1
|
Iqbal M, Zaman M, Ojha N, Gau YTA, Young EI. The known and unknown of post-pump chorea: a case report on robust steroid responsiveness implicating occult neuroinflammation. Front Immunol 2024; 15:1458022. [PMID: 39318628 PMCID: PMC11419990 DOI: 10.3389/fimmu.2024.1458022] [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: 07/01/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Post-pump chorea (PPC) is characterized by the development of choreiform movements following cardiopulmonary bypass (CPB) surgery. PPC occurs almost exclusively in children, and its pathophysiology remains unclear. Here we present an adult case of PPC after bovine aortic valve replacement (AVR) which exhibited dramatic and reproducible response to steroid, suggesting the presence of occult neuroinflammation. This observation suggests a novel underlying mechanism in certain subgroups of PPC, which is likely a heterogeneous condition to start with. Further research into the pathomechanisms of PPC could offer insights into managing this otherwise symptomatic control-only condition.
Collapse
Affiliation(s)
- Muhammad Iqbal
- Department of Neurology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Muizz Zaman
- Department of Neurology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Niranjan Ojha
- Department of Neurology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Yung-Tian A Gau
- Department of Neurology, SUNY Upstate Medical University, Syracuse, NY, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Eufrosina I Young
- Department of Neurology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
2
|
Puzio M, Moreton N, Sullivan M, Scaife C, Glennon JC, O'Connor JJ. An Electrophysiological and Proteomic Analysis of the Effects of the Superoxide Dismutase Mimetic, MnTMPyP, on Synaptic Signalling Post-Ischemia in Isolated Rat Hippocampal Slices. Antioxidants (Basel) 2023; 12:antiox12040792. [PMID: 37107167 PMCID: PMC10135248 DOI: 10.3390/antiox12040792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic stress and the increased production of reactive oxygen species (ROS) are two main contributors to neuronal damage and synaptic plasticity in acute ischemic stroke. The superoxide scavenger MnTMPyP has been previously reported to have a neuroprotective effect in organotypic hippocampal slices and to modulate synaptic transmission after in vitro hypoxia and oxygen-glucose deprivation (OGD). However, the mechanisms involved in the effect of this scavenger remain elusive. In this study, two concentrations of MnTMPyP were evaluated on synaptic transmission during ischemia and post-ischemic synaptic potentiation. The complex molecular changes supporting cellular adaptation to metabolic stress, and how these are modulated by MnTMPyP, were also investigated. Electrophysiological data showed that MnTMPyP causes a decrease in baseline synaptic transmission and impairment of synaptic potentiation. Proteomic analysis performed on MnTMPyP and hypoxia-treated tissue indicated an impairment in vesicular trafficking mechanisms, including reduced expression of Hsp90 and actin signalling. Alterations of vesicular trafficking may lead to reduced probability of neurotransmitter release and AMPA receptor activity, resulting in the observed modulatory effect of MnTMPyP. In OGD, protein enrichment analysis highlighted impairments in cell proliferation and differentiation, such as TGFβ1 and CDKN1B signalling, in addition to downregulation of mitochondrial dysfunction and an increased expression of CAMKII. Taken together, our results may indicate modulation of neuronal sensitivity to the ischemic insult, and a complex role for MnTMPyP in synaptic transmission and plasticity, potentially providing molecular insights into the mechanisms mediating the effects of MnTMPyP during ischemia.
Collapse
Affiliation(s)
- Martina Puzio
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Niamh Moreton
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Mairéad Sullivan
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
- UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Caitriona Scaife
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Jeffrey C Glennon
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
- UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - John J O'Connor
- UCD School of Biomolecular & Biomedical Science, University College Dublin, Dublin 4, Ireland
- Mass Spectrometry Core Facility, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
3
|
Morrison T, Watts ER, Sadiku P, Walmsley SR. The emerging role for metabolism in fueling neutrophilic inflammation. Immunol Rev 2023; 314:427-441. [PMID: 36326284 PMCID: PMC10953397 DOI: 10.1111/imr.13157] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neutrophils are a critical element of host defense and are rapidly recruited to inflammatory sites. Such sites are frequently limited in oxygen and/or nutrient availability, presenting a metabolic challenge for infiltrating cells. Long believed to be uniquely dependent on glycolysis, it is now clear that neutrophils possess far greater metabolic plasticity than previously thought, with the capacity to generate energy stores and utilize extracellular proteins to fuel central carbon metabolism and biosynthetic activity. Out-with cellular energetics, metabolic programs have also been implicated in the production of neutrophils and their progenitors in the bone marrow compartment, activation of neutrophil antimicrobial responses, inflammatory and cell survival signaling cascades, and training of the innate immune response. Thus, understanding the mechanisms by which metabolic processes sustain changes in neutrophil effector functions and how these are subverted in disease states provides exciting new avenues for the treatment of dysfunctional neutrophilic inflammation which are lacking in clinical practice to date.
