1
|
Zhang X, van Greevenbroek MMJ, Scheijen JLJM, Eussen SJPM, Kelly J, Stehouwer CDA, Schalkwijk CG, Wouters K. Fasting plasma methylglyoxal concentrations are associated with higher numbers of circulating intermediate and non-classical monocytes but with lower activation of intermediate monocytes: the Maastricht Study. J Endocrinol Invest 2025; 48:1257-1268. [PMID: 39847265 DOI: 10.1007/s40618-025-02536-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/12/2025] [Indexed: 01/24/2025]
Abstract
PURPOSE Elevated methylglyoxal (MGO) levels and altered immune cell responses are observed in diabetes. MGO is thought to modulate immune cell activation. The current study investigated whether fasting or post-glucose-load plasma MGO concentrations are associated with circulating immune cell counts and activation in a large cohort study. METHODS 696 participants of The Maastricht Study (age 60.3 ± 8.4 years, 51.9% women) underwent an oral glucose tolerance test (OGTT). Fasting and post-OGTT plasma MGO concentrations were measured using mass spectrometry. Numbers and activation of circulating immune cells at fasting state were quantified using flow cytometry. Activation scores were calculated by averaging individual marker z-scores for neutrophils (CD11b, CD11c, CD16) and classical, intermediate, and non-classical monocytes (CD11b, CD11c, CX3XR1, HLA-DR). Associations were analysed using multiple linear regression adjusted for potential confounders. Stratified analyses were performed for glucose metabolism status for associations between plasma MGO levels and immune cell counts. RESULTS Higher fasting plasma MGO concentrations were significantly associated with higher numbers of intermediate (β = 0.09 [95%CI 0.02; 0.17]) and non-classical monocytes (0.08 [0.002; 0.15]), but with lower activation scores for the intermediate monocytes (-0.14 [-0.22; -0.06]). Stratified analyses showed that positive associations between fasting plasma MGO levels and numbers of intermediate and non-classical monocytes appear only in participants with type 2 diabetes. Post-OGTT plasma MGO concentrations were not consistently associated with immune cells counts or activation. CONCLUSION Higher fasting plasma MGO concentrations are associated with higher intermediate and non-classical monocyte counts but with lower activation of intermediate monocytes.
Collapse
Affiliation(s)
- Xiaodi Zhang
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, 6229ER, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, the Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, 6229ER, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, the Netherlands
| | - Jean L J M Scheijen
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, 6229ER, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, the Netherlands
| | - Simone J P M Eussen
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, the Netherlands
- Department of Epidemiology, Maastricht University, Maastricht, 6229HA, the Netherlands
- CAPHRI School for Care and Public Health Research Unit, Maastricht University, Maastricht, 6229ER, the Netherlands
| | - Jaycey Kelly
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, 6229ER, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, 6229ER, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, 6229ER, the Netherlands.
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, the Netherlands.
| | - Kristiaan Wouters
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, 6229ER, the Netherlands.
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, 6229ER, the Netherlands.
| |
Collapse
|
2
|
Herta T, Bhattacharyya A, Hippenstiel S, Zahlten J. The role of KLF4 in phagocyte activation during infectious diseases. Front Immunol 2025; 16:1584873. [PMID: 40313940 PMCID: PMC12044337 DOI: 10.3389/fimmu.2025.1584873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Phagocytes, including granulocytes (especially neutrophils), monocytes, macrophages, and dendritic cells, are essential components of the innate immune system, bridging innate and adaptive immunity. Their activation and function are tightly regulated by transcription factors that coordinate immune responses. Among these, Krüppel-like factor 4 (KLF4) has gained attention as a regulator of phagocyte differentiation, polarization, and inflammatory modulation. However, its role is highly context-dependent, exhibiting both pro- and anti-inflammatory properties based on environmental signals, cellular states, and the invading pathogen. KLF4 influences monocyte-to-macrophage differentiation and shapes macrophage polarization, promoting either inflammatory or regulatory phenotypes depending on external cues. In neutrophils, it affects reactive oxygen species production and immune activation, while in dendritic cells, it regulates monocyte-to-dendritic cell differentiation and cytokine secretion. Its diverse involvements in immune responses suggests that it contributes to maintaining a balance between effective pathogen defense and the prevention of excessive and potentially harmful inflammation. This review summarizes current knowledge on the function of KLF4 in phagocytes during infections, highlighting its regulatory mechanisms, context-dependent roles, and its impact on immune activation and resolution. Additionally, potential implications for therapeutic interventions targeting KLF4 are discussed.
Collapse
Affiliation(s)
- Toni Herta
- Department of Hepatology and Gastroenterology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité –Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Aritra Bhattacharyya
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Janine Zahlten
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
3
|
Oliveira LPG, Xavier RG, Nora CCV, Mangueira CLP, Rosseto EA, Aloia T, Gil JZ, Neto AS, Lopes FBTP, Carvalho KI. Exhaustion profile on classical monocytes after LPS stimulation on Crohn's disease patients. Hum Immunol 2025; 86:111257. [PMID: 39952081 DOI: 10.1016/j.humimm.2025.111257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
Crohn's disease is a type of inflammatory bowel disease that leads to symptoms such as diarrhea, abdominal pain, weight loss, and increased risk of developing tumors. The immune system plays a vital role in the gastrointestinal tract by maintaining tolerance to commensal antigens and food. However, in Crohn's disease, this tolerance mechanism is disrupted, resulting in chronic inflammatory responses. The involvement of the immune system is central to Crohn's disease, with a wide range of immune cells including monocytes, being affected. Due to the limited understanding of the role of monocytes in Crohn's disease, our study aimed to clarify the cytokine production and activation profile of monocytes subsets in the context of this condition. We used multiparametric flow cytometry to analyze the status of monocyte, quantified gene expression using qPCR, and created a correlation matrix to connect the flow cytometry data with the qPCR results through a bioinformatics approach. Our findings indicate that patients with Crohn's disease show a reduction in all monocyte subsets. Additionally, classical monocytes exhibit an exhaustion profile characterized by increased CD38 expression and reduced IL-1β production following LPS stimulation in patient groups. These results suggest that monocyte subsets play distinct roles in the disease's pathophysiology of Crohn's disease, potentially contributing to chronic inflammation and impairing the resolution of the immune response.
Collapse
Affiliation(s)
| | - Rafaela Gomes Xavier
- Instituto de Ensino e Pesquisa Hospital Israelita Albert Einstein São Paulo Brazil
| | | | | | | | - Thiago Aloia
- Instituto de Ensino e Pesquisa Hospital Israelita Albert Einstein São Paulo Brazil
| | | | | | | | - Karina Inacio Carvalho
- Instituto de Ensino e Pesquisa Hospital Israelita Albert Einstein São Paulo Brazil; Case Comprehensive Cancer Center, Case Western Reserve University Cleveland OH USA.
| |
Collapse
|
4
|
Jernigan JE, Staley HA, Baty Z, Bolen ML, Gomes BN, Holt J, Cole CL, Neighbarger NK, Dheeravath K, Merchak AR, Menees KB, Coombes SA, Tansey MG. RGS10 attenuates systemic immune dysregulation induced by chronic inflammatory stress. J Neuroinflammation 2025; 22:49. [PMID: 39994765 PMCID: PMC11852585 DOI: 10.1186/s12974-024-03322-1] [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: 11/01/2024] [Accepted: 12/10/2024] [Indexed: 02/26/2025] Open
Abstract
Regulator of G-protein signaling 10 (RGS10), a key homeostatic regulator of immune cells, has been implicated in multiple diseases associated with aging and chronic inflammation including Parkinson's Disease (PD). Interestingly, subjects with idiopathic PD display reduced levels of RGS10 in subsets of peripheral immune cells. Additionally, individuals with PD have been shown to have increased activated peripheral immune cells in cerebrospinal fluid (CSF) compared to age-matched healthy controls. However, it is unknown whether peripheral immune cells in the CSF of individuals with PD also exhibit decreased levels of RGS10. Utilizing the Michael J. Fox Foundation Parkinson's Progression Markers Initiative (PPMI) study we found that RGS10 levels are decreased in the CSF of individuals with PD compared to healthy controls and prodromal individuals. As RGS10 levels are decreased in the CSF and circulating peripheral immune cells of individuals with PD, we hypothesized that RGS10 regulates peripheral immune cell responses to chronic systemic inflammation (CSI) prior to the onset of neurodegeneration. To test this, we induced CSI for 6 weeks in C57BL6/J mice and RGS10 KO mice to assess circulating and CNS-associated immune cell responses. We found that RGS10 deficiency synergizes with CSI to induce a bias for inflammatory and cytotoxic cell populations, a reduction in antigen presentation machinery in peripheral blood immune cells, as well as in and around the brain that is most notable in males. These results highlight RGS10 as an important regulator of the systemic immune response to CSI and implicate RGS10 as a potential contributor to the development of immune dysregulation in PD.
Collapse
Affiliation(s)
- Janna E Jernigan
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Hannah A Staley
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Zachary Baty
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL, USA
| | - MacKenzie L Bolen
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Beatriz Nuñes Gomes
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jenny Holt
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Cassandra L Cole
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Noelle K Neighbarger
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kruthika Dheeravath
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Andrea R Merchak
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kelly B Menees
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Stephen A Coombes
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL, USA
| | - Malú Gámez Tansey
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA.
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
5
|
Tian X, Li J, van Bentem K, Lindelauf C, Kapsenberg JM, van der Keur C, Lashley LEELO, van Unen V, Roelen DL, Koning F, Eikmans M, van der Hoorn MLP. Peripheral monocyte subsets are altered during gestation in oocyte donation pregnancy complicated with pre-eclampsia. Scand J Immunol 2025; 101:e13432. [PMID: 39920890 PMCID: PMC11806125 DOI: 10.1111/sji.13432] [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: 06/18/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 02/09/2025]
Abstract
Oocyte donation (OD) pregnancies show a higher fetal-maternal incompatibility and a higher risk of developing pre-eclampsia (PE) than autologous pregnancies. As maternal monocytes play a role in the tolerization of the allogeneic fetus, the aim of this study was to analyse monocyte phenotypes in healthy and PE OD pregnancies. We collected maternal peripheral blood at different gestational time points in healthy (n = 10) and PE (n = 5) OD pregnancies. Fetal-maternal human leukocyte antigen (HLA) mismatches were calculated. We used a 35-colour antibody panel for Aurora spectral flow cytometry to analyse the composition and surface marker expression of monocyte subsets. Expression of CD38 on intermediate monocytes significantly increased throughout gestation in healthy OD pregnancies. Compared with the healthy group, the PE group exhibited even higher CD38 expression on monocyte subsets, with statistical significance. Immune inhibiting receptors CD85j (LILRB1) and CD85d (LILRB2), as well as monocyte recruitment regulating molecules CCR2 and CD91, also showed significantly enhanced expression on monocyte subsets during PE. When comparing healthy and PE OD only in pregnancies with high HLA mismatches, the different CD38 and CD85j expression in monocyte subsets was still significant. In conclusion, in healthy OD pregnancies, the upregulated CD38 expression might reflect a proinflammatory condition specifically at the third trimester. In PE OD pregnancies, expression of both inflammatory and immune regulatory markers is increased in maternal peripheral monocyte subsets. The elevated expression of CCR2 and CD91 on these subsets might reflect monocyte chemotaxis and the effect from systemic vascular dysfunction at the late stage of PE.
Collapse
Affiliation(s)
- Xuezi Tian
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- Department of Obstetrics and Gynecology, Leiden University Medical Center, Leiden, Netherlands
| | - Jia Li
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Kim van Bentem
- Department of Obstetrics and Gynecology, Leiden University Medical Center, Leiden, Netherlands
| | - Ciska Lindelauf
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Carin van der Keur
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Lisa E E L O Lashley
- Department of Obstetrics and Gynecology, Leiden University Medical Center, Leiden, Netherlands
| | - Vincent van Unen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Dave L Roelen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Frits Koning
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | |
Collapse
|
6
|
Rao M, McGonagill PW, Brackenridge S, Remy KE, Caldwell CC, Hotchkiss RS, Moldawer LL, Griffith TS, Badovinac VP. FUNCTIONAL IMMUNOPHENOTYPING FOR PRECISION THERAPIES IN SEPSIS. Shock 2025; 63:189-201. [PMID: 39617419 DOI: 10.1097/shk.0000000000002511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
ABSTRACT Sepsis remains a significant cause of morbidity and mortality worldwide. Although many more patients are surviving the acute event, a substantial number enters a state of persistent inflammation and immunosuppression, rendering them more vulnerable to infections. Modulating the host immune response has been a focus of sepsis research for the past 50 years, yet novel therapies have been few and far between. Although many septic patients have similar clinical phenotypes, pathways affected by the septic event differ not only between individuals but also within an individual over the course of illness. These differences ultimately impact overall immune function and response to treatment. Defining the immune state, or endotype, of an individual is critical to understanding which patients will respond to a particular therapy. In this review, we highlight current approaches to define the immune endotype and propose that these technologies may be used to "prescreen" individuals to determine which therapies are most likely to be beneficial.
