirrhosis Is Mediated by Prostaglandin E2. Nat Med (2014) 20(5):518–23. doi: 10.1038/nm.3516
    1. Bernsmeier C, Pop OT, Singanayagam A, Triantafyllou E, Patel VC, Weston CJ, et al. . Patients With Acute-on-Chronic Liver Failure Have Increased Numbers of Regulatory Immune Cells Expressing the Receptor Tyrosine Kinase MERTK. Gastroenterology (2015) 148(3):603–15.e14. doi: 10.1053/j.gastro.2014.11.045
    1. Korf H, du Plessis J, van Pelt J, De Groote S, Cassiman D, Verbeke L, et al. . Inhibition of Glutamine Synthetase in Monocytes From Patients With Acute-on-Chronic Liver Failure Resuscitates Their Antibacterial and Inflammatory Capacity. Gut (2019) 68(10):1872–83. doi: 10.1136/gutjnl-2018-316888
    1. O'Neill LA, Hardie DG. Metabolism of Inflammation Limited by AMPK and Pseudo-Starvation. Nature (2013) 493(7432):346–55. doi: 10.1038/nature11862
    1. Zhu X, Meyers A, Long D, Ingram B, Liu T, Yoza BK, et al. . Frontline Science: Monocytes Sequentially Rewire Metabolism and Bioenergetics During an Acute Inflammatory Response. J Leukoc Biol (2019) 105(2):215–28. doi: 10.1002/JLB.3HI0918-373R
    1. Biswas SK, Mantovani A. Orchestration of Metabolism by Macrophages. Cell Metab (2012) 15(4):432–7. doi: 10.1016/j.cmet.2011.11.013
    1. Garaude J, Acin-Perez R, Martinez-Cano S, Enamorado M, Ugolini M, Nistal-Villan E, et al. . Mitochondrial Respiratory-Chain Adaptations in Macrophages Contribute to Antibacterial Host Defense. Nat Immunol (2016) 17(9):1037–45. doi: 10.1038/ni.3509
    1. Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, et al. . Succinate is an Inflammatory Signal That Induces IL-1beta Through HIF-1alpha. Nature (2013) 496(7444):238–42. doi: 10.1038/nature11986
    1. Mills EL, Kelly B, Logan A, Costa ASH, Varma M, Bryant CE, et al. . Succinate Dehydrogenase Supports Metabolic Repurposing of Mitochondria to Drive Inflammatory Macrophages. Cell (2016) 167(2):457–70.e13. doi: 10.1016/j.cell.2016.08.064
    1. Rodriguez-Prados JC, Traves PG, Cuenca J, Rico D, Aragones J, Martin-Sanz P, et al. . Substrate Fate in Activated Macrophages: A Comparison Between Innate, Classic, and Alternative Activation. J Immunol (2010) 185(1):605–14. doi: 10.4049/jimmunol.0901698
    1. Zhang S, Weinberg S, DeBerge M, Gainullina A, Schipma M, Kinchen JM, et al. . Efferocytosis Fuels Requirements of Fatty Acid Oxidation and the Electron Transport Chain to Polarize Macrophages for Tissue Repair. Cell Metab (2019) 29(2):443–56.e5. doi: 10.1016/j.cmet.2018.12.004
    1. Zhao Q, Chu Z, Zhu L, Yang T, Wang P, Liu F, et al. . 2-Deoxy-D-Glucose Treatment Decreases Anti-Inflammatory M2 Macrophage Polarization in Mice With Tumor and Allergic Airway Inflammation. Front Immunol (2017) 8:637. doi: 10.3389/fimmu.2017.00637
    1. Lachmandas E, Boutens L, Ratter JM, Hijmans A, Hooiveld GJ, Joosten LA, et al. . Microbial Stimulation of Different Toll-Like Receptor Signalling Pathways Induces Diverse Metabolic Programmes in Human Monocytes. Nat Microbiol (2016) 2:16246. doi: 10.1038/nmicrobiol.2016.246
    1. Cheng SC, Scicluna BP, Arts RJ, Gresnigt MS, Lachmandas E, Giamarellos-Bourboulis EJ, et al. . Broad Defects in the Energy Metabolism of Leukocytes Underlie Immunoparalysis in Sepsis. Nat Immunol (2016) 17(4):406–13. doi: 10.1038/ni.3398
    1. Wiest R, Lawson M, Geuking M. Pathological Bacterial Translocation in Liver Cirrhosis. J Hepatol (2014) 60(1):197–209. doi: 10.1016/j.jhep.2013.07.044
    1. Schierwagen R, Alvarez-Silva C, Madsen MSA, Kolbe CC, Meyer C, Thomas D, et al. . Circulating Microbiome in Blood of Different Circulatory Compartments. Gut (2019) 68(3):578–80. doi: 10.1136/gutjnl-2018-316227
    1. Hackstein CP, Assmus LM, Welz M, Klein S, Schwandt T, Schultze J, et al. . Gut Microbial Translocation Corrupts Myeloid Cell Function to Control Bacterial Infection During Liver Cirrhosis. Gut (2017) 66(3):507–18. doi: 10.1136/gutjnl-2015-311224