Collapse
Affiliation(s)
- Tyler Morrison
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of EdinburghEdinburghUK
| | - Emily R. Watts
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of EdinburghEdinburghUK
| | - Pranvera Sadiku
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of EdinburghEdinburghUK
| | - Sarah R. Walmsley
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of EdinburghEdinburghUK
| |
Collapse
|
4
|
Schlegel C, Liu K, Spring B, Dietz S, Poets CF, Hudalla H, Lajqi T, Köstlin-Gille N, Gille C. Decreased expression of hypoxia-inducible factor 1α (HIF-1α) in cord blood monocytes under anoxia. Pediatr Res 2023; 93:870-877. [PMID: 35906309 PMCID: PMC10033401 DOI: 10.1038/s41390-022-02193-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/24/2022] [Accepted: 06/28/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Infections are a major cause for morbidity and mortality in neonates; however, the underling mechanisms for increased infection susceptibility are incompletely understood. Hypoxia, which is present in inflamed tissues, has been identified as an important activation signal for innate immune cells in adults and is mainly mediated by hypoxia-inducible factor 1α (HIF-1α). Fetal tissue pO2 physiologically is low but rises immediately after birth. METHODS In this study, the effect of low oxygen partial pressure (pO2) on HIF-1α expression and its targets phagocytosis, reactive oxygen species (ROS) production and vascular endothelial growth factor (VEGF) secretion was compared in vitro between immune cells from adult peripheral blood and cord blood using anoxia, HIF-1α stabilizer desferroxamin (DFO) and E. coli as stimuli. RESULTS We show that anoxia-induced HIF-1α protein accumulation, phagocytosis, ROS-production and VEGF-expression were greatly diminished in cord blood compared to adult cells. E. coli led to HIF-1α gene expression in adult and cord blood immune cells; however, cord blood cells failed to accumulate HIF-1α protein and VEGF upon E. coli stimulation. CONCLUSIONS Taken together, our results show a diminished activation of cord blood immune cells by low pO2, which might contribute to impaired reactivity in the context of infection. IMPACT Neonatal immune cells do not accumulate HIF-1α under low oxygen partial pressure leading to decreased phagocytosis and decreased ROS production. We demonstrate a previously unknown mechanism of reduced activation of neonatal immune cells in the context of an inflammatory response. This could contribute to the increased susceptibility of newborns and preterm infants to infection.
Collapse
Affiliation(s)
- Christiane Schlegel
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Kai Liu
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Bärbel Spring
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Christian F Poets
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany
| | - Hannes Hudalla
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Trim Lajqi
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tübingen University Children's Hospital, Tübingen, Germany.
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany.
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Heidelberg, Germany
| |
Collapse
|
5
|
Wang YT, Trzeciak AJ, Rojas WS, Saavedra P, Chen YT, Chirayil R, Etchegaray JI, Lucas CD, Puleston DJ, Keshari KR, Perry JSA. Metabolic adaptation supports enhanced macrophage efferocytosis in limited-oxygen environments. Cell Metab 2023; 35:316-331.e6. [PMID: 36584675 PMCID: PMC9908853 DOI: 10.1016/j.cmet.2022.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/23/2022] [Accepted: 12/06/2022] [Indexed: 12/30/2022]
Abstract
Apoptotic cell (AC) clearance (efferocytosis) is performed by phagocytes, such as macrophages, that inhabit harsh physiological environments. Here, we find that macrophages display enhanced efferocytosis under prolonged (chronic) physiological hypoxia, characterized by increased internalization and accelerated degradation of ACs. Transcriptional and translational analyses revealed that chronic physiological hypoxia induces two distinct but complimentary states. The first, "primed" state, consists of concomitant transcription and translation of metabolic programs in AC-naive macrophages that persist during efferocytosis. The second, "poised" state, consists of transcription, but not translation, of phagocyte function programs in AC-naive macrophages that are translated during efferocytosis. Mechanistically, macrophages efficiently flux glucose into a noncanonical pentose phosphate pathway (PPP) loop to enhance NADPH production. PPP-derived NADPH directly supports enhanced efferocytosis under physiological hypoxia by ensuring phagolysosomal maturation and redox homeostasis. Thus, macrophages residing under physiological hypoxia adopt states that support cell fitness and ensure performance of essential homeostatic functions rapidly and safely.