Collapse
Affiliation(s)
- Mahil Rao
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Patrick W McGonagill
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Scott Brackenridge
- Department of Surgery, Harborview Medical Center, University of Washington School of Medicine, Seattle, Washington
| | - Kenneth E Remy
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Charles C Caldwell
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Lyle L Moldawer
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, Florida
| | | | | |
Collapse
|
7
|
Caldwell BA, Ie S, Lucas A, Li L. Ticam2 ablation facilitates monocyte exhaustion recovery after sepsis. Sci Rep 2025; 15:2059. [PMID: 39814939 PMCID: PMC11735619 DOI: 10.1038/s41598-025-86103-x] [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: 09/10/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Sepsis is a leading cause of death worldwide, with most patient mortality stemming from lingering immunosuppression in sepsis survivors. This is due in part to immune dysfunction resulting from monocyte exhaustion, a phenotype of reduced antigen presentation, altered CD14/CD16 inflammatory subtypes, and disrupted cytokine production. Whereas previous research demonstrated improved sepsis survival in Ticam2-/- mice, the contribution of TICAM2 to long-term exhaustion memory remained unknown. Using a cecal slurry injection sepsis model, we monitored the establishment and recovery of monocyte exhaustion in Ticam2-/- mice. After one week of recovery, we profiled bone marrow and splenic reservoir monocytes in Ticam2-/- mice and found that, in contrast to the persistent exhaustion observed in wild-type monocytes, Ticam2-/- monocytes largely resembled healthy controls. To determine the impact of TICAM2 ablation on innate epigenetic memory in sepsis, we measured genome-wide DNA methylation in bone marrow monocytes and found that Ticam2-/- cells exhibit a unique profile of altered methylation at CEBPE binding sites and regulatory features for key immune genes such as Dmkn and Btg1. Bearing human translational relevance, a case study of time course blood samples collected from a sepsis patient presenting with SIRS and a positive qSOFA revealed a similar effect in human monocytes, which steadily transition into an exhausted memory characterized by a CD38high; CX3CR1low; HLA-DRlow state within four days of hospital admittance. Together, our data reveal the chronic preservation of monocyte exhaustion, partially controlled by TICAM2.
Collapse
Affiliation(s)
- Blake A Caldwell
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Susanti Ie
- Carillion Roanoke Memorial Hospital, Roanoke, VA, 24014, USA
| | - Amy Lucas
- Carillion Roanoke Memorial Hospital, Roanoke, VA, 24014, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA.
| |
Collapse
|
8
|
Drago E, Fioredda F, Penco F, Prigione I, Bertoni A, Del Zotto G, Bocca P, Massaccesi E, Lanciotti M, Moratto D, Thurner L, Caorsi R, Gattorno M, Volpi S. Inborn Error of WAS Presenting with SARS-CoV-2-Related Multisystem Inflammatory Syndrome in Children. J Clin Immunol 2024; 45:49. [PMID: 39581942 DOI: 10.1007/s10875-024-01840-4] [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: 12/03/2023] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) has been reported in patients with inborn errors of immunity (IEI), providing insights into disease pathogenesis. Here, we present the first case of MIS-C in a child affected by Wiskott-Aldrich syndrome (WAS) gene mutation, elucidating underlying predisposing factors and the involved inflammatory pathways. Genetic analysis revealed a frameshift truncating variant in the WAS gene, resulting in WAS protein expression between mild and severe forms, despite a clinical phenotype resembling X-linked thrombocytopenia (XLT). IL-1β secretion by LPS-stimulated peripheral blood mononuclear cells from patient during MIS-C was lower compared to healthy subjects but increased during follow-up. Conversely, the percentage of ASC (apoptosis-associated speck-like protein containing a CARD) specks in the patient's circulating monocytes during the acute phase was higher than in healthy subjects. The type I interferon (IFN) signature during MIS-C was normal, in contrast to the raised IFN signature measured far from the acute event. This case supports the association of IEI with MIS-C, potentially linked to delayed immune responses to SARS-CoV-2. The XLT phenotype underlies a subclinical immunodysregulation involving the NLRP3 inflammasome and the type-I IFN response.
Collapse
Affiliation(s)
- Enrico Drago
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Università degli Studi di Genova, Genova, Italy.
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy.
| | | | - Federica Penco
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Ignazia Prigione
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Arinna Bertoni
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Genny Del Zotto
- Core Facilities Laboratory, Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Paola Bocca
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | | | | | - Daniele Moratto
- Flow Cytometry Unit, Clinical Chemistry Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Lorenz Thurner
- José Carreras Center for Immuno and Gene Therapy, Department of Internal Medicine I, Saarland University, Homburg, Germany
| | - Roberta Caorsi
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Marco Gattorno
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Stefano Volpi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Università degli Studi di Genova, Genova, Italy
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
9
|
Cumming BM, Addicott KW, Maruri F, Pillay V, Asmal R, Moodley S, Barreto-Durate B, Araújo-Pereira M, Mazibuko M, Mhlane Z, Mbatha N, Khan K, Makhari S, Karim F, Peetluk L, Pym AS, Moosa MYS, van der Heijden YF, Sterling TS, Andrade BB, Leslie A, Steyn AJC. Longitudinal mitochondrial bioenergetic signatures of blood monocytes and lymphocytes improve during treatment of drug-susceptible pulmonary tuberculosis patients Monocyte/lymphocyte bioenergetic signatures post-TB treatment. Front Immunol 2024; 15:1465448. [PMID: 39606220 PMCID: PMC11599235 DOI: 10.3389/fimmu.2024.1465448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024] Open
Abstract
The impact of human pulmonary tuberculosis (TB) on the bioenergetic metabolism of circulating immune cells remains elusive, as does the resolution of these effects with TB treatment. In this study, the rates of oxidative phosphorylation (OXPHOS) and glycolysis in circulating lymphocytes and monocytes of patients with drug-susceptible TB at diagnosis, 2 months, and 6 months during treatment, and 12 months after diagnosis were investigated using extracellular flux analysis. At diagnosis, the bioenergetic parameters of both blood lymphocytes and monocytes of TB patients were severely impaired in comparison to non-TB and non-HIV-infected controls. However, most bioenergetic parameters were not affected by HIV status or glycemic index. Treatment of TB patients restored the % spare respiratory capacity (%SRC) of the circulating lymphocytes to that observed in non-TB and non-HIV infected controls by 12 months. Treatment also improved the maximal respiration of circulating lymphocytes and the %SRC of circulating monocytes of the TB patients. Notably, the differential correlation of the clinical and bioenergetic parameters of the monocytes and lymphocytes from the controls and TB patients at baseline and month 12 was consistent with improved metabolic health and resolution of inflammation following successful TB treatment. Network analysis of the bioenergetic parameters of circulating immune cells with serum cytokine levels indicated a highly coordinated immune response at month 6. These findings underscore the importance of metabolic health in combating TB, supporting the need for further investigation of the bioenergetic immunometabolism associated with TB infection for novel therapeutic approaches aimed at bolstering cellular energetics to enhance immune responses and expedite recovery in TB patients.
Collapse
Affiliation(s)
- Bridgette M. Cumming
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Kelvin W. Addicott
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Fernanda Maruri
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Vanessa Pillay
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Rukaya Asmal
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Sashen Moodley
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Beatriz Barreto-Durate
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | - Mariana Araújo-Pereira
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | - Matilda Mazibuko
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Zoey Mhlane
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Nikiwe Mbatha
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Khadija Khan
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Senamile Makhari
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Farina Karim
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Lauren Peetluk
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Alexander S. Pym
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | | | - Yuri F. van der Heijden
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
- Global Division, The Aurum Institute, Johannesburg, South Africa
| | - Timothy S. Sterling
- Vanderbilt Tuberculosis Center, Vanderbilt University School of Medicine, Nashville, TN, United States
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Bruno B. Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Laboratório de Pesquisa Clínica e Translacional, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
| | - Alasdair Leslie
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Infectious Diseases, University of KwaZulu-Natal, Durban, South Africa
- Department of Infection and Immunity, University College of London, London, United Kingdom
| | - Adrie J. C. Steyn
- Africa Health Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
10
|
Wallings RL, McFarland K, Staley HA, Neighbarger N, Schaake S, Brüggemann N, Zittel S, Usnich T, Klein C, Sammler EM, Tansey MG. The R1441C-Lrrk2 mutation induces myeloid immune cell exhaustion in an age- and sex-dependent manner in mice. Sci Transl Med 2024; 16:eadl1535. [PMID: 39504353 DOI: 10.1126/scitranslmed.adl1535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/19/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024]
Abstract
Age is the greatest risk factor for many neurodegenerative diseases, yet immune system aging, a contributor to neurodegeneration, is understudied. Genetic variation in the LRRK2 gene affects risk for both familial and sporadic Parkinson's disease (PD). The leucine-rich repeat kinase 2 (LRRK2) protein is implicated in peripheral immune cell signaling, but the effects of an aging immune system on LRRK2 function remain unclear. We analyzed peritoneal macrophages from R1441C-Lrrk2 knock-in mice and observed a biphasic, age-dependent effect of the R1441C-Lrrk2 mutation on peritoneal macrophage function. We report increases in antigen presentation, anti-inflammatory cytokine production, lysosomal activity, and pathogen uptake in peritoneal macrophages from young (2- to 3-month-old) female R1441C-Lrrk2 mice. Conversely, macrophages from aged (18- to 21-month-old) female R1441C-Lrrk2 mice exhibited decreased antigen presentation after inflammatory insult, decreased lysosomal function, and pathogen uptake, with a concomitant increase in DNA fragmentation in the presence of pathogens. This immune cell exhaustion phenotype was not observed in male R1441C-Lrrk2 mice and was driven by increased LRRK2 protein kinase activity. This phenotype was also observed in human peripheral myeloid cells, with monocyte-derived macrophages from patients with PD who had either the R1441C- or Y1699C-LRRK2 mutation exhibiting decreased pathogen uptake and increased PDL1 expression, consistent with immune cell exhaustion. Our findings that LRRK2 mutations conferred an immunological advantage at a young age but could predispose the carrier to age-acquired immune cell exhaustion have implications for the therapeutic development of LRRK2 inhibitors.
Collapse
Affiliation(s)
- Rebecca L Wallings
- Department of Neuroscience, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Karen McFarland
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Department of Neurology and Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
- Department of Pharmacology, Emory University, Atlanta, GA, USA
| | - Hannah A Staley
- Department of Neuroscience, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
| | - Noelle Neighbarger
- Department of Neuroscience, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
| | - Susen Schaake
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Norbert Brüggemann
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Zittel
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tatiana Usnich
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Klein
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Esther M Sammler
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
- Department of Neurology, School of Medicine, Ninewells Hospital, Ninewells Drive, Dundee DD1 9SY, UK
| | - Malú Gámez Tansey
- Department of Neuroscience, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, College of Medicine, McKnight Brain Institute, Gainesville, FL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Neurology and Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| |
Collapse
|
11
|
Jernigan JE, Staley HA, Baty Z, Bolen ML, Gomes BN, Holt J, Cole CL, Neighbarger NK, Dheeravath K, Merchak AR, Menees KB, Coombes SA, Tansey MG. RGS10 Attenuates Systemic Immune Dysregulation Induced by Chronic Inflammatory Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620078. [PMID: 39554164 PMCID: PMC11566001 DOI: 10.1101/2024.10.24.620078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Regulator of G-protein signaling 10 (RGS10), a key homeostatic regulator of immune cells, has been implicated in multiple diseases associated with aging and chronic inflammation including Parkinson's Disease (PD). Interestingly, subjects with idiopathic PD display reduced levels of RGS10 in subsets of peripheral immune cells. Additionally, individuals with PD have been shown to have increased activated peripheral immune cells in cerebral spinal fluid (CSF) compared to age-matched healthy controls. However, it is unknown whether CSF-resident peripheral immune cells in individuals with PD also exhibit decreased levels of RGS10. Therefore, we performed an analysis of RGS10 levels in the proteomic database of the CSF from the Michael J. Fox Foundation Parkinson's Progression Markers Initiative (PPMI) study. We found that RGS10 levels are decreased in the CSF of individuals with PD compared to healthy controls and prodromal individuals. Moreover, we find that RGS10 levels decrease with age but not PD progression and that males have less RGS10 than females in PD. Importantly, studies have established an association between chronic systemic inflammation (CSI) and neurodegenerative diseases, such as PD, and known sources of CSI have been identified as risk factors for developing PD; however, the role of peripheral immune cell dysregulation in this process has been underexplored. As RGS10 levels are decreased in the CSF and circulating peripheral immune cells of individuals with PD, we hypothesized that RGS10 regulates peripheral immune cell responses to CSI prior to the onset of neurodegeneration. To test this, we induced CSI for 6 weeks in C57BL6/J mice and RGS10 KO mice to assess circulating and CNS-associated peripheral immune cell responses. We found that RGS10 deficiency synergizes with CSI to induce a bias for inflammatory and cytotoxic cell populations, a reduction in antigen presentation in peripheral blood immune cells, as well as in and around the brain that is most notable in males. These results highlight RGS10 as an important regulator of the systemic immune response to CSI and implicate RGS10 as a potential contributor to the development of immune dysregulation in PD.
Collapse
Affiliation(s)
- Janna E. Jernigan
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Hannah A. Staley
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Zachary Baty
- Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL USA
| | - MacKenzie L. Bolen
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Beatriz Nuñes Gomes
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jenny Holt
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Cassandra L. Cole
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Noelle K. Neighbarger
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kruthika Dheeravath
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Andrea R. Merchak
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kelly B. Menees
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Stephen A. Coombes
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL USA
| | - Malú Gámez Tansey
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| |
Collapse
|
12
|
Geng S, Lu R, Zhang Y, Wu Y, Xie L, Caldwell BA, Pradhan K, Yi Z, Hou J, Xu F, Chen X, Li L. Monocytes Reprogrammed by 4-PBA Potently Contribute to the Resolution of Inflammation and Atherosclerosis. Circ Res 2024; 135:856-872. [PMID: 39224974 PMCID: PMC11424066 DOI: 10.1161/circresaha.124.325023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Chronic inflammation initiated by inflammatory monocytes underlies the pathogenesis of atherosclerosis. However, approaches that can effectively resolve chronic low-grade inflammation targeting monocytes are not readily available. The small chemical compound 4-phenylbutyric acid (4-PBA) exhibits broad anti-inflammatory effects in reducing atherosclerosis. Selective delivery of 4-PBA reprogrammed monocytes may hold novel potential in providing targeted and precision therapeutics for the treatment of atherosclerosis. METHODS Systems analyses integrating single-cell RNA sequencing and complementary immunologic approaches characterized key resolving characteristics as well as defining markers of reprogrammed monocytes trained by 4-PBA. Molecular mechanisms responsible for monocyte reprogramming were assessed by integrated biochemical and genetic approaches. The intercellular propagation of homeostasis resolution was evaluated by coculture assays with donor monocytes trained by 4-PBA and recipient naive monocytes. The in vivo effects of monocyte resolution and atherosclerosis prevention by 4-PBA were assessed with the high-fat diet-fed ApoE-/- mouse model with IP 4-PBA administration. Furthermore, the selective efficacy of 4-PBA-trained monocytes was examined by IV transfusion of ex vivo trained monocytes by 4-PBA into recipient high-fat diet-fed ApoE-/- mice. RESULTS In this study, we found that monocytes can be potently reprogrammed by 4-PBA into an immune-resolving state characterized by reduced adhesion and enhanced expression of anti-inflammatory mediator CD24. Mechanistically, 4-PBA reduced the expression of ICAM-1 (intercellular adhesion molecule 1) via reducing peroxisome stress and attenuating SYK (spleen tyrosine kinase)-mTOR (mammalian target of rapamycin) signaling. Concurrently, 4-PBA enhanced the expression of resolving mediator CD24 through promoting PPARγ (peroxisome proliferator-activated receptor γ) neddylation mediated by TOLLIP (toll-interacting protein). 4-PBA-trained monocytes can effectively propagate anti-inflammation activity to neighboring monocytes through CD24. Our data further demonstrated that 4-PBA-trained monocytes effectively reduce atherosclerosis pathogenesis when administered in vivo. CONCLUSIONS Our study describes a robust and effective approach to generate resolving monocytes, characterizes novel mechanisms for targeted monocyte reprogramming, and offers a precision therapeutics for atherosclerosis based on delivering reprogrammed resolving monocytes.