    1. Nautiyal N, Maheshwari D, Tripathi DM, Kumar D, Kumari R, Gupta S, et al. . Establishment of a Murine Model of Acute-on-Chronic Liver Failure With Multi-Organ Dysfunction. Hepatol Int (2021) 15(6):1389–1401. doi: 10.21203/-438031/v1
    1. Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, et al. . Mitochondrial Transfer From Bone-Marrow-Derived Stromal Cells to Pulmonary Alveoli Protects Against Acute Lung Injury. Nat Med (2012) 18(5):759–65. doi: 10.1038/nm.2736
    1. Phinney DG, Di Giuseppe M, Njah J, Sala E, Shiva S, St Croix CM, et al. . Mesenchymal Stem Cells Use Extracellular Vesicles to Outsource Mitophagy and Shuttle microRNAs. Nat Commun (2015) 6:8472. doi: 10.1038/ncomms9472
    1. Li C, Cheung MKH, Han S, Zhang Z, Chen L, Chen J, et al. . Mesenchymal Stem Cells and Their Mitochondrial Transfer: A Double-Edged Sword. Biosci Rep (2019) 39(5):BSR20182417. doi: 10.1042/BSR20182417
    1. Xiang X, Feng D, Hwang S, Ren T, Wang X, Trojnar E, et al. . Interleukin-22 Ameliorates Acute-on-Chronic Liver Failure by Reprogramming Impaired Regeneration Pathways in Mice. J Hepatol (2020) 72(4):736–45. doi: 10.1016/j.jhep.2019.11.013
    1. Claria J, Stauber RE, Coenraad MJ, Moreau R, Jalan R, Pavesi M, et al. . Systemic Inflammation in Decompensated Cirrhosis: Characterization and Role in Acute-on-Chronic Liver Failure. Hepatology (2016) 64(4):1249–64. doi: 10.1002/hep.28740
    1. Serezani CH, Chung J, Ballinger MN, Moore BB, Aronoff DM, Peters-Golden M. Prostaglandin E2 Suppresses Bacterial Killing in Alveolar Macrophages by Inhibiting NADPH Oxidase. Am J Respir Cell Mol Biol (2007) 37(5):562–70. doi: 10.1165/rcmb.2007-0153OC
    1. Aronoff DM, Canetti C, Peters-Golden M. Prostaglandin E2 Inhibits Alveolar Macrophage Phagocytosis Through an E-Prostanoid 2 Receptor-Mediated Increase in Intracellular Cyclic AMP. J Immunol (2004) 173(1):559–65. doi: 10.4049/jimmunol.173.1.559
    1. Minhas PS, Latif-Hernandez A, McReynolds MR, Durairaj AS, Wang Q, Rubin A, et al. . Restoring Metabolism of Myeloid Cells Reverses Cognitive Decline in Ageing. Nature (2021) 590(7844):122–8. doi: 10.1038/s41586-020-03160-0
    1. Moreau R, Claria J, Aguilar F, Fenaille F, Lozano JJ, Junot C, et al. . Blood Metabolomics Uncovers Inflammation-Associated Mitochondrial Dysfunction as a Potential Mechanism Underlying ACLF. J Hepatol (2020) 72(4):688–701. doi: 10.1016/j.jhep.2019.11.009
    1. Minhas PS, Liu L, Moon PK, Joshi AU, Dove C, Mhatre S, et al. . Macrophage De Novo NAD(+) Synthesis Specifies Immune Function in Aging and Inflammation. Nat Immunol (2019) 20(1):50–63. doi: 10.1038/s41590-018-0255-3
    1. Maiwall R, Bajpai M, Choudhury AK, Kumar A, Sharma MK, Duan Z, et al. . Therapeutic Plasma-Exchange Improves Systemic Inflammation and Survival in Acute-on-Chronic Liver Failure: A Propensity-Score Matched Study From AARC. Liver Int (2021) 41(5):1083–96. doi: 10.1111/liv.14806
    1. Horak J, Nalos L, Martinkova V, Benes J, Stengl M, Matejovic M. Mesenchymal Stem Cells in Sepsis and Associated Organ Dysfunction: A Promising Future or Blind Alley? Stem Cells Int (2017) 2017:7304121. doi: 10.1155/2017/7304121
    1. Shi M, Zhang Z, Xu R, Lin H, Fu J, Zou Z, et al. . Human Mesenchymal Stem Cell Transfusion Is Safe and Improves Liver Function in Acute-on-Chronic Liver Failure Patients. Stem Cells Transl Med (2012) 1(10):725–31. doi: 10.5966/sctm.2012-0034
    1. Li YH, Xu Y, Wu HM, Yang J, Yang LH, Yue-Meng W. Umbilical Cord-Derived Mesenchymal Stem Cell Transplantation in Hepatitis B Virus Related Acute-On-Chronic Liver Failure Treated With Plasma Exchange and Entecavir: A 24-Month Prospective Study. Stem Cell Rev Rep (2016) 12(6):645–53. doi: 10.1007/s12015-016-9683-3
    1. Starkey Lewis P, Campana L, Aleksieva N, Cartwright JA, Mackinnon A, O'Duibhir E, et al. . Alternatively Activated Macrophages Promote Resolution of Necrosis Following Acute Liver Injury. J Hepatol (2020) 73(2):349–60. doi: 10.1016/j.jhep.2020.02.031

Source: PubMed

3
Abonnere