Collapse
Affiliation(s)
- Ya-Ting Wang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alissa J Trzeciak
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Waleska Saitz Rojas
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedro Saavedra
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yan-Ting Chen
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rachel Chirayil
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jon Iker Etchegaray
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Christopher D Lucas
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, Scotland, UK; Institute for Regeneration and Repair, Edinburgh BioQuarter, Edinburgh, Scotland, UK
| | - Daniel J Puleston
- Bloomberg, Kimmel Institute of Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Kayvan R Keshari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin S A Perry
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
6
|
Tang YY, Wang DC, Wang YQ, Huang AF, Xu WD. Emerging role of hypoxia-inducible factor-1α in inflammatory autoimmune diseases: A comprehensive review. Front Immunol 2023; 13:1073971. [PMID: 36761171 PMCID: PMC9905447 DOI: 10.3389/fimmu.2022.1073971] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/13/2022] [Indexed: 01/26/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a primary metabolic sensor, and is expressed in different immune cells, such as macrophage, dendritic cell, neutrophil, T cell, and non-immune cells, for instance, synovial fibroblast, and islet β cell. HIF-1α signaling regulates cellular metabolism, triggering the release of inflammatory cytokines and inflammatory cells proliferation. It is known that microenvironment hypoxia, vascular proliferation, and impaired immunological balance are present in autoimmune diseases. To date, HIF-1α is recognized to be overexpressed in several inflammatory autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis, and function of HIF-1α is dysregulated in these diseases. In this review, we narrate the signaling pathway of HIF-1α and the possible immunopathological roles of HIF-1α in autoimmune diseases. The collected information will provide a theoretical basis for the familiarization and development of new clinical trials and treatment based on HIF-1α and inflammatory autoimmune disorders in the future.
Collapse
Affiliation(s)
- Yang-Yang Tang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Da-Cheng Wang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - You-Qiang Wang
- Department of Laboratory Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China,*Correspondence: Wang-Dong Xu,
| |
Collapse
|
7
|
Winning S, Fandrey J. Oxygen Sensing in Innate Immune Cells: How Inflammation Broadens Classical Hypoxia-Inducible Factor Regulation in Myeloid Cells. Antioxid Redox Signal 2022; 37:956-971. [PMID: 35088604 DOI: 10.1089/ars.2022.0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Significance: Oxygen deprivation (hypoxia) is a common feature at sites of inflammation. Immune cells and all other cells present at the inflamed site have to adapt to these conditions. They do so by stabilization and activation of hypoxia-inducible factor subunit α (HIF-1α and HIF-2α, respectively), enabling constant generation of adenosine triphosphate (ATP) under these austere conditions by the induction of, for example, glycolytic pathways. Recent Advances: During recent years, it has become evident that HIFs play a far more important role than initially believed because they shape the inflammatory phenotype of immune cells. They are indispensable for migration, phagocytosis, and the induction of inflammatory cytokines by innate immune cells and thereby enable a crosstalk between innate and adaptive immunity. In short, they ensure the survival and function of immune cells under critical conditions. Critical Issues: Up to now, there are still open questions regarding the individual roles of HIF-1 and HIF-2 for the different cell types. In particular, the loss of both HIF-1 and HIF-2 in myeloid cells led to unexpected and contradictory results in the mouse models analyzed so far. Similarly, the role of HIF-1 in dendritic cell maturation is unclear due to inconsistent results from in vitro experiments. Future Directions: The HIFs are indispensable for immune cell survival and action under inflammatory conditions, but they might also trigger over-activation of immune cells. Therefore, they might be excellent setscrews to adjust the inflammatory response by pharmaceuticals. China and Japan and very recently (August 2021) Europe have approved prolyl hydroxylase inhibitors (PHIs) to stabilize HIF such as roxadustat for clinical use to treat anemia by increasing the production of erythropoietin, the classical HIF target gene. Nonetheless, we need further work regarding the use of PHIs under inflammatory conditions, because HIFs show specific activation and distinct expression patterns in innate immune cells. The extent to which HIF-1 or HIF-2 as a transcription factor regulates the adaptation of immune cells to inflammatory hypoxia differs not only by the cell type but also with the inflammatory challenge and the surrounding tissue. Therefore, we urgently need isoform- and cell type-specific modulators of the HIF pathway. Antioxid. Redox Signal. 37, 956-971.
Collapse
Affiliation(s)
- Sandra Winning
- Institut für Physiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Joachim Fandrey
- Institut für Physiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| |
Collapse
|
8
|
The Possible Importance of Glutamine Supplementation to Mood and Cognition in Hypoxia from High Altitude. Nutrients 2020; 12:nu12123627. [PMID: 33255790 PMCID: PMC7760805 DOI: 10.3390/nu12123627] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 11/16/2022] Open
Abstract
Hypoxia induced by low O2 pressure is responsible for several physiological and behavioral alterations. Changes in physiological systems are frequent, including inflammation and psychobiological declines such as mood and cognition worsening, resulting in increased reaction time, difficulty solving problems, reduced memory and concentration. The paper discusses the possible relationship between glutamine supplementation and worsening cognition mediated by inflammation induced by high altitude hypoxia. The paper is a narrative literature review conducted to verify the effects of glutamine supplementation on psychobiological aspects. We searched MEDLINE/PubMed and Web of Science databases and gray literature by Google Scholar for English articles. Mechanistic pathways mediated by glutamine suggest potential positive effects of its supplementation on mood and cognition, mainly its potential effect on inflammation. However, clinical studies are scarce, making any conclusions impossible. Although glutamine plays an important role and seems to mitigate inflammation, clinical studies should test this hypothesis, which will contribute to a better mood and cognition state for several people who suffer from problems mediated by hypoxia.