Collapse
Affiliation(s)
- Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Ran Lu
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Yajun Wu
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Ling Xie
- Department of Biochemistry and Molecular Biology, University of North Carolina at Chappell Hill, NC (L.X., X.C.)
| | - Blake A Caldwell
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Kisha Pradhan
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Ziyue Yi
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Jacqueline Hou
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Feng Xu
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| | - Xian Chen
- Department of Biochemistry and Molecular Biology, University of North Carolina at Chappell Hill, NC (L.X., X.C.)
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg (S.G., R.L., Y.Z., Y.W., B.A.C., K.P., Z.Y., J.H., F.X., L.L.)
| |
Collapse
|
13
|
Clanchy FI, Borghese F, Bystrom J, Balog A, Penn H, Hull DN, Mageed RA, Taylor PC, Williams RO. Inflammatory disease status and response to TNF blockade are associated with mechanisms of endotoxin tolerance. J Autoimmun 2024; 148:103300. [PMID: 39116634 DOI: 10.1016/j.jaut.2024.103300] [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: 03/16/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024]
Abstract
The mechanisms of endotoxin tolerance (ET), which down-regulate inflammation, are well described in response to exogenous toll-like receptor ligands, but few studies have focused on ET-associated mechanisms in inflammatory disease. As blocking TNF can attenuate the development of ET, the effect of anti-TNF on the expression of key ET-associated molecules in inflammatory auto-immune disease was measured; changes in inflammatory gene expression were confirmed using an ET bioassay. The expression of immunomodulatory molecules was measured in a murine model of arthritis treated with anti-TNF and the expression of ET-associated molecules was measured in whole blood in rheumatoid arthritis (RA) and ankylosing spondylitis (AS) patients, before and after therapy. The expression of ET-associated genes was also measured in RA patient monocytes before and after therapy, in anti-TNF responders and non-responders. Tnfaip3, Ptpn6 and Irak3 were differentially expressed in affected paws, spleens, lymph nodes and circulating leucocytes in experimental murine arthritis treated with anti-TNF. Prior to therapy, the expression of TNFAIP3, INPP5D, PTPN6, CD38 and SIGIRR in whole blood differed between human healthy controls and RA or AS patients. In blood monocytes from RA patients, the expression of TNFAIP3 was significantly reduced by anti-TNF therapy in non-responders. Prior to therapy, anti-TNF non-responders had higher expression of TNFAIP3 and SLPI, compared to responders. Although the expression of TNFAIP3 was significantly higher in RA non-responders prior to treatment, the post-treatment reduction to a level similar to responders did not coincide with a clinical response to therapy.
Collapse
Affiliation(s)
- Felix Il Clanchy
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, United Kingdom; Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.
| | - Federica Borghese
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Jonas Bystrom
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Attila Balog
- Department of Rheumatology and Immunology, Szent-Györgyi Albert Clinical Centre, University of Szeged, Szeged, Hungary
| | - Henry Penn
- Northwick Park Hospital, Harrow, United Kingdom
| | - Dobrina N Hull
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Rizgar A Mageed
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Richard O Williams
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| |
Collapse
|
14
|
Wang W, Ma L, Liu B, Ouyang L. The role of trained immunity in sepsis. Front Immunol 2024; 15:1449986. [PMID: 39221248 PMCID: PMC11363069 DOI: 10.3389/fimmu.2024.1449986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction syndrome caused by dysregulated host response to infection, characterized by a systemic inflammatory response to infection. The use of antibiotics, fluid resuscitation, and organ support therapy has limited prognostic benefit in patients with sepsis, and its incidence is not diminishing, which is attracting increased attention in medicine. Sepsis remains one of the most debilitating and expensive illnesses. One of the main reasons of septic mortality is now understood to be disruption of immune homeostasis. Immunotherapy is revolutionizing the treatment of illnesses in which dysregulated immune responses play a significant role. This "trained immunity", which is a potent defense against infection regardless of the type of bacteria, fungus, or virus, is attributed to the discovery that the innate immune cells possess immune memory via metabolic and epigenetic reprogramming. Here we reviewed the immunotherapy of innate immune cells in sepsis, the features of trained immunity, and the relationship between trained immunity and sepsis.
Collapse
Affiliation(s)
| | | | | | - Liangliang Ouyang
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
15
|
Zhang Y, Lee C, Geng S, Wang J, Bohara U, Hou J, Yi Z, Li L. Immune-enhancing neutrophils reprogrammed by subclinical low-dose endotoxin in cancer treatment. EMBO Mol Med 2024; 16:1886-1900. [PMID: 39009886 PMCID: PMC11319772 DOI: 10.1038/s44321-024-00100-7] [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: 10/24/2023] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024] Open
Abstract
Despite the re-emergence of the pioneering "Coley's toxin" concept in anti-cancer immune therapies highlighted by check-point inhibitors and CAR-T approaches, fundamental mechanisms responsible for the immune-enhancing efficacy of low-dose "Coley's toxin" remain poorly understood. This study examines the novel reprogramming of immune-enhancing neutrophils by super-low dose endotoxin conducive for anti-cancer therapies. Through integrated analyses including scRNAseq and functional characterizations, we examined the efficacy of reprogrammed neutrophils in treating experimental cancer. We observed that neutrophils trained by super-low dose endotoxin adopt a potent immune-enhancing phenotype characterized by CD177loCD11bloCD80hiCD40hiDectin2hi. Both murine and human neutrophils trained by super-low dose endotoxin exhibit relieved suppression of adaptive T cells as compared to un-trained neutrophils. Functionally, neutrophils trained by super-low dose endotoxin can potently reduce tumor burden when transfused into recipient tumor-bearing mice. Mechanistically, Super-low dose endotoxin enables the generation of immune-enhancing neutrophils through activating STAT5 and reducing innate suppressor IRAK-M. Together, our data clarify the long-held mystery of "Coley's toxin" in rejuvenating anti-tumor immune defense, and provide a proof-of-concept in developing innate neutrophil-based anti-tumor therapeutics.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Christina Lee
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Jing Wang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Udipta Bohara
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Jacqueline Hou
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Ziyue Yi
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA.
| |
Collapse
|
16
|
Barrios EL, Rincon JC, Willis M, Polcz VE, Leary J, Darden DB, Balch JA, Larson SD, Loftus TJ, Mohr AM, Wallet S, Brusko MA, Balzano-Nogueira L, Cai G, Sharma A, Upchurch GR, Kladde MP, Mathews CE, Maile R, Moldawer LL, Bacher R, Efron PA. TRANSCRIPTOMIC DIFFERENCES IN PERIPHERAL MONOCYTE POPULATIONS IN SEPTIC PATIENTS BASED ON OUTCOME. Shock 2024; 62:208-216. [PMID: 38713581 PMCID: PMC11892173 DOI: 10.1097/shk.0000000000002379] [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: 05/09/2024]
Abstract
ABSTRACT Postsepsis early mortality is being replaced by survivors who experience either a rapid recovery and favorable hospital discharge or the development of chronic critical illness with suboptimal outcomes. The underlying immunological response that determines these clinical trajectories remains poorly defined at the transcriptomic level. As classical and nonclassical monocytes are key leukocytes in both the innate and adaptive immune systems, we sought to delineate the transcriptomic response of these cell types. Using single-cell RNA sequencing and pathway analyses, we identified gene expression patterns between these two groups that are consistent with differences in TNF-α production based on clinical outcome. This may provide therapeutic targets for those at risk for chronic critical illness in order to improve their phenotype/endotype, morbidity, and long-term mortality.
Collapse
Affiliation(s)
- Evan L. Barrios
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Jaimar C. Rincon
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Micah Willis
- Department of Oral Biology, College of Dentistry, Gainesville, FL, USA
| | - Valerie E. Polcz
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - John Leary
- Department of Biostatistics, College of Medicine, Gainesville, FL, USA
| | - Dijoia B. Darden
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Jeremy A. Balch
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Shawn D. Larson
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Tyler J. Loftus
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Alicia M. Mohr
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Shannon Wallet
- Department of Oral Biology, College of Dentistry, Gainesville, FL, USA
| | - Maigan A. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Gainesville, FL, USA
| | | | - Guoshuai Cai
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Ashish Sharma
- Department of Surgery, College of Medicine, Gainesville, FL, USA
| | | | - Michael P. Kladde
- Department of Biochemistry and Molecular Biology, College of Medicine, Gainesville, FL, USA
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Gainesville, FL, USA
| | - Robert Maile
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Lyle L. Moldawer
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Rhonda Bacher
- Department of Biostatistics, College of Medicine, Gainesville, FL, USA
| | - Philip A. Efron
- Sepsis and Critical Illness Research Center, Department of Surgery, College of Medicine, Gainesville, FL, USA
| |
Collapse
|
17
|
Wang Q, Tang J, Li Y, Lu J, Yang D, He C, Li T, Fu K, Liu R. EFFECT OF STRATIFIED DOSE OF NOREPINEPHRINE ON CELLULAR IMMUNE RESPONSE IN PATIENTS WITH SEPTIC SHOCK AND THE CONSTRUCTION OF A PROGNOSTIC RISK MODEL. Shock 2024; 62:32-43. [PMID: 38517239 DOI: 10.1097/shk.0000000000002363] [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: 03/23/2024]
Abstract
ABSTRACT Objective: To explore the effect of a stratified dose of norepinephrine (NE) on cellular immune response in patients with septic shock, and to construct a prognostic model of septic shock. Methods: A total of 160 patients with septic shock (B group) and 58 patients with sepsis (A group) were given standard cluster therapy. Patients with septic shock were divided into four groups (B1-B4 groups: 0.01-0.2, 0.2-0.5, 0.5-1.0, and >1 μg/kg/min) according to the quartile method of the early (72 h) time-weighted average dose of NE and clinical application. The cellular immune indexes at 24 h (T0) and 4-7 days (T1) after admission were collected. The difference method was used to explore the effect of NE stratified dose on cellular immune effect in patients with septic shock. A multivariate COX proportional risk regression model was used to analyze the independent prognostic risk factors, and a prognostic risk model was constructed. Results: The differences of ΔIL-1β, ΔIL-6, ΔIL-10, absolute value difference of T lymphocyte (ΔCD3+/CD45+#) and Th helper T cell (ΔCD3+ CD4+/CD45+#), CD64 infection index difference, ΔmHLA-DR, regulatory T lymphocyte ratio difference (ΔTregs%) between group A, B1, B2, B3, and B4 were statistically significant ( P < 0.05). There was a nonlinear relation between the stratified dose of NE and ΔIL-6, ΔIL-10, ΔCD3+/CD45+#, ΔmHLA-DR%. The threshold periods of NE-induced proinflammatory and anti-inflammatory immune changes were 0.3-0.5 μg/kg/min. Multivariate COX model regression analysis showed that age, nutritional patterns, weighted average dose of norepinephrine, IL-6, absolute value of T lymphocytes, and mHLA-DR were independent risk factors affecting the prognosis of patients with septic shock ( P < 0.05). The prognostic risk model was constructed (AUC value = 0.813, 95% CI: 0.752-0.901). Conclusion: NE has a certain inhibitory effect on cellular immune function in patients with septic shock. A prognostic risk model was constructed with stronger prediction efficiency for the prognosis of patients with septic shock.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Geriatric Intensive Care Medicine, the First Affiliated Hospital of Kunming Medical University, Yunnan Geriatric Medicine Center, Kunming, China
| | - Jiefu Tang
- Department of Geriatric Intensive Care Medicine, the First Affiliated Hospital of Kunming Medical University, Yunnan Geriatric Medicine Center, Kunming, China
| | - Yao Li
- Stomatology Research Center, The First People's Hospital of Yunnan Province, Kunming, China
| | - Jiafei Lu
- Department of Geriatric Intensive Care Medicine, the First Affiliated Hospital of Kunming Medical University, Yunnan Geriatric Medicine Center, Kunming, China
| | - Dexing Yang
- Department of Emergency Medicine, Yunnan First People's Hospital, Kunming, China
| | - Chen He
- Department of Geriatric Intensive Care Medicine, the First Affiliated Hospital of Kunming Medical University, Yunnan Geriatric Medicine Center, Kunming, China
| | - Ting Li
- Department of Geriatric Intensive Care Medicine, the First Affiliated Hospital of Kunming Medical University, Yunnan Geriatric Medicine Center, Kunming, China
| | - Kai Fu
- Department of Geriatric Intensive Care Medicine, the First Affiliated Hospital of Kunming Medical University, Yunnan Geriatric Medicine Center, Kunming, China
| | - Rong Liu
- Department of Geriatric Intensive Care Medicine, the First Affiliated Hospital of Kunming Medical University, Yunnan Geriatric Medicine Center, Kunming, China
| |
Collapse
|
18
|
Jolly KJ, Zhang F. IVT-mRNA reprogramming of myeloid cells for cancer immunotherapy. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:247-288. [PMID: 39034054 DOI: 10.1016/bs.apha.2024.04.004] [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: 07/23/2024]
Abstract
In the past decade, in vitro transcribed messenger RNAs (IVT-mRNAs) have emerged as promising therapeutic molecules. The clinical success of COVID-19 mRNA vaccines developed by Pfizer-BioNTech and Moderna, have demonstrated that IVT-mRNAs can be safely and successfully used in a clinical setting, and efforts are underway to develop IVT-mRNAs for therapeutic applications. Current applications of mRNA-based therapy have been focused on (1) mRNA vaccines for infectious diseases and cancer treatment; (2) protein replacement therapy; (3) gene editing therapy; and (4) cell-reprogramming therapies. Due to the recent clinical progress of cell-based immunotherapies, the last direction-the use of IVT-mRNAs as a therapeutic approach to program immune cells for the treatment of cancer has received extensive attention from the cancer immunotherapy field. Myeloid cells are important components of our immune system, and they play critical roles in mediating disease progression and regulating immunity against diseases. In this chapter, we discussed the progress of using IVT-mRNAs as a therapeutic approach to program myeloid cells against cancer and other immune-related diseases. Towards this direction, we first reviewed the pharmacology of IVT-mRNAs and the biology of myeloid cells as well as myeloid cell-targeting therapeutics. We then presented a few cases of current IVT-mRNA-based approaches to target and reprogram myeloid cells for disease treatment and discussed the advantages and limitations of these approaches. Finally, we presented our considerations in designing mRNA-based approaches to target myeloid cells for disease treatment.