Collapse
|
9
|
Kling L, Schreiber A, Eckardt KU, Kettritz R. Hypoxia-inducible factors not only regulate but also are myeloid-cell treatment targets. J Leukoc Biol 2020; 110:61-75. [PMID: 33070368 DOI: 10.1002/jlb.4ri0820-535r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
Hypoxia describes limited oxygen availability at the cellular level. Myeloid cells are exposed to hypoxia at various bodily sites and even contribute to hypoxia by consuming large amounts of oxygen during respiratory burst. Hypoxia-inducible factors (HIFs) are ubiquitously expressed heterodimeric transcription factors, composed of an oxygen-dependent α and a constitutive β subunit. The stability of HIF-1α and HIF-2α is regulated by oxygen-sensing prolyl-hydroxylases (PHD). HIF-1α and HIF-2α modify the innate immune response and are context dependent. We provide a historic perspective of HIF discovery, discuss the molecular components of the HIF pathway, and how HIF-dependent mechanisms modify myeloid cell functions. HIFs enable myeloid-cell adaptation to hypoxia by up-regulating anaerobic glycolysis. In addition to effects on metabolism, HIFs control chemotaxis, phagocytosis, degranulation, oxidative burst, and apoptosis. HIF-1α enables efficient infection defense by myeloid cells. HIF-2α delays inflammation resolution and decreases antitumor effects by promoting tumor-associated myeloid-cell hibernation. PHDs not only control HIF degradation, but also regulate the crosstalk between innate and adaptive immune cells thereby suppressing autoimmunity. HIF-modifying pharmacologic compounds are entering clinical practice. Current indications include renal anemia and certain cancers. Beneficial and adverse effects on myeloid cells should be considered and could possibly lead to drug repurposing for inflammatory disorders.
Collapse
Affiliation(s)
- Lovis Kling
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Adrian Schreiber
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ralph Kettritz
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
10
|
Behera J, Nagarajan S, Saran U, Kumar R, Keshri GK, Suryakumar G, Chatterjee S. Nitric oxide restores peripheral blood mononuclear cell adhesion against hypoxia via NO-cGMP signalling. Cell Biochem Funct 2020; 38:319-329. [PMID: 31989682 DOI: 10.1002/cbf.3502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/01/2019] [Accepted: 12/12/2019] [Indexed: 11/10/2022]
Abstract
Hypoxia is the most detrimental threat to humans residing at high altitudes, affecting multifaceted cellular responses that are crucial for normal homeostasis. Inhalation of nitric oxide has been successfully implemented to combat the hypoxia effect in the high altitude patients. We hypothesize that nitric oxide (NO) restores the peripheral blood mononuclear cell-matrix deadhesion during hypoxia. In the present study, we investigate the cellular action of exogenous NO in the hypoxia-mediated diminution of cell-matrix adhesion of PBMNC and NO bioavailability in vitro. The result showed that NO level and cell-matrix adhesion of PBMNC were significantly reduced in hypoxia as compared with normoxia, as assessed by the DAF-FM and cell adhesion assay, respectively. In contrast, cellular oxidative damage response was indeed upregulated in hypoxic PBMNC. Further, gene expression analysis revealed that mRNA transcripts of cell adhesion molecules (Integrin α5 and β1) and eNOS expressions were significantly downregulated. The mechanistic study revealed that administration of NO and 8-Br-cGMP and overexpression of eNOS-GFP restored the basal NO level and recovers cell-matrix adhesion in PBMNC via cGMP-dependent protein kinase I (PKG I) signalling. In conclusion, NO-cGMP/PKG signalling may constitute a novel target to recover high altitude-afflicted cellular deadhesion. SIGNIFICANCE OF THIS STUDY: Cellular adhesion is a complex multistep process. The ability of cells to adhere to extracellular matrix is an essential physiological process for normal homeostasis and function. Hypoxia exposure in the PBMNC culture has been proposed to induce oxidative damage and cellular deadhesion and is generally believed to be the key factor in the reduction of NO bioavailability. In the present study, we demonstrated that NO donor or overexpression of eNOS-GFP has a protective effect against hypoxia-induced cellular deadhesion and greatly improves the redox balance by inhibiting the oxidative stress. Furthermore, this protective effect of NO is mediated by the NO-cGMP/PKG signal pathway, which may provide a potential strategy against hypoxia.
Collapse
Affiliation(s)
- Jyotirmaya Behera
- Vascular Biology Lab, AU-KBC Research Centre, MIT Campus of Anna University, Chennai, India
| | - Shunmugam Nagarajan
- Vascular Biology Lab, AU-KBC Research Centre, MIT Campus of Anna University, Chennai, India
| | - Uttara Saran
- Department of Biotechnology, Anna University, Chennai, India
| | - Ravi Kumar
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Gaurav K Keshri
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | | | - Suvro Chatterjee
- Vascular Biology Lab, AU-KBC Research Centre, MIT Campus of Anna University, Chennai, India.,Department of Biotechnology, Anna University, Chennai, India
| |
Collapse
|
11
|
Caris AV, Tavares-Silva E, Thomatieli-Santos RV. Effects of carbohydrate and glutamine supplementation on cytokine production by monocytes after exercise in hypoxia: A crossover, randomized, double-blind pilot study. Nutrition 2019; 70:110592. [PMID: 31743809 DOI: 10.1016/j.nut.2019.110592] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/24/2019] [Accepted: 08/25/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the combined effects of carbohydrate (CHO) and glutamine (Gln) supplementation on cytokine production by monocytes after exercise until exhaustion performed in hypoxia. METHODS Fifteen physically active men underwent three exercises until exhaustion with an intensity of 70% maximal oxygen intake at a simulated height of 4500 m under the following supplementation: placebo, CHO (maltodextrin 8%/200 mL for 20 min), and CHO + Gln (Gln 20 g/d for 6 d and maltodextrin 8%/200 mL for 20 min) during exercise and for 2 h of recovery. Analysis of variance for repeated measures followed by the Tukey's post hoc test was realized and P < 0.05 was considered statistically significant. RESULTS Oxygen saturation of arterial blood (SaO2%) decreased in the three trials compared with baseline. Two hours post-exercise, the SaO2% was high in CHO + Gln condition compared with placebo. Two hours after exercise, interleukin (IL)-1β decreased compared with post-exercise in placebo and was lower compared with baseline in the CHO + Gln condition. Tumor necrosis factor-α decreased 2 h after exercise compared with baseline and pre-exercise in the CHO + Gln condition. No changes were observed in myeloperoxidase or IL-6 production. Two hours after exercise, Gln decreased compared with baseline and post-exercise in placebo and decreased 2 h after exercise in relation to post-exercise in the CHO condition. Gln increased post-exercise compared with pre-exercise in the CHO + Gln condition. Although erythropoietin did not change in this condition, it was high post-exercise and 2 h after exercise in the placebo condition compared with baseline and 2 h after exercise compared with baseline and pre-exercise in the CHO condition. CONCLUSIONS Gln supplementation for 6 d before exercise, associated with CHO supplementation during exercise, was able to revert Gln reduction after exercise and after 2 h of recovery and may have contributed to reducing tumor necrosis factor-α production, suggesting a possible anti-inflammatory effect of supplementation.
Collapse
Affiliation(s)
- Aline V Caris
- Universidade Federal de São Paulo, Campus São Paulo, Brazil
| | | | - Ronaldo V Thomatieli-Santos
- Universidade Federal de São Paulo, Campus Baixada Santista, Brazil; Postgraduate program in Psychobiology, Universidade Federal de São Paulo, Campus São Paulo, Brazil.
| |
Collapse
|
12
|
Hajdamowicz NH, Hull RC, Foster SJ, Condliffe AM. The Impact of Hypoxia on the Host-Pathogen Interaction between Neutrophils and Staphylococcus aureus. Int J Mol Sci 2019; 20:ijms20225561. [PMID: 31703398 PMCID: PMC6888323 DOI: 10.3390/ijms20225561] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 10/26/2019] [Accepted: 10/31/2019] [Indexed: 12/20/2022] Open
Abstract
Neutrophils are key to host defence, and impaired neutrophil function predisposes to infection with an array of pathogens, with Staphylococcus aureus a common and sometimes life-threatening problem in this setting. Both infiltrating immune cells and replicating bacteria consume oxygen, contributing to the profound tissue hypoxia that characterises sites of infection. Hypoxia in turn has a dramatic effect on both neutrophil bactericidal function and the properties of S. aureus, including the production of virulence factors. Hypoxia thereby shapes the host-pathogen interaction and the progression of infection, for example promoting intracellular bacterial persistence, enabling local tissue destruction with the formation of an encaging abscess capsule, and facilitating the establishment and propagation of bacterial biofilms which block the access of host immune cells. Elucidating the molecular mechanisms underlying host-pathogen interactions in the setting of hypoxia will enable better understanding of persistent and recalcitrant infections due to S. aureus and may uncover novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Natalia H Hajdamowicz
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Beech Hill Road, Sheffield S10 2TN, UK; (N.H.H.); (R.C.H.)
- Florey Institute, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
| | - Rebecca C Hull
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Beech Hill Road, Sheffield S10 2TN, UK; (N.H.H.); (R.C.H.)
- Florey Institute, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
| | - Simon J Foster
- Florey Institute, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Beech Hill Road, Sheffield S10 2TN, UK; (N.H.H.); (R.C.H.)