Collapse
Affiliation(s)
- Kevon J Jolly
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Fan Zhang
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, United States; Department of Chemical Engineering, College of Engineering, University of Florida, Gainesville, FL, United States; Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
19
|
Rees A, Jenkins BJ, Angelini R, Davies LC, Cronin JG, Jones N, Thornton CA. Immunometabolic adaptation in monocytes underpins functional changes during pregnancy. iScience 2024; 27:109779. [PMID: 38736550 PMCID: PMC11088341 DOI: 10.1016/j.isci.2024.109779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/02/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Metabolic heterogeneity is a determinant of immune cell function. The normal physiological metabolic reprogramming of pregnancy that ensures the fuel requirements of mother and baby are met, might also underpin changes in immunity that occur with pregnancy and manifest as altered responses to pathogens and changes to autoimmune disease symptoms. Using peripheral blood from pregnant women at term, we reveal that monocytes lose M2-like and gain M1-like properties accompanied by reductions in mitochondrial mass, maximal respiration, and cardiolipin content in pregnancy; glycolysis is unperturbed. We establish that muramyl dipeptide (MDP)-stimulated cytokine production relies on oxidative metabolism, then show in pregnancy reduced cytokine production in response to MDP but not LPS. Overall, mitochondrially centered metabolic capabilities of late gestation monocytes are down-regulated revealing natural plasticity in monocyte phenotype and function that could reveal targets for improving pregnancy outcomes but also yield alternative therapeutic approaches to diverse metabolic and/or immune-mediated diseases beyond pregnancy.
Collapse
Affiliation(s)
- April Rees
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, Wales, UK
| | - Benjamin J. Jenkins
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, Wales, UK
| | - Roberto Angelini
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, Wales, UK
| | - Luke C. Davies
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, Wales, UK
| | - James G. Cronin
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, Wales, UK
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, Wales, UK
| | - Catherine A. Thornton
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, Wales, UK
| |
Collapse
|
20
|
Wallings R, McFarland K, Staley H, Neighbarger N, Schaake S, Brueggemann N, Zittel S, Usnich T, Klein C, Sammler E, Tansey MG. The R1441C-LRRK2 mutation induces myeloid immune cell exhaustion in an age- and sex-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.12.562063. [PMID: 37905053 PMCID: PMC10614788 DOI: 10.1101/2023.10.12.562063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Considering age is the greatest risk factor for many neurodegenerative diseases, aging, in particular aging of the immune system, is the most underappreciated and understudied contributing factor in the neurodegeneration field. Genetic variation around the LRRK2 gene affects risk of both familial and sporadic Parkinson's disease (PD). The leucine-rich repeat kinase 2 (LRRK2) protein has been implicated in peripheral immune signaling, however, the effects of an aging immune system on LRRK2 function have been neglected to be considered. We demonstrate here that the R1441C mutation induces a hyper-responsive phenotype in macrophages from young female mice, characterized by increased effector functions, including stimulation-dependent antigen presentation, cytokine release, phagocytosis, and lysosomal function. This is followed by age-acquired immune cell exhaustion in a Lrrk2-kinase-dependent manner. Immune-exhausted macrophages exhibit suppressed antigen presentation and hypophagocytosis, which is also demonstrated in myeloid cells from R1441C and Y1699C-PD patients. Our novel findings that LRRK2 mutations confer immunological advantage at a young age but may predispose the carrier to age-acquired immune exhaustion have significant implications for LRRK2 biology and therapeutic development. Indeed, LRRK2 has become an appealing target in PD, but our findings suggest that more research is required to understand the cell-type specific consequences and optimal timing of LRRK2-targeting therapeutics.
Collapse
|
21
|
Barrios EL, Leary JR, Darden DB, Rincon JC, Willis M, Polcz VE, Gillies GS, Munley JA, Dirain ML, Ungaro R, Nacionales DC, Gauthier MPL, Larson SD, Morel L, Loftus TJ, Mohr AM, Maile R, Kladde MP, Mathews CE, Brusko MA, Brusko TM, Moldawer LL, Bacher R, Efron PA. The post-septic peripheral myeloid compartment reveals unexpected diversity in myeloid-derived suppressor cells. Front Immunol 2024; 15:1355405. [PMID: 38720891 PMCID: PMC11076668 DOI: 10.3389/fimmu.2024.1355405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Sepsis engenders distinct host immunologic changes that include the expansion of myeloid-derived suppressor cells (MDSCs). These cells play a physiologic role in tempering acute inflammatory responses but can persist in patients who develop chronic critical illness. Methods Cellular Indexing of Transcriptomes and Epitopes by Sequencing and transcriptomic analysis are used to describe MDSC subpopulations based on differential gene expression, RNA velocities, and biologic process clustering. Results We identify a unique lineage and differentiation pathway for MDSCs after sepsis and describe a novel MDSC subpopulation. Additionally, we report that the heterogeneous response of the myeloid compartment of blood to sepsis is dependent on clinical outcome. Discussion The origins and lineage of these MDSC subpopulations were previously assumed to be discrete and unidirectional; however, these cells exhibit a dynamic phenotype with considerable plasticity.
Collapse
Affiliation(s)
- Evan L. Barrios
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jack R. Leary
- Department of Biostatistics, University of Florida College of Medicine and Public Health and Health Sciences, Gainesville, FL, United States
| | - Dijoia B. Darden
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jaimar C. Rincon
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Micah Willis
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Valerie E. Polcz
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Gwendolyn S. Gillies
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jennifer A. Munley
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Marvin L. Dirain
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ricardo Ungaro
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Dina C. Nacionales
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Marie-Pierre L. Gauthier
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Shawn D. Larson
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Laurence Morel
- Department of Microbiology and Immunology, University of Texas San Antonio School of Medicine, San Antonio, TX, United States
| | - Tyler J. Loftus
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Alicia M. Mohr
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Robert Maile
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Michael P. Kladde
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Clayton E. Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Maigan A. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Lyle L. Moldawer
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida College of Medicine and Public Health and Health Sciences, Gainesville, FL, United States
| | - Philip A. Efron
- Sepsis and Critical Illness Research Center, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
22
|
Padovani CM, Wilson RM, Rodriguez A, Spur BW, Yin K. Resolvin D2 attenuates LPS-induced macrophage exhaustion. FASEB J 2024; 38:e23569. [PMID: 38551610 DOI: 10.1096/fj.202302521r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Early in sepsis, a hyperinflammatory response is dominant, but later, an immunosuppressive phase dominates, and the host is susceptible to opportunistic infections. Anti-inflammatory agents may accelerate the host into immunosuppression, and few agents can reverse immunosuppression without causing inflammation. Specialized pro-resolving mediators (SPMs) such as resolvin D2 (RvD2) have been reported to resolve inflammation without being immunosuppressive, but little work has been conducted to examine their effects on immunosuppression. To assess the effects of RvD2 on immunosuppression, we established a model of macrophage exhaustion using two lipopolysaccharide (LPS) treatments or hits. THP-1 monocyte-derived macrophages were first treated with RvD2 or vehicle for 1 h. One LPS hit increased NF-κB activity 11-fold and TNF-α release 60-fold compared to unstimulated macrophages. RvD2 decreased LPS-induced NF-κB activity and TNF-α production but increased bacterial clearance. Two LPS hits reduced macrophage bacterial clearance and decreased macrophage NF-κB activity (45%) and TNF-α release (75%) compared to one LPS hit, demonstrating exhaustion. RvD2 increased NF-κB activity, TNF-α release, and bacterial clearance following two LPS hits compared to controls. TLR2 inhibition abolished RvD2-mediated changes. In a mouse sepsis model, splenic macrophage response to exogenous LPS was reduced compared to controls and was restored by in vivo administration of RvD2, supporting the in vitro results. If RvD2 was added to monocytes before differentiation into macrophages, however, RvD2 reduced LPS responses and increased bacterial clearance following both one and two LPS hits. The results show that RvD2 attenuated macrophage suppression in vitro and in vivo and that this effect was macrophage-specific.
Collapse
Affiliation(s)
- Cristina M Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Rachael M Wilson
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Ana Rodriguez
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Bernd W Spur
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| | - Kingsley Yin
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, New Jersey, USA
| |
Collapse
|
23
|
Kempaiah P, Libertin CR, Chitale RA, Naeyma I, Pleqi V, Sheele JM, Iandiorio MJ, Hoogesteijn AL, Caulfield TR, Rivas AL. Decoding Immuno-Competence: A Novel Analysis of Complete Blood Cell Count Data in COVID-19 Outcomes. Biomedicines 2024; 12:871. [PMID: 38672225 PMCID: PMC11048687 DOI: 10.3390/biomedicines12040871] [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: 02/10/2024] [Revised: 03/14/2024] [Accepted: 03/23/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND While 'immuno-competence' is a well-known term, it lacks an operational definition. To address this omission, this study explored whether the temporal and structured data of the complete blood cell count (CBC) can rapidly estimate immuno-competence. To this end, one or more ratios that included data on all monocytes, lymphocytes and neutrophils were investigated. MATERIALS AND METHODS Longitudinal CBC data collected from 101 COVID-19 patients (291 observations) were analyzed. Dynamics were estimated with several approaches, which included non-structured (the classic CBC format) and structured data. Structured data were assessed as complex ratios that capture multicellular interactions among leukocytes. In comparing survivors with non-survivors, the hypothesis that immuno-competence may exhibit feedback-like (oscillatory or cyclic) responses was tested. RESULTS While non-structured data did not distinguish survivors from non-survivors, structured data revealed immunological and statistical differences between outcomes: while survivors exhibited oscillatory data patterns, non-survivors did not. In survivors, many variables (including IL-6, hemoglobin and several complex indicators) showed values above or below the levels observed on day 1 of the hospitalization period, displaying L-shaped data distributions (positive kurtosis). In contrast, non-survivors did not exhibit kurtosis. Three immunologically defined data subsets included only survivors. Because information was based on visual patterns generated in real time, this method can, potentially, provide information rapidly. DISCUSSION The hypothesis that immuno-competence expresses feedback-like loops when immunological data are structured was not rejected. This function seemed to be impaired in immuno-suppressed individuals. While this method rapidly informs, it is only a guide that, to be confirmed, requires additional tests. Despite this limitation, the fact that three protective (survival-associated) immunological data subsets were observed since day 1 supports many clinical decisions, including the early and personalized prognosis and identification of targets that immunomodulatory therapies could pursue. Because it extracts more information from the same data, structured data may replace the century-old format of the CBC.
Collapse
Affiliation(s)
- Prakasha Kempaiah
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (P.K.); (V.P.)
| | | | - Rohit A. Chitale
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Islam Naeyma
- Department of Neuroscience, Division of QHS Computational Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (I.N.); (T.R.C.)
| | - Vasili Pleqi
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic, Jacksonville, FL 32224, USA; (P.K.); (V.P.)
| | | | - Michelle J. Iandiorio
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA;
| | | | - Thomas R. Caulfield
- Department of Neuroscience, Division of QHS Computational Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (I.N.); (T.R.C.)
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ariel L. Rivas
- Center for Global Health, Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| |
Collapse
|
24
|
Caldwell BA, Li L. Epigenetic regulation of innate immune dynamics during inflammation. J Leukoc Biol 2024; 115:589-606. [PMID: 38301269 PMCID: PMC10980576 DOI: 10.1093/jleuko/qiae026] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/03/2024] Open
Abstract
Innate immune cells play essential roles in modulating both immune defense and inflammation by expressing a diverse array of cytokines and inflammatory mediators, phagocytizing pathogens to promote immune clearance, and assisting with the adaptive immune processes through antigen presentation. Rudimentary innate immune "memory" states such as training, tolerance, and exhaustion develop based on the nature, strength, and duration of immune challenge, thereby enabling dynamic transcriptional reprogramming to alter present and future cell behavior. Underlying transcriptional reprogramming are broad changes to the epigenome, or chromatin alterations above the level of DNA sequence. These changes include direct modification of DNA through cytosine methylation as well as indirect modifications through alterations to histones that comprise the protein core of nucleosomes. In this review, we will discuss recent advances in our understanding of how these epigenetic changes influence the dynamic behavior of the innate immune system during both acute and chronic inflammation, as well as how stable changes to the epigenome result in long-term alterations of innate cell behavior related to pathophysiology.