- Florey Institute, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK;
- Correspondence:
| |
Collapse
|
13
|
Santos SA, Lira FS, Silva ET, Caris AV, Oyama LM, Thomatieli-Santos RV. Effect of moderate exercise under hypoxia on Th1/Th2 cytokine balance. CLINICAL RESPIRATORY JOURNAL 2019; 13:583-589. [PMID: 31310707 DOI: 10.1111/crj.13061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/09/2018] [Accepted: 07/08/2019] [Indexed: 11/30/2022]
Abstract
INTRODUCTION AND OBJECTIVE Moderate exercise performed in normoxia can be immunostimulatory, while strenuous exercise can be immunosuppressive. However, less is known about the effects of exercise under hypoxia on cytokines. The aim of this study was to evaluate the effects of an acute exercise session performed under hypoxia similar to an altitude of 4200 m on cytokine balance. Our hypothesis was that exercise, even of moderate intensity, associated with hypoxia may induce different changes in relation to the normoxic condition. METHODS Eight healthy male volunteers were exercised on a treadmill for 1 hour at an intensity of 50% VO2peak under normoxic or hypoxic condition (4200 m). Blood samples were collected at rest and immediately 1 hour after the exercise, respectively to determine cytokines, hormones and metabolites. The two-way ANOVA and the Bonferroni post hoc test were used and the significance adopted was P < .05. RESULTS While IL-2, the IL-2/IL-4 ratio and glutamine decreased under hypoxia, IL-6 and IL-1ra increased. There were increases in the IL-2/IL-4 ratio, IL-6, IL-1ra and IL-10/TNF-α in normoxia. There were no differences in cortisol or glucose. CONCLUSION Moderate exercise under hypoxia condition changes the Th1/Th2 balance including IL-2, IL-4 and TNF-α cytokines, suggesting a Th2 response after 1 hour rest.
Collapse
Affiliation(s)
- Samile A Santos
- Department of Bioscience, Universidade Federal de São Paulo, Santos, Brazil
| | - Fábio S Lira
- Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Edgar T Silva
- Department of Bioscience, Universidade Federal de São Paulo, Santos, Brazil
| | - Aline V Caris
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lila M Oyama
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
14
|
Caris AV, Santos RVT. Performance and altitude: Ways that nutrition can help. Nutrition 2018; 60:35-40. [PMID: 30529882 DOI: 10.1016/j.nut.2018.09.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/29/2018] [Indexed: 12/18/2022]
Abstract
High altitudes are a challenge for human physiology and for sports enthusiasts. Several reasons lead to deterioration in performance at high altitudes. Hypoxia owing to high altitude causes a breakdown of homeostasis with imbalance in several physiological systems, including the immune system. The reduction in mucosal immunity and inflammation and the predominance of the humoral immune response causes a condition of immunosuppression and an increased likelihood of infection. In addition, it is known that worsening of the immune response is associated with reduced performance. On the other hand, immunonutrition plays an important role in modulating the effects of physical exercise on the immune system. However, to our knowledge, few studies have evaluated the effect of nutrition on the immune system after exercise in hypoxia. Although the association between exercise and hypoxia has been shown to be more severe for the body owing to the sum of stressful agents, supplementation with carbohydrates and glutamine seems to play a relevant role in mitigating immunosuppressive effects. These findings, although limited by the fact that they are the result of very few studies, shed light on a relevant theme for sports physiology and nutrition and suggest that both supplements may be useful for athletes, visitors, and workers in high-altitude regions. The aim of this review was to discuss the effects of high-altitude hypoxia on the human body from the point of view of exercise immunology because it is known that transient immunosuppression after strenuous exercise and competition should be followed by reduction in training overload and worse performance.
Collapse
Affiliation(s)
- Aline V Caris
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
15
|
Volatile Anesthetic Attenuates Phagocyte Function and Worsens Bacterial Loads in Wounds. J Surg Res 2018; 233:323-330. [PMID: 30502266 DOI: 10.1016/j.jss.2018.07.075] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/19/2018] [Accepted: 07/23/2018] [Indexed: 01/21/2023]
Abstract
BACKGROUND Previously we have shown that volatile anesthetic isoflurane attenuated neutrophil recruitment and phagocytosis in mouse sepsis and skin inflammation models. The objectives of this study were to test ex vivo function of neutrophils in patients who underwent cardiac catheterization under volatile anesthesia versus intravenous anesthesia (IA), and also to assess the effect of anesthesia on surgical site infections (SSIs) using mouse model to understand the clinical relevance of anesthesia-induced immunomodulation. METHODS Whole blood from patients who underwent cardiac catheterization procedures either by volatile anesthesia or IA was collected and subjected to phagocytosis assay and a lipopolysaccharide-induced tumor necrosis factor-α assay. Mouse SSI with Staphylococcus aureus USA300 was created, and the effect of isoflurane and propofol exposure (short or long exposure) on bacterial loads was tested. RESULTS Neutrophil phagocytosis was significantly attenuated after the induction of volatile anesthesia in patients, but not by IA. Monocyte phagocytosis was not affected by the anesthesia regimen. Bacterial loads following SSIs were significantly higher in mice receiving long, but not short, isoflurane exposure. Propofol exposure did not affect bacterial loads. DISCUSSION Neutrophil phagocytosis can be affected by the type of anesthesia, and preclinical model of SSIs showed potential clinical relevance. The effects of anesthesia regimen on SSIs in patients needs to be studied extensively in the future.
Collapse
|
16
|
Wernly B, Zappe AK, Unbehaun A, Sinning JM, Jung C, Kim WK, Fichtlscherer S, Lichtenauer M, Hoppe UC, Alushi B, Beckhoff F, Wewetzer C, Franz M, Kretzschmar D, Navarese E, Landmesser U, Falk V, Lauten A. Transcatheter valve-in-valve implantation (VinV-TAVR) for failed surgical aortic bioprosthetic valves. Clin Res Cardiol 2018; 108:83-92. [DOI: 10.1007/s00392-018-1326-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022]
|
17
|
Caris AV, Ysis W, Lemos VDA, Bottura R, Santos RVTD. Nutrition and exercise can attenuate inflammatory and psychobiological changes in hypoxia? Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2016.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
18
|
Lodge KM, Thompson AAR, Chilvers ER, Condliffe AM. Hypoxic regulation of neutrophil function and consequences for Staphylococcus aureus infection. Microbes Infect 2016; 19:166-176. [PMID: 27789256 DOI: 10.1016/j.micinf.2016.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/07/2016] [Accepted: 10/12/2016] [Indexed: 01/22/2023]
Abstract
Staphylococcal infection and neutrophilic inflammation can act in concert to establish a profoundly hypoxic environment. In this review we summarise how neutrophils and Staphylococcus aureus are adapted to function under hypoxic conditions, with a particular focus on the impaired ability of hypoxic neutrophils to effect Staphylococcus aureus killing.
Collapse
Affiliation(s)
- Katharine M Lodge
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge University Hospitals, Hills Road, Cambridge, CB2 0QQ, UK
| | - A A Roger Thompson
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge University Hospitals, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| |
Collapse
|
19
|
Murinello S, Moreno SK, Macauley MS, Sakimoto S, Westenskow PD, Friedlander M. Assessing Retinal Microglial Phagocytic Function In Vivo Using a Flow Cytometry-based Assay. J Vis Exp 2016. [PMID: 27805590 DOI: 10.3791/54677] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Microglia are the tissue resident macrophages of the central nervous system (CNS) and they perform a variety of functions that support CNS homeostasis, including phagocytosis of damaged synapses or cells, debris, and/or invading pathogens. Impaired phagocytic function has been implicated in the pathogenesis of diseases such as Alzheimer's and age-related macular degeneration, where amyloid-β plaque and drusen accumulate, respectively. Despite its importance, microglial phagocytosis has been challenging to assess in vivo. Here, we describe a simple, yet robust, technique for precisely monitoring and quantifying the in vivo phagocytic potential of retinal microglia. Previous methods have relied on immunohistochemical staining and imaging techniques. Our method uses flow cytometry to measure microglial uptake of fluorescently labeled particles after intravitreal delivery to the eye in live rodents. This method replaces conventional practices that involve laborious tissue sectioning, immunostaining, and imaging, allowing for more precise quantification of microglia phagocytic function in just under six hours. This procedure can also be adapted to test how various compounds alter microglial phagocytosis in physiological settings. While this technique was developed in the eye, its use is not limited to vision research.
Collapse
Affiliation(s)
- Salome Murinello
- Department of Cell and Molecular Biology, The Scripps Research Institute;
| | - Stacey K Moreno
- Department of Cell and Molecular Biology, The Scripps Research Institute
| | | | - Susumu Sakimoto
- Department of Cell and Molecular Biology, The Scripps Research Institute
| | - Peter D Westenskow
- Department of Cell and Molecular Biology, The Scripps Research Institute; The Lowy Medical Research Institute
| | - Martin Friedlander
- Department of Cell and Molecular Biology, The Scripps Research Institute
| |
Collapse
|
20
|
Kiers HD, Scheffer GJ, van der Hoeven JG, Eltzschig HK, Pickkers P, Kox M. Immunologic Consequences of Hypoxia during Critical Illness. Anesthesiology 2016; 125:237-49. [PMID: 27183167 PMCID: PMC5119461 DOI: 10.1097/aln.0000000000001163] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hypoxia and immunity are highly intertwined at clinical, cellular, and molecular levels. The prevention of tissue hypoxia and modulation of systemic inflammation are cornerstones of daily practice in the intensive care unit. Potentially, immunologic effects of hypoxia may contribute to outcome and represent possible therapeutic targets. Hypoxia and activation of downstream signaling pathways result in enhanced innate immune responses, aimed to augment pathogen clearance. On the other hand, hypoxia also exerts antiinflammatory and tissue-protective effects in lymphocytes and other tissues. Although human data on the net immunologic effects of hypoxia and pharmacologic modulation of downstream pathways are limited, preclinical data support the concept of tailoring the immune response through modulation of the oxygen status or pharmacologic modulation of hypoxia-signaling pathways in critically ill patients.