Collapse
Affiliation(s)
- Blake A. Caldwell
- Department of Biological Sciences, Virginia Tech, 970 Washington St. SW, Blacksburg, VA 24061-0910, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, 970 Washington St. SW, Blacksburg, VA 24061-0910, USA
| |
Collapse
|
25
|
Caldwell BA, Wu Y, Wang J, Li L. Altered DNA methylation underlies monocyte dysregulation and immune exhaustion memory in sepsis. Cell Rep 2024; 43:113894. [PMID: 38442017 PMCID: PMC11654472 DOI: 10.1016/j.celrep.2024.113894] [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: 09/14/2023] [Revised: 01/12/2024] [Accepted: 02/14/2024] [Indexed: 03/07/2024] Open
Abstract
Monocytes can develop an exhausted memory state characterized by reduced differentiation, pathogenic inflammation, and immune suppression that drives immune dysregulation during sepsis. Chromatin alterations, notably via histone modifications, underlie innate immune memory, but the contribution of DNA methylation remains poorly understood. Using an ex vivo sepsis model, we show altered DNA methylation throughout the genome of exhausted monocytes, including genes implicated in immune dysregulation during sepsis and COVID-19 infection (e.g., Plac8). These changes are recapitulated in septic mice induced by cecal slurry injection. Methylation profiles developed in septic mice are maintained during ex vivo culture, supporting the involvement of DNA methylation in stable monocyte exhaustion memory. Methylome reprogramming is driven in part by Wnt signaling inhibition in exhausted monocytes and can be reversed with DNA methyltransferase inhibitors, Wnt agonists, or immune training molecules. Our study demonstrates the significance of altered DNA methylation in the maintenance of stable monocyte exhaustion memory.
Collapse
Affiliation(s)
- Blake A Caldwell
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061-0910, USA
| | - Yajun Wu
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061-0910, USA
| | - Jing Wang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061-0910, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24061-0910, USA.
| |
Collapse
|
26
|
Wu Y, Caldwell B, Wang J, Zhang Y, Li L. Alleviation of monocyte exhaustion by BCG derivative mycolic acid. iScience 2024; 27:108978. [PMID: 38323001 PMCID: PMC10845070 DOI: 10.1016/j.isci.2024.108978] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/27/2023] [Accepted: 01/16/2024] [Indexed: 02/08/2024] Open
Abstract
Monocyte exhaustion with sustained pathogenic inflammation and immune-suppression, a hallmark of sepsis resulting from systemic infections, presents a challenge with limited therapeutic solutions. This study identified Methoxy-Mycolic Acid (M-MA), a branched mycolic acid derived from Mycobacterium bovis Bacillus Calmette-Guérin (BCG), as a potent agent in alleviating monocyte exhaustion and restoring immune homeostasis. Co-treatment of monocytes with M-MA effectively blocked the expansion of Ly6Chi/CD38hi/PD-L1hi monocytes induced by LPS challenges and restored the expression of immune-enhancing CD86. M-MA treatment restored mitochondrial functions of exhausted monocytes and alleviated their suppressive activities on co-cultured T cells. Independent of TREM2, M-MA blocks Src-STAT1-mediated inflammatory polarization and reduces the production of immune suppressors TAX1BP1 and PLAC8. Whole genome methylation analyses revealed M-MA's ability to erase the methylation memory of exhausted monocytes, particularly restoring Plac8 methylation. Together, our data suggest M-MA as an effective agent in restoring monocyte homeostasis with a therapeutic potential for treating sepsis.
Collapse
Affiliation(s)
- Yajun Wu
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| | - Blake Caldwell
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| | - Jing Wang
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech; Blacksburg, VA 24061-0910, USA
| |
Collapse
|
27
|
Kosmider O, Possémé C, Templé M, Corneau A, Carbone F, Duroyon E, Breillat P, Chirayath TW, Oules B, Sohier P, Luka M, Gobeaux C, Lazaro E, Outh R, Le Guenno G, Lifermann F, Berleur M, Le Mene M, Friedrich C, Lenormand C, Weitten T, Guillotin V, Burroni B, Boussier J, Willems L, Aractingi S, Dionet L, Tharaux PL, Vergier B, Raynaud P, Ea HK, Ménager M, Duffy D, Terrier B. VEXAS syndrome is characterized by inflammasome activation and monocyte dysregulation. Nat Commun 2024; 15:910. [PMID: 38291039 PMCID: PMC10828464 DOI: 10.1038/s41467-024-44811-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Acquired mutations in the UBA1 gene were recently identified in patients with severe adult-onset auto-inflammatory syndrome called VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic). However, the precise physiological and clinical impact of these mutations remains poorly defined. Here we study a unique prospective cohort of VEXAS patients. We show that monocytes from VEXAS are quantitatively and qualitatively impaired and display features of exhaustion with aberrant expression of chemokine receptors. In peripheral blood from VEXAS patients, we identify an increase in circulating levels of many proinflammatory cytokines, including IL-1β and IL-18 which reflect inflammasome activation and markers of myeloid cells dysregulation. Gene expression analysis of whole blood confirms these findings and also reveals a significant enrichment of TNF-α and NFκB signaling pathways that can mediate cell death and inflammation. This study suggests that the control of the nflammasome activation and inflammatory cell death could be therapeutic targets in VEXAS syndrome.
Collapse
Affiliation(s)
- Olivier Kosmider
- Université de Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France.
- Hematology Laboratory, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris Cité, Cochin Hospital, Paris, France.
| | - Céline Possémé
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Marie Templé
- Université de Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
- Hematology Laboratory, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris Cité, Cochin Hospital, Paris, France
| | - Aurélien Corneau
- Sorbonne Université, Faculté de Médecine, UMS037, PASS, Plateforme de Cytométrie de la Pitié-Salpêtrière CyPS, Paris, France
| | - Francesco Carbone
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Eugénie Duroyon
- Université de Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
- Hematology Laboratory, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris Cité, Cochin Hospital, Paris, France
| | - Paul Breillat
- Université de Paris Cité, INSERM, U970, PARCC, F-, Paris, France
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP-CUP, Hôpital Cochin, Paris, France
| | | | - Bénédicte Oules
- Department of Pathology, AP-HP, APHP-CUP, Hôpital Cochin, Paris, France
| | - Pierre Sohier
- Department of Pathology, AP-HP, APHP-CUP, Hôpital Cochin, Paris, France
| | - Marine Luka
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Camille Gobeaux
- Biochemistry Laboratory, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris Cité, Cochin Hospital, Paris, France
| | - Estibaliz Lazaro
- Department of Internal Medicine, Bordeaux University Hospital-Haut-Lévêque, Pessac, France
| | - Roderau Outh
- Department of Internal Medicine, Centre Hospitalier de Perpignan, Perpignan, France
| | - Guillaume Le Guenno
- Department of Internal Medicine, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | | | - Marie Berleur
- Department of Internal Medicine, AP-HP, APHP-NUP, Hôpital Bichat, Paris, France
| | - Melchior Le Mene
- Université de Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
- Hematology Laboratory, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris Cité, Cochin Hospital, Paris, France
| | - Chloé Friedrich
- Université de Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
- Hematology Laboratory, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris Cité, Cochin Hospital, Paris, France
| | - Cédric Lenormand
- Université de Strasbourg, Department of Dermatology, CHRU Strasbourg, Strasbourg, France
| | - Thierry Weitten
- Department of Internal Medicine, Centre Hospitalier (CHICAS), Gap, France
| | - Vivien Guillotin
- Department of Internal Medicine, Bordeaux University Hospital-Saint-André, Bordeaux, France
| | - Barbara Burroni
- Department of Pathology, AP-HP, APHP-CUP, Hôpital Cochin, Paris, France
| | - Jeremy Boussier
- Sorbonne University - 47-83 Boulevard de l'Hopital, Paris, France
| | - Lise Willems
- Université de Paris Cité, Institut Cochin, CNRS UMR8104, INSERM U1016, Paris, France
- Hematology Department, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris Cité, Cochin Hospital, Paris, France
| | - Selim Aractingi
- Dermatology Department, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris Cité, Cochin Hospital, Paris, France
| | - Léa Dionet
- Université de Paris Cité, INSERM, U970, PARCC, F-, Paris, France
| | | | - Béatrice Vergier
- Pathology Department, Bordeaux University Hospital-Haut-Lévêque, Pessac, France
| | - Pierre Raynaud
- Pathology Department, Centre Hospitalier de Perpignan, Perpignan, France
| | - Hang-Korng Ea
- Université de Paris Cité, INSERM, UMR-S 1132 BIOSCAR, Paris, France
- Rheumatology Department, AP- HP, Lariboisière Hospital, Paris, France
| | - Mickael Ménager
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Darragh Duffy
- Institut Pasteur, Université de Paris Cité, Translational Immunology Unit, Paris, France
| | - Benjamin Terrier
- Université de Paris Cité, INSERM, U970, PARCC, F-, Paris, France.
- Department of Internal Medicine, National Reference Center for Rare Systemic Autoimmune Diseases, AP-HP, APHP-CUP, Hôpital Cochin, Paris, France.
| |
Collapse
|
28
|
Padovani CM, Yin K. Immunosuppression in Sepsis: Biomarkers and Specialized Pro-Resolving Mediators. Biomedicines 2024; 12:175. [PMID: 38255280 PMCID: PMC10813323 DOI: 10.3390/biomedicines12010175] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Severe infection can lead to sepsis. In sepsis, the host mounts an inappropriately large inflammatory response in an attempt to clear the invading pathogen. This sustained high level of inflammation may cause tissue injury and organ failure. Later in sepsis, a paradoxical immunosuppression occurs, where the host is unable to clear the preexisting infection and is susceptible to secondary infections. A major issue with sepsis treatment is that it is difficult for physicians to ascertain which stage of sepsis the patient is in. Sepsis treatment will depend on the patient's immune status across the spectrum of the disease, and these immune statuses are nearly polar opposites in the early and late stages of sepsis. Furthermore, there is no approved treatment that can resolve inflammation without contributing to immunosuppression within the host. Here, we review the major mechanisms of sepsis-induced immunosuppression and the biomarkers of the immunosuppressive phase of sepsis. We focused on reviewing three main mechanisms of immunosuppression in sepsis. These are lymphocyte apoptosis, monocyte/macrophage exhaustion, and increased migration of myeloid-derived suppressor cells (MDSCs). The biomarkers of septic immunosuppression that we discuss include increased MDSC production/migration and IL-10 levels, decreased lymphocyte counts and HLA-DR expression, and increased GPR18 expression. We also review the literature on the use of specialized pro-resolving mediators (SPMs) in different models of infection and/or sepsis, as these compounds have been reported to resolve inflammation without being immunosuppressive. To obtain the necessary information, we searched the PubMed database using the keywords sepsis, lymphocyte apoptosis, macrophage exhaustion, MDSCs, biomarkers, and SPMs.
Collapse
Affiliation(s)
- Cristina M. Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, NJ 08084, USA;
| | | |
Collapse
|
29
|
Ravkov EV, Williams ESCP, Elgort M, Barker AP, Planelles V, Spivak AM, Delgado JC, Lin L, Hanley TM. Reduced monocyte proportions and responsiveness in convalescent COVID-19 patients. Front Immunol 2024; 14:1329026. [PMID: 38250080 PMCID: PMC10797708 DOI: 10.3389/fimmu.2023.1329026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/06/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction The clinical manifestations of acute severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection and coronavirus disease 2019 (COVID-19) suggest a dysregulation of the host immune response that leads to inflammation, thrombosis, and organ dysfunction. It is less clear whether these dysregulated processes persist during the convalescent phase of disease or during long COVID. We sought to examine the effects of SARS-CoV-2 infection on the proportions of classical, intermediate, and nonclassical monocytes, their activation status, and their functional properties in convalescent COVID-19 patients. Methods Peripheral blood mononuclear cells (PBMCs) from convalescent COVID-19 patients and uninfected controls were analyzed by multiparameter flow cytometry to determine relative percentages of total monocytes and monocyte subsets. The expression of activation markers and proinflammatory cytokines in response to LPS treatment were measured by flow cytometry and ELISA, respectively. Results We found that the percentage of total monocytes was decreased in convalescent COVID-19 patients compared to uninfected controls. This was due to decreased intermediate and non-classical monocytes. Classical monocytes from convalescent COVID-19 patients demonstrated a decrease in activation markers, such as CD56, in response to stimulation with bacterial lipopolysaccharide (LPS). In addition, classical monocytes from convalescent COVID-19 patients showed decreased expression of CD142 (tissue factor), which can initiate the extrinsic coagulation cascade, in response to LPS stimulation. Finally, we found that monocytes from convalescent COVID-19 patients produced less TNF-α and IL-6 in response to LPS stimulation, than those from uninfected controls. Conclusion SARS-CoV-2 infection exhibits a clear effect on the relative proportions of monocyte subsets, the activation status of classical monocytes, and proinflammatory cytokine production that persists during the convalescent phase of disease.