Collapse
Affiliation(s)
- Harmke D. Kiers
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Gert-Jan Scheffer
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes G. van der Hoeven
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Holger K. Eltzschig
- Organ Protection Program; Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| |
Collapse
|
21
|
Yilmaz A, Ratka J, Rohm I, Pistulli R, Goebel B, Asadi Y, Petri A, Kiehntopf M, Figulla HR, Jung C. Decrease in circulating plasmacytoid dendritic cells during short-term systemic normobaric hypoxia. Eur J Clin Invest 2016; 46:115-22. [PMID: 25652640 DOI: 10.1111/eci.12416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/31/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND During exposure to high altitude, the immune system is altered. During hypoxia, an increase in interleukin (IL)-6 and high sensitivity C-reactive protein (hs-CRP), and an increase in natural killer cells and decrease in T cells in blood was shown. However, the impact of hypoxia on dendritic cells has not been investigated yet. MATERIAL AND METHODS Twelve healthy volunteers were subjected to a transient normobaric hypoxia for 6·5 h simulating an oxygen concentration at 5500 m. During exposure to hypoxia, blood samples were collected and analysed by flow cytometrical cell sorting (FACS) for circulating myeloid (mDCs) and plasmacytoid (pDCs) DCs. Serum levels of IL-6 and tumour necrosis factor (TNF)-α were analysed. In a cell culture hypoxia chamber, blood samples were subjected to the same hypoxia and analysed regarding DCs. RESULTS Exposure to normobaric hypoxia induced a significant decrease in circulating pDCs about 45% (P = 0·001) but not of mDC compared to baseline normoxia. Furthermore, we observed a significant increase of TNF-α about 340% (P = 0·03) and of IL-6 about 286% (P = 0·002). In cell culture experiments exposure of blood to hypoxia led to no significant changes in DCs, so that a direct cytotoxic effect was excluded. During hypoxia, we observed a transient increase in stromal-derived factor 1 (SDF-1) which is important for pDC tissue recruitment. CONCLUSIONS We show a significant decrease in circulating pDCs during hypoxia in parallel to a pro-inflammatory response. Further studies are necessary to evaluate whether the decrease in circulating pDCs might be the result of an enhanced tissue recruitment.
Collapse
Affiliation(s)
- Atilla Yilmaz
- Internal Medicine II, Elisabeth Hospital Schmalkalden, Schmalkalden, Germany
| | - Josi Ratka
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Ilonka Rohm
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Rudin Pistulli
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Bjorn Goebel
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Yahya Asadi
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Alexander Petri
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Michael Kiehntopf
- Department of Clinical Chemistry & Laboratory Medicine, University Hospital of Jena, Jena, Germany
| | - Hans R Figulla
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Christian Jung
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| |
Collapse
|
22
|
Lichtenauer M, Goebel B, Fritzenwanger M, Förster M, Betge S, Lauten A, Figulla HR, Jung C. Simulated temporary hypoxia triggers the release of CD31+/Annexin+ endothelial microparticles: A prospective pilot study in humans. Clin Hemorheol Microcirc 2015; 61:83-90. [DOI: 10.3233/ch-141908] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - Bjoern Goebel
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Michael Fritzenwanger
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Martin Förster
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Stefan Betge
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Alexander Lauten
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Hans-Reiner Figulla
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Christian Jung
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| |
Collapse
|
23
|
Mishra KP, Ganju L, Singh SB. Hypoxia modulates innate immune factors: A review. Int Immunopharmacol 2015; 28:425-8. [PMID: 26184693 DOI: 10.1016/j.intimp.2015.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/19/2015] [Accepted: 07/07/2015] [Indexed: 12/30/2022]
Abstract
Hypoxia is an important factor for transcriptional regulation of cell metabolism and the adaptation to cellular stress. It modulates the function of phagocytic cells by stimulating surface receptors such as scavenger receptors, toll like receptors and their downstream signaling cascades. In response to hypoxia, innate immune modifiers are upregulated through pathways involving the key immune response master regulator nuclear factor-κB leading to the modulation of inflammatory cytokines. In this review, we highlighted the effects of hypoxia on different innate immune factors and consequences thereof.
Collapse
Affiliation(s)
- K P Mishra
- Immunomodulation Laboratory, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi 110054, India.
| | - Lilly Ganju
- Immunomodulation Laboratory, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi 110054, India
| | - Shashi Bala Singh
- Immunomodulation Laboratory, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi 110054, India
| |
Collapse
|
24
|
Jung C, Quitter F, Lichtenauer M, Fritzenwanger M, Pfeil A, Shemyakin A, Franz M, Figulla HR, Pfeifer R, Pernow J. Increased arginase levels contribute to impaired perfusion after cardiopulmonary