Collapse
Affiliation(s)
- Eugene V. Ravkov
- ARUP Laboratories Institute for Clinical and Experimental Pathology, Salt Lake City, UT, United States
| | - Elizabeth S. C. P. Williams
- Department of Internal Medicine, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Marc Elgort
- ARUP Laboratories Institute for Clinical and Experimental Pathology, Salt Lake City, UT, United States
| | - Adam P. Barker
- ARUP Laboratories Institute for Clinical and Experimental Pathology, Salt Lake City, UT, United States
- Department of Pathology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Vicente Planelles
- Department of Pathology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Adam M. Spivak
- Department of Internal Medicine, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Julio C. Delgado
- ARUP Laboratories Institute for Clinical and Experimental Pathology, Salt Lake City, UT, United States
- Department of Pathology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Leo Lin
- ARUP Laboratories Institute for Clinical and Experimental Pathology, Salt Lake City, UT, United States
- Department of Pathology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Timothy M. Hanley
- ARUP Laboratories Institute for Clinical and Experimental Pathology, Salt Lake City, UT, United States
- Department of Pathology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
30
|
Wang J, Wu Y, Lin R, Zhang Y, Li L. TRAM deletion attenuates monocyte exhaustion and alleviates sepsis severity. Front Immunol 2023; 14:1297329. [PMID: 38162637 PMCID: PMC10756061 DOI: 10.3389/fimmu.2023.1297329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Monocyte exhaustion characterized by immune-suppressive features can develop during sepsis and contribute to adverse patient outcomes. However, molecular mechanisms responsible for the establishment of immune-suppressive monocytes with reduced expression of immune-enhancing mediators such as CD86 during sepsis are not well understood. In this study, we identified that the TLR4 intracellular adaptor TRAM plays a key role in mediating the sustained reduction of CD86 expression on exhausted monocytes and generating an immune-suppressive monocyte state. TRAM contributes to the prolonged suppression of CD86 through inducing TAX1BP1 as well as SARM1, collectively inhibiting Akt and NFκB. TRAM deficient mice are protected from cecal slurry-induced experimental sepsis and retain immune-competent monocytes with CD86 expression. Our data reveal a key molecular circuitry responsible for monocyte exhaustion and provide a viable target for rejuvenating functional monocytes and treating sepsis.
Collapse
Affiliation(s)
| | | | | | | | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
31
|
Geng S, Lin R, Wu Y, Wang J, Li L. Modulation of Innate Immune Memory Dynamics by Subcellular Reactive Oxygen Species. Antioxid Redox Signal 2023; 39:1027-1038. [PMID: 37082952 PMCID: PMC10715440 DOI: 10.1089/ars.2023.0304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 04/22/2023]
Abstract
Significance: Innate immune cells adopt distinct memory states during the pathogenesis of acute and chronic inflammatory diseases. Intracellular generations of reactive oxygen species (ROS) play key roles during the programming dynamics of innate immune cells such as monocytes and macrophages. Recent Advances: ROS modulate the adaptation of innate leukocytes to varying intensities and durations of inflammatory signals, facilitate fundamental reprogramming dynamics such as priming, tolerance, and exhaustion, in addition to fundamental processes of proliferation, differentiation, phagocytosis, chemotaxis, as well as expression of pro- and anti-inflammatory mediators. ROS can be generated at distinct subcellular compartments including cellular membrane, mitochondria, and peroxisome. Complex inflammatory signals may finely regulate ROS generation within distinct subcellular compartments, which in turn may differentially facilitate innate memory dynamics. Critical Issues: Complex inflammatory signals with varying strengths and durations may differentially trigger ROS generation at peroxisome, mitochondria, and other subcellular organelles. Peroxisomal or mitochondrial ROS may facilitate the assembly of distinct signaling platforms involved in the programming of memory innate leukocytes. Despite the emerging connection of subcellular ROS with innate immune memory, underlying mechanisms are still not well defined. Future Directions: Recent important discoveries linking subcellular ROS and innate memory as critically reviewed here hold novel translational relevance related to acute and chronic inflammatory diseases. Capitalizing on these novel findings, future systems studies that use next-generation single-cell dynamic analyses in response to complex inflammatory environments are urgently needed to comprehensively decipher the programming dynamics of innate immune memory, finely modulated by subcellular ROS. Antioxid. Redox Signal. 39, 1027-1038.
Collapse
Affiliation(s)
- Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - RuiCi Lin
- Program of Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, Virginia, USA
| | - Yajun Wu
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Jing Wang
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
- Program of Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, Virginia, USA
- Program in Genetics, Bioinformatics and Computational Biology, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
32
|
Zhang WY, Chen ZH, An XX, Li H, Zhang HL, Wu SJ, Guo YQ, Zhang K, Zeng CL, Fang XM. Analysis and validation of diagnostic biomarkers and immune cell infiltration characteristics in pediatric sepsis by integrating bioinformatics and machine learning. World J Pediatr 2023; 19:1094-1103. [PMID: 37115484 PMCID: PMC10533616 DOI: 10.1007/s12519-023-00717-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/10/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND Pediatric sepsis is a complicated condition characterized by life-threatening organ failure resulting from a dysregulated host response to infection in children. It is associated with high rates of morbidity and mortality, and rapid detection and administration of antimicrobials have been emphasized. The objective of this study was to evaluate the diagnostic biomarkers of pediatric sepsis and the function of immune cell infiltration in the development of this illness. METHODS Three gene expression datasets were available from the Gene Expression Omnibus collection. First, the differentially expressed genes (DEGs) were found with the use of the R program, and then gene set enrichment analysis was carried out. Subsequently, the DEGs were combined with the major module genes chosen using the weighted gene co-expression network. The hub genes were identified by the use of three machine-learning algorithms: random forest, support vector machine-recursive feature elimination, and least absolute shrinkage and selection operator. The receiver operating characteristic curve and nomogram model were used to verify the discrimination and efficacy of the hub genes. In addition, the inflammatory and immune status of pediatric sepsis was assessed using cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT). The relationship between the diagnostic markers and infiltrating immune cells was further studied. RESULTS Overall, after overlapping key module genes and DEGs, we detected 402 overlapping genes. As pediatric sepsis diagnostic indicators, CYSTM1 (AUC = 0.988), MMP8 (AUC = 0.973), and CD177 (AUC = 0.986) were investigated and demonstrated statistically significant differences (P < 0.05) and diagnostic efficacy in the validation set. As indicated by the immune cell infiltration analysis, multiple immune cells may be involved in the development of pediatric sepsis. Additionally, all diagnostic characteristics may correlate with immune cells to varying degrees. CONCLUSIONS The candidate hub genes (CD177, CYSTM1, and MMP8) were identified, and the nomogram was constructed for pediatric sepsis diagnosis. Our study could provide potential peripheral blood diagnostic candidate genes for pediatric sepsis patients.
Collapse
Affiliation(s)
- Wen-Yuan Zhang
- Department of Anesthesiology and Intensive Care, School of Medicine, The First Affiliated Hospital, Zhejiang University, QingChun Road 79, Hangzhou, 310003, China
| | - Zhong-Hua Chen
- Department of Anesthesiology and Intensive Care, School of Medicine, The First Affiliated Hospital, Zhejiang University, QingChun Road 79, Hangzhou, 310003, China
- Department of Anesthesiology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, China
| | | | - Hui Li
- Department of Anesthesiology and Intensive Care, School of Medicine, The First Affiliated Hospital, Zhejiang University, QingChun Road 79, Hangzhou, 310003, China
| | - Hua-Lin Zhang
- Department of Anesthesiology and Intensive Care, School of Medicine, The First Affiliated Hospital, Zhejiang University, QingChun Road 79, Hangzhou, 310003, China
| | - Shui-Jing Wu
- Department of Anesthesiology and Intensive Care, School of Medicine, The First Affiliated Hospital, Zhejiang University, QingChun Road 79, Hangzhou, 310003, China
| | - Yu-Qian Guo
- Department of Anesthesiology and Intensive Care, School of Medicine, The First Affiliated Hospital, Zhejiang University, QingChun Road 79, Hangzhou, 310003, China
| | - Kai Zhang
- Department of Anesthesiology and Intensive Care, School of Medicine, The First Affiliated Hospital, Zhejiang University, QingChun Road 79, Hangzhou, 310003, China
| | - Cong-Li Zeng
- Department of Anesthesiology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Xiang-Ming Fang
- Department of Anesthesiology and Intensive Care, School of Medicine, The First Affiliated Hospital, Zhejiang University, QingChun Road 79, Hangzhou, 310003, China.
| |
Collapse
|
33
|
Montserrat-de la Paz S, Del Carmen Naranjo M, Lopez S, Del Carmen Millan-Linares M, Rivas-Dominguez A, Jaramillo-Carmona SM, Abia R, Muriana FJG, Bermudez B. Immediate-release niacin and a monounsaturated fatty acid-rich meal on postprandial inflammation and monocyte characteristics in men with metabolic syndrome. Clin Nutr 2023; 42:2138-2150. [PMID: 37774650 DOI: 10.1016/j.clnu.2023.08.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND & AIM When considered separately, long-term immediate-release niacin and fatty meals enriched in monounsaturated fatty acids (MUFA) decrease postprandial triglycerides, but their effects on postprandial inflammation, which is common in individuals with metabolic syndrome, are less known. Moreover, successful combination is lacking and its impact on acute disorders of the innate immune cells in the metabolic syndrome remains unclear. Here, we aimed to establish the effects from combination with niacin of different fats [butter, enriched in saturated fatty acids (SFA), olive oil, enriched in MUFA, and olive oil supplemented with eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids] on plasma inflammatory markers and circulating monocyte subsets, activation and priming at the postprandial period in individuals with metabolic syndrome. METHODS A random-order within-subject crossover experiment was performed, in which 16 individuals with metabolic syndrome and 16 age-matched healthy volunteers took 2 g immediate-release niacin together with the corresponding fatty meal or a meal with no fat as control. In total, 128 postprandial curves were analysed. We sampled hourly over 6 h for plasma concentrations of soluble inflammatory markers and triglycerides. Circulating monocyte subsets (CD14/CD16 balance), activation (CCL2/CCR2 axis) and priming (M1/M2-like phenotype) at the time of postprandial hypertriglyceridemic peak were also addressed. RESULTS Dietary SFA (combined with niacin) promote postprandial excursions of circulating IL-6, IL-1β, TNF-α and CD14/CCR2-rich monocytes with a pro-inflammatory M1-like phenotype, particularly in individuals with metabolic syndrome. In contrast, dietary MUFA (combined with niacin) postprandially increased circulating CD16-rich monocytes with an anti-inflammatory M2-like phenotype. Omega-3 PUFA did not add to the effects of MUFA. CONCLUSION The co-administration of a single-dose of immediate-release niacin with a fatty meal rich in MUFA, in contrast to SFA, suppresses postprandial inflammation at the levels of both secretory profile and monocyte response in individuals with metabolic syndrome. These findings highlight a potential role of combining niacin and dietary MUFA for the homeostatic control of inflammation and the innate immune system, identifying a new search direction for the management of disorders associated with the metabolic syndrome.
Collapse
Affiliation(s)
- Sergio Montserrat-de la Paz
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain; Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Maria Del Carmen Naranjo
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain
| | - Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain; Department of Cell Biology, Faculty of Biology, University of Seville, 41012 Seville, Spain; Instituto de Biomedicina de Sevilla (IBiS/CSIC), Hospital Universitario Virgen del Rocio, University of Seville, 41013 Seville, Spain
| | - Maria Del Carmen Millan-Linares
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain; Cell Biology Unit, Instituto de la Grasa, CSIC, 41013 Seville, Spain
| | | | | | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain
| | - Francisco J G Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain
| | - Beatriz Bermudez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41013 Seville, Spain; Department of Cell Biology, Faculty of Biology, University of Seville, 41012 Seville, Spain.
| |
Collapse
|
34
|
Geng S, Lu R, Zhang Y, Wu Y, Xie L, Caldwell B, Pradhan K, Yi Z, Hou J, Xu F, Chen X, Li L. Monocytes reprogrammed by 4-PBA potently contribute to the resolution of inflammation and atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563200. [PMID: 37961551 PMCID: PMC10634693 DOI: 10.1101/2023.10.19.563200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Chronic inflammation initiated by inflammatory monocytes underlies the pathogenesis of atherosclerosis. However, approaches that can effectively resolve chronic low-grade inflammation targeting monocytes are not readily available. The small chemical compound 4-phenylbutyric acid (4-PBA) exhibits broad anti-inflammatory effects in reducing atherosclerosis. Selective delivery of 4-PBA reprogrammed monocytes may hold novel potential in providing targeted and precision therapeutics for the treatment of atherosclerosis. Methods Systems analyses integrating single-cell RNA-sequencing and complementary immunological approaches characterized key resolving characteristics as well as defining markers of reprogrammed monocytes trained by 4-PBA. Molecular mechanisms responsible for monocyte reprogramming was assessed by integrated biochemical and genetic approaches. The inter-cellular propagation of homeostasis resolution was evaluated by co-culture assays with donor monocytes trained by 4-PBA and recipient naïve monocytes. The in vivo effects of monocyte resolution and atherosclerosis prevention by 4-PBA were assessed with the high fat diet-fed ApoE -/- mouse model with i.p. 4-PBA administration. Furthermore, the selective efficacy of 4-PBA trained monocytes were examined by i.v. transfusion of ex vivo trained monocytes by 4-PBA into recipient high fat diet-fed ApoE -/- mice. Results In this study, we found that monocytes can be potently reprogrammed by 4-PBA into an immune-resolving state characterized by reduced adhesion and enhanced expression of anti-inflammatory mediator CD24. Mechanistically, 4-PBA reduced the expression of ICAM-1 via reducing peroxisome stress and attenuating SYK-mTOR signaling. Concurrently, 4-PBA enhanced the expression of resolving mediator CD24 through promoting PPARγ neddylation mediated by TOLLIP. 4-PBA trained monocytes can effectively propagate anti-inflammation activity to neighboring monocytes through CD24. Our data further demonstrated that 4-PBA trained monocytes effectively reduce atherosclerosis pathogenesis when administered in vivo . Conclusion Our study describes a robust and effective approach to generate resolving monocytes, characterizes novel mechanisms for targeted monocyte reprogramming, and offers a precision-therapeutics for atherosclerosis based on delivering reprogrammed resolving monocytes.
Collapse
|
35
|
Hicks J, Olson M, Mitchell C, Juran CM, Paul AM. The Impact of Microgravity on Immunological States. Immunohorizons 2023; 7:670-682. [PMID: 37855736 PMCID: PMC10615652 DOI: 10.4049/immunohorizons.2200063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/13/2023] [Indexed: 10/20/2023] Open
Abstract
As we explore other planetary bodies, astronauts will face unique environmental and physiological challenges. The human immune system has evolved under Earth's gravitational force. Consequently, in the microgravity environment of space, immune function is altered. This can pose problematic consequences for astronauts on deep space missions where medical intervention will be limited. Studying the unique environment of microgravity has its challenges, yet current research has uncovered immunological states that are probable during exploration missions. As microgravity-induced immune states are uncovered, novel countermeasure developments and personalized mitigation programs can be designed to improve astronaut health. This can also benefit immune-related monitoring programs for disorders on Earth. This is a comprehensive review, including gaps in knowledge, of simulated and spaceflight microgravity studies in human and rodent models.
Collapse
Affiliation(s)
- Janelle Hicks
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL
| | - Makaila Olson
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL
| | - Carol Mitchell
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL
| | - Cassandra M. Juran
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA
- Blue Marble Space Institute of Science, Seattle, WA
| | - Amber M. Paul
- Department of Human Factors and Behavioral Neurobiology, Embry-Riddle Aeronautical University, Daytona Beach, FL
- NASA Ames Research Center, Space Biosciences Division, Moffett Field, CA
- Blue Marble Space Institute of Science, Seattle, WA
| |
Collapse
|
36
|
Lin R, Wang J, Wu Y, Yi Z, Zhang Y, Li L. Resolving neutrophils due to TRAM deletion renders protection against experimental sepsis. Inflamm Res 2023; 72:1733-1744. [PMID: 37563334 PMCID: PMC10727485 DOI: 10.1007/s00011-023-01779-z] [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: 06/02/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE Proper inflammation resolution is crucial to prevent runaway inflammation during sepsis and reduce sepsis-related mortality/morbidity. Previous studies suggest that deleting TRAM, a key TLR4 signaling adaptor, can reprogram the first inflammatory responder cell-neutrophil from an inflammatory state to a resolving state. In this study, we aim to examine the therapeutic potential of TRAM-deficient neutrophils in vivo with recipient mice undergoing experimental sepsis. MATERIAL AND METHODS Wild-type or Tram-/- mice were intraperitoneally injected with cecal slurry to induce either severe or mild sepsis. Phenotypic examinations of sepsis and neutrophil characteristics were examined in vivo and ex vivo. The propagations of resolution from donor neutrophils to recipient cells such as monocytes, T cells, and endothelial cells were examined through co-culture assays in vitro. The efficacies of Tram-/- neutrophils in reducing inflammation were studied by transfusing either wild-type or Tram-/- neutrophils into septic recipient mice. RESULTS Tram-/- septic mice had improved survival and attenuated injuries within the lung and kidney tissues as compared to wild-type septic mice. Wild-type septic mice transfused with Tram-/- resolving neutrophils exhibited reduced multi-organ damages and improved cellular homeostasis. In vitro co-culture studies revealed that donor Tram-/- neutrophils can effectively propagate cellular homeostasis to co-cultured neighboring monocytes, neutrophils, T cells as well as endothelial cells. CONCLUSIONS Neutrophils with TRAM deletion render effective reprogramming into a resolving state beneficial for ameliorating experimental sepsis, with therapeutic potential in propagating cellular and tissue homeostasis as well as treating sepsis.
Collapse
Affiliation(s)
- RuiCi Lin
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA
| | - Jing Wang
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA
| | - Yajun Wu
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA
| | - Ziyue Yi
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA.
| |
Collapse
|
37
|
Yi Z, Geng S, Li L. Comparative analyses of monocyte memory dynamics from mice to humans. Inflamm Res 2023; 72:1539-1549. [PMID: 37453943 PMCID: PMC10499745 DOI: 10.1007/s00011-023-01762-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/13/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Innate monocytes can adopt dynamic "memory" states ranging from low-grade inflammation to pathogenic exhaustion, dependent upon signal strength and history of challenges. Low-grade inflammatory monocytes facilitate the pathogenesis of chronic inflammatory diseases, while exhausted monocytes drive the pathogenesis of severe sepsis. Although clinical and basic studies suggest the conservation of key features of exhausted monocytes from human and murine sepsis, systems analyses of monocyte exhaustion among human and murine monocytes are lacking. METHODS We performed cross examination of septic monocytes scRNAseq data recently collected from human sepsis patients as well as experimental septic mice, in reference to monocytes experimentally exhausted in vitro. Furthermore, we performed pseudo-time analyses of in vitro programmed monocytes following prolonged challenges causing either low-grade inflammation or exhaustion. Additional comparative analyses of low-grade inflammatory monocytes were performed with scRNAseq data from selected human patients with chronic low-grade inflammatory diseases. RESULTS Our systems analyses reveal key features of monocyte exhaustion including reduced differentiation, pathogenic inflammation and immune suppression that are highly conserved in human and murine septic monocytes, and captured by in vitro experimental exhaustion. Pseudo-time analyses reveal that monocytes initially transition into a less-differentiated state with proliferative potential. The expansion of proliferative monocytes can be observed not only in experimentally challenged monocytes, but also in tissues of murine sepsis and human septic blood. We observed that monocytes similarly transition into the less-differentiated state when challenged with a subclinical dose endotoxin under chronic inflammatory conditions. Instead of being exhausted, monocytes with prolonged challenges with super-low dose endotoxin bifurcate into the low-grade inflammatory immune-enhancing or the chemotactic/adhesive state, often see in atherosclerosis or auto-immune diseases. CONCLUSIONS Key features of monocyte memory dynamics are identified and conserved in human and murine monocytes, which can be captured by prolonged challenges of innate signals with varying signal strength.
Collapse
Affiliation(s)
- Ziyue Yi
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA
| | - Shuo Geng
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, 149 Life Science 1 Bldg, Blacksburg, VA, 24061-0910, USA.
| |
Collapse
|
38
|
Cabana-Puig X, Lu R, Geng S, Michaelis JS, Oakes V, Armstrong C, Testerman JC, Liao X, Alajoleen R, Appiah M, Zhang Y, Reilly CM, Li L, Luo XM. CX 3CR1 modulates SLE-associated glomerulonephritis and cardiovascular disease in MRL/lpr mice. Inflamm Res 2023; 72:1083-1097. [PMID: 37060359 PMCID: PMC10748465 DOI: 10.1007/s00011-023-01731-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023] Open
Abstract
OBJECTIVE Patients with systemic lupus erythematosus (SLE) often develop multi-organ damages including heart and kidney complications. We sought to better define the underlying mechanisms with a focus on the chemokine receptor CX3CR1. METHODS We generated Cx3cr1-deficient MRL/lpr lupus-prone mice through backcrossing. We then employed heterozygous intercross to generate MRL/lpr littermates that were either sufficient or deficient of CX3CR1. The mice were also treated with either Lactobacillus spp. or a high-fat diet (HFD) followed by assessments of the kidney and heart, respectively. RESULTS Cx3cr1-/- MRL/lpr mice exhibited a distinct phenotype of exacerbated glomerulonephritis compared to Cx3cr1+/+ littermates, which was associated with a decrease of spleen tolerogenic marginal zone macrophages and an increase of double-negative T cells. Interestingly, upon correction of the gut microbiota with Lactobacillus administration, the phenotype of exacerbated glomerulonephritis was reversed, suggesting that CX3CR1 controls glomerulonephritis in MRL/lpr mice through a gut microbiota-dependent mechanism. Upon treatment with HFD, Cx3cr1-/- MRL/lpr mice developed significantly more atherosclerotic plaques that were promoted by Ly6C+ monocytes. Activated monocytes expressed ICOS-L that interacted with ICOS-expressing follicular T-helper cells, which in turn facilitated a germinal center reaction to produce more autoantibodies. Through a positive feedback mechanism, the increased circulatory autoantibodies further promoted the activation of Ly6C+ monocytes and their display of ICOS-L. CONCLUSIONS We uncovered novel, Cx3cr1 deficiency-mediated pathogenic mechanisms contributing to SLE-associated glomerulonephritis and cardiovascular disease.
Collapse
Affiliation(s)
- Xavier Cabana-Puig
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Ran Lu
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Jacquelyn S Michaelis
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, USA
| | - Vanessa Oakes
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Caitlin Armstrong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - James C Testerman
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Xiaofeng Liao
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Razan Alajoleen
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Michael Appiah
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | | | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
| | - Xin M Luo
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
39
|
Oates JR, Sawada K, Giles DA, Alarcon PC, Damen MS, Szabo S, Stankiewicz TE, Moreno-Fernandez ME, Divanovic S. Thermoneutral housing shapes hepatic inflammation and damage in mouse models of non-alcoholic fatty liver disease. Front Immunol 2023; 14:1095132. [PMID: 36875069 PMCID: PMC9982161 DOI: 10.3389/fimmu.2023.1095132] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/27/2023] [Indexed: 02/19/2023] Open
Abstract
Introduction Inflammation is a common unifying factor in experimental models of non-alcoholic fatty liver disease (NAFLD) progression. Recent evidence suggests that housing temperature-driven alterations in hepatic inflammation correlate with exacerbated hepatic steatosis, development of hepatic fibrosis, and hepatocellular damage in a model of high fat diet-driven NAFLD. However, the congruency of these findings across other, frequently employed, experimental mouse models of NAFLD has not been studied. Methods Here, we examine the impact of housing temperature on steatosis, hepatocellular damage, hepatic inflammation, and fibrosis in NASH diet, methionine and choline deficient diet, and western diet + carbon tetrachloride experimental models of NAFLD in C57BL/6 mice. Results We show that differences relevant to NAFLD pathology uncovered by thermoneutral housing include: (i) augmented NASH diet-driven hepatic immune cell accrual, exacerbated serum alanine transaminase levels and increased liver tissue damage as determined by NAFLD activity score; (ii) augmented methionine choline deficient diet-driven hepatic immune cell accrual and increased liver tissue damage as indicated by amplified hepatocellular ballooning, lobular inflammation, fibrosis and overall NAFLD activity score; and (iii) dampened western diet + carbon tetrachloride driven hepatic immune cell accrual and serum alanine aminotransferase levels but similar NAFLD activity score. Discussion Collectively, our findings demonstrate that thermoneutral housing has broad but divergent effects on hepatic immune cell inflammation and hepatocellular damage across existing experimental NAFLD models in mice. These insights may serve as a foundation for future mechanistic interrogations focused on immune cell function in shaping NAFLD progression.
Collapse
Affiliation(s)
- Jarren R. Oates
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Daniel A. Giles
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Pablo C. Alarcon
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michelle S.M.A. Damen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Sara Szabo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Pathology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Traci E. Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Maria E. Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
- Immunology Graduate Program, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
40
|
Masyutina AM, Maximchik PV, Chkadua GZ, Pashenkov MV. Inhibition of specific signaling pathways rather than epigenetic silencing of effector genes is the leading mechanism of innate tolerance. Front Immunol 2023; 14:1006002. [PMID: 36776861 PMCID: PMC9909295 DOI: 10.3389/fimmu.2023.1006002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction Macrophages activated through a pattern-recognition receptor (PRR) enter a transient state of tolerance characterized by diminished responsiveness to restimulation of the same receptor. Signaling-based and epigenetic mechanisms are invoked to explain this innate tolerance. However, these two groups of mechanisms should result in different outcomes. The epigenetic scenario (silencing of effector genes) predicts that activation of a PRR should broadly cross-tolerize to agonists of unrelated PRRs, whereas in the signaling-based scenario (inhibition of signaling pathways downstream of specific PRRs), cross-tolerization should occur only between agonists utilizing the same PRR and/or signaling pathway. Also, the so-called non-tolerizeable genes have been described, which acquire distinct epigenetic marks and increased responsiveness to rechallenge with the same agonist. The existence of such genes is well explained by epigenetic mechanisms but difficult to explain solely by signaling mechanisms. Methods To evaluate contribution of signaling and epigenetic mechanisms to innate tolerance, we tolerized human macrophages with agonists of TLR4 or NOD1 receptors, which signal via distinct pathways, and assessed responses of tolerized cells to homologous restimulation and to cross-stimulation using different signaling, metabolic and transcriptomic read-outs. We developed a transcriptomics-based approach to distinguish responses to secondary stimulation from continuing responses to primary stimulation. Results We found that macrophages tolerized with a NOD1 agonist lack responses to homologous restimulation, whereas LPS-tolerized macrophages partially retain the ability to activate NF-κB pathway upon LPS rechallenge, which allows to sustain low-level expression of a subset of pro-inflammatory genes. Contributing to LPS tolerance is blockade of signaling pathways required for IFN-β production, resulting in 'pseudo-tolerization' of IFN-regulated genes. Many genes in NOD1- or TLR4-tolerized macrophages are upregulated as the result of primary stimulation (due to continuing transcription and/or high mRNA stability), but do not respond to homologous restimulation. Hyperresponsiveness of genes to homologous rechallenge is a rare and inconsistent phenomenon. However, most genes that have become unresponsive to homologous stimuli show unchanged or elevated responses to agonists of PRRs signaling via distinct pathways. Discussion Thus, inhibition of specific signaling pathways rather than epigenetic silencing is the dominant mechanism of innate tolerance.
Collapse
Affiliation(s)
- Anna M. Masyutina
- Laboratory of Clinical Immunology, National Research Center “Institute of Immunology” of the Federal Medical-Biological Agency of Russia, Moscow, Russia,Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Polina V. Maximchik
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Georgy Z. Chkadua
- Laboratory of experimental diagnostics and biotherapy of tumors, N.N.Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Mikhail V. Pashenkov
- Laboratory of Clinical Immunology, National Research Center “Institute of Immunology” of the Federal Medical-Biological Agency of Russia, Moscow, Russia,*Correspondence: Mikhail V. Pashenkov,
| |
Collapse
|
41
|
Rincon JC, Efron PA, Moldawer LL. Immunopathology of chronic critical illness in sepsis survivors: Role of abnormal myelopoiesis. J Leukoc Biol 2022; 112:1525-1534. [PMID: 36193662 PMCID: PMC9701155 DOI: 10.1002/jlb.4mr0922-690rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 02/01/2023] Open
Abstract
Sepsis remains the single most common cause of mortality and morbidity in hospitalized patients requiring intensive care. Although earlier detection and improved treatment bundles have reduced in-hospital mortality, long-term recovery remains dismal. Sepsis survivors who experience chronic critical illness often demonstrate persistent inflammation, immune suppression, lean tissue wasting, and physical and functional cognitive declines, which often last in excess of 1 year. Older patients and those with preexisting comorbidities may never fully recover and have increased mortality compared with individuals who restore their immunologic homeostasis. Many of these responses are shared with individuals with advanced cancer, active autoimmune diseases, chronic obstructive pulmonary disease, and chronic renal disease. Here, we propose that this resulting immunologic endotype is secondary to a persistent maladaptive reprioritization of myelopoiesis and pathologic activation of myeloid cells. Driven in part by the continuing release of endogenous alarmins from chronic organ injury and muscle wasting, as well as by secondary opportunistic infections, ongoing myelopoiesis at the expense of lymphopoiesis and erythropoiesis leads to anemia, recurring infections, and lean tissue wasting. Early recognition and intervention are required to interrupt this pathologic activation of myeloid populations.
Collapse
Affiliation(s)
- Jaimar C Rincon
- Sepsis and Critical Illness Research Center, Laboratory of Inflammation Biology and Surgical Science, Department of Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Philip A Efron
- Sepsis and Critical Illness Research Center, Laboratory of Inflammation Biology and Surgical Science, Department of Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Lyle L Moldawer
- Sepsis and Critical Illness Research Center, Laboratory of Inflammation Biology and Surgical Science, Department of Surgery, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
42
|
Awasthi NP, Mishra S, Tiwari V, Agarwal J, Das PK, Jain P, Husain N. Monocyte HLADR and Immune Dysregulation Index as Biomarkers for COVID-19 Severity and Mortality. Indian J Clin Biochem 2022; 38:204-211. [PMID: 36246016 PMCID: PMC9540126 DOI: 10.1007/s12291-022-01087-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 08/20/2022] [Indexed: 01/08/2023]
Abstract
Immune dysregulation in COVID-19 is the major causal factor associated with disease progression and mortality. Role of monocyte HLA-DR (mHLA-DR), neutrophil CD64 (nCD64) and Immune dysregulation index (IDI) were studied in COVID-19 patients for assessing severity and outcome. Results were compared with other laboratory parameters. Antibody bound per cell for mHLA-DR, nCD64 and IDI were measured in 100 COVID-19 patients by flow cytometry within 12 h of hospital admission. Thirty healthy controls (HC) were included. Clinical and laboratory parameters like C - reactive protein (CRP), Procalcitonin (PCT), Absolute Lymphocyte count (ALC), Absolute Neutrophil count (ANC) and Neutrophil to Lymphocyte ratio (NLR) were recorded. Patients were followed up until recovery with discharge or death. Parameters from 54 mild (MCOV-19), 46 severe (SCOV-19) and 30 HC were analysed. mHLA-DR revealed significant and graded down regulation in MCOV-19 and SCOV-19 as compared to HC whereas IDI was lowest in HC with increasing values in MCOV-19 and SCOV-19. For diagnostic discrimination of MCOV-19 and SCOV-19, IDI revealed highest AUC (0.99). All three immune parameters revealed significant difference between survivors (n = 78) and non-survivors (n = 22). mHLA-DR < 7010 and IDI > 12 had significant association with mortality. Four best performing parameters to identify patients with SCOV-19 at higher risk of mortality were IDI, NLR, ALC and PCT. mHLA-DR and IDI, in addition to NLR and ALC at admission and during hospital stay can be utilized for patient triaging, monitoring, early intervention, and mortality prediction. IDI reported for the first time in this study, appears most promising. Immune monitoring of ‘in hospital’ cases may provide optimized treatment options.
Collapse
Affiliation(s)
- Namrata Punit Awasthi
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Gomti Nagar, 226010 Lucknow, Uttar Pradesh India
| | - Sridhar Mishra
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Gomti Nagar, 226010 Lucknow, Uttar Pradesh India
| | - Vandana Tiwari
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Gomti Nagar, 226010 Lucknow, Uttar Pradesh India
| | - Jyotsna Agarwal
- Department of Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Gomti Nagar, 226010 Lucknow, Uttar Pradesh India
| | - Pravin Kumar Das
- Department of Anaesthesiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Gomti Nagar, 226010 Lucknow, Uttar Pradesh India
| | - Paresh Jain
- BD Biosciences, Central and South Asia and Japan, Tokyo, Japan
| | - Nuzhat Husain
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Gomti Nagar, 226010 Lucknow, Uttar Pradesh India
| |
Collapse
|
43
|
Lu X, Zhang K, Jiang W, Li H, Huang Y, Du M, Wan J, Cao Y, Du L, Liu X, Pan W. Single-cell RNA sequencing combined with whole exome sequencing reveals the landscape of the immune pathogenic response to chronic mucocutaneous candidiasis with STAT1 GOF mutation. Front Immunol 2022; 13:988766. [PMID: 36225936 PMCID: PMC9549386 DOI: 10.3389/fimmu.2022.988766] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic mucocutaneous candidiasis (CMC) is characterized by recurrent or persistent infections with Candida of the skin, nails, and mucous membranes (e.g., mouth, esophagus, and vagina). Compared with that of other infectious diseases, the immune pathogenic mechanism of CMC is still poorly understood. We identified a signal transducer and activator of transcription 1 gain-of-function (c.Y289C) mutation in a CMC patient. Single-cell transcriptional profiling on peripheral blood mononuclear cells from this patient revealed decreases in immature B cells and monocytes. Further analysis revealed several differentially expressed genes related to immune regulation, including RGS1, TNFAIP3, S100A8/A9, and CTSS. In our review of the literature on signal transducer and activator of transcription 1 gain-of-function (c.Y289C) mutations, we identified seven cases in total. The median age of onset for CMC (n=4, data lacking for three cases) was 10.5 years (range: birth to 11 years), with an average onset age of 8 years. There were no reports linking tumors to the c.Y289C mutation, and the incidence of pre-existing clinical disease in patients with the c.Y289C mutation was similar to previous data.
Collapse
Affiliation(s)
- Xiaodi Lu
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Keming Zhang
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Weiwei Jiang
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
- Department of Dermatology, 72nd Group Army Hospital of People’s Liberation Army (PLA), Huzhou, China
| | - Hang Li
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Yue Huang
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Mingwei Du
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
| | - Jian Wan
- Department of Dermatology, Pudong New Area People’s Hospital, Shanghai, China
| | - Yanyun Cao
- Department of Dermatology, Pudong New Area People’s Hospital, Shanghai, China
- Shanghai Key Laboratory of Pathogenic Fungi Medical Testing, Pudong New Area People’s Hospital, Shanghai, China
| | - Lin Du
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
- *Correspondence: Lin Du, ; Xiaogang Liu, ; Weihua Pan,
| | - Xiaogang Liu
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
- *Correspondence: Lin Du, ; Xiaogang Liu, ; Weihua Pan,
| | - Weihua Pan
- Shanghai Key Laboratory of Molecular Medicine Mycology, Naval Medical University, Shanghai, China
- *Correspondence: Lin Du, ; Xiaogang Liu, ; Weihua Pan,
| |
Collapse
|
44
|
Lupu L, Horst K, Greven J, Mert Ü, Ludviksen JA, Pettersen K, Lau C, Li Y, Palmer A, Qin K, Zhang X, Mayer B, van Griensven M, Huber-Lang M, Hildebrand F, Mollnes TE. Simultaneous C5 and CD14 inhibition limits inflammation and organ dysfunction in pig polytrauma. Front Immunol 2022; 13:952267. [PMID: 36059503 PMCID: PMC9433645 DOI: 10.3389/fimmu.2022.952267] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Dysfunctional complement activation and Toll-like receptor signaling immediately after trauma are associated with development of trauma-induced coagulopathy and multiple organ dysfunction syndrome. We assessed the efficacy of the combined inhibition therapy of complement factor C5 and the TLR co-receptor CD14 on thrombo-inflammation and organ damage in an exploratory 72-h polytrauma porcine model, conducted under standard surgical and intensive care management procedures. Twelve male pigs were subjected to polytrauma, followed by resuscitation (ATLS® guidelines) and operation of the femur fracture (intramedullary nailing technique). The pigs were allocated to combined C5 and CD14 inhibition therapy group (n=4) and control group (n=8). The therapy group received intravenously C5 inhibitor (RA101295) and anti-CD14 antibody (rMil2) 30 min post-trauma. Controls received saline. Combined C5 and CD14 inhibition reduced the blood levels of the terminal complement complex (TCC) by 70% (p=0.004), CRP by 28% (p=0.004), and IL-6 by 52% (p=0.048). The inhibition therapy prevented the platelet consumption by 18% and TAT formation by 77% (p=0.008). Moreover, the norepinephrine requirements in the treated group were reduced by 88%. The inhibition therapy limited the organ damage, thereby reducing the blood lipase values by 50% (p=0.028), LDH by 30% (p=0.004), AST by 33%, and NGAL by 30%. Immunofluorescent analysis of the lung tissue revealed C5b-9 deposition on blood vessels in five from the untreated, and in none of the treated animals. In kidney and liver, the C5b-9 deposition was similarly detected mainly the untreated as compared to the treated animals. Combined C5 and CD14 inhibition limited the inflammatory response, the organ damage, and reduced the catecholamine requirements after experimental polytrauma and might be a promising therapeutic approach.
Collapse
Affiliation(s)
- Ludmila Lupu
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Klemens Horst
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Johannes Greven
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Ümit Mert
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | | | | | - Corinna Lau
- Research Laboratory, Nordland Hospital Bodø, Bodø, Norway
| | - Yang Li
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Kang Qin
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Xing Zhang
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Martijn van Griensven
- Department Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Frank Hildebrand
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Tom Eirik Mollnes
- Research Laboratory, Nordland Hospital Bodø, Bodø, Norway
- Department of Immunology, Oslo University Hospital, and University of Oslo, Oslo, Norway
- Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
- *Correspondence: Tom Eirik Mollnes,
| |
Collapse
|
45
|
Decreased leukocyte exhaustion is associated with decreased IFN-β and increased α-defensin-1 levels in type-2 diabetes. Cytokine 2022; 156:155918. [DOI: 10.1016/j.cyto.2022.155918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/15/2022] [Accepted: 05/16/2022] [Indexed: 11/21/2022]
|
46
|
Giral H, Franke V, Moobed M, Müller MF, Lübking L, James DM, Hartung J, Kuschnerus K, Meteva D, Seppelt C, Jakob P, Klingenberg R, Kränkel N, Leistner D, Zeller T, Blankenberg S, Zimmermann F, Haghikia A, Lüscher TF, Akalin A, Landmesser U, Kratzer A. Rapid Inflammasome Activation Is Attenuated in Post-Myocardial Infarction Monocytes. Front Immunol 2022; 13:857455. [PMID: 35558073 PMCID: PMC9090500 DOI: 10.3389/fimmu.2022.857455] [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: 01/19/2022] [Accepted: 03/28/2022] [Indexed: 12/02/2022] Open
Abstract
Inflammasomes are crucial gatekeepers of the immune response, but their maladaptive activation associates with inflammatory pathologies. Besides canonical activation, monocytes can trigger non-transcriptional or rapid inflammasome activation that has not been well defined in the context of acute myocardial infarction (AMI). Rapid transcription-independent inflammasome activation induced by simultaneous TLR priming and triggering stimulus was measured by caspase-1 (CASP1) activity and interleukin release. Both classical and intermediate monocytes from healthy donors exhibited robust CASP1 activation, but only classical monocytes produced high mature interleukin-18 (IL18) release. We also recruited a limited number of coronary artery disease (CAD, n=31) and AMI (n=29) patients to evaluate their inflammasome function and expression profiles. Surprisingly, monocyte subpopulations isolated from blood collected during percutaneous coronary intervention (PCI) from AMI patients presented diminished CASP1 activity and abrogated IL18 release despite increased NLRP3 gene expression. This unexpected attenuated rapid inflammasome activation was accompanied by a significant increase of TNFAIP3 and IRAKM expression. Moreover, TNFAIP3 protein levels of circulating monocytes showed positive correlation with high sensitive troponin T (hsTnT), implying an association between TNFAIP3 upregulation and the severity of tissue injury. We suggest this monocyte attenuation to be a protective phenotype aftermath following a very early inflammatory wave in the ischemic area. Damage-associated molecular patterns (DAMPs) or other signals trigger a transitory negative feedback loop within newly recruited circulating monocytes as a mechanism to reduce post-injury tissue damage.
Collapse
Affiliation(s)
- Hector Giral
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Vedran Franke
- Max Delbrück Center, The Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Minoo Moobed
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Maja F Müller
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Laura Lübking
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Divya Maria James
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes Hartung
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kira Kuschnerus
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Denitsa Meteva
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Claudio Seppelt
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Philipp Jakob
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, University Hospital Zurich, Zurich, Switzerland.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Klingenberg
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Nicolle Kränkel
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - David Leistner
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tanja Zeller
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg, Lübeck, Kiel, Hamburg, Germany
| | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Hamburg, Lübeck, Kiel, Hamburg, Germany
| | - Friederike Zimmermann
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Arash Haghikia
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Altuna Akalin
- Max Delbrück Center, The Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Adelheid Kratzer
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
47
|
Laudanski K. Quo Vadis Anesthesiologist? The Value Proposition of Future Anesthesiologists Lies in Preserving or Restoring Presurgical Health after Surgical Insult. J Clin Med 2022; 11:1135. [PMID: 35207406 PMCID: PMC8879076 DOI: 10.3390/jcm11041135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/18/2022] [Indexed: 12/26/2022] Open
Abstract
This Special Issue of the Journal of Clinical Medicine is devoted to anesthesia and perioperative care [...].
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA 19104, USA; ; Tel.: +1-215-662-8000
- Leonard Davis Institute for Healthcare Economics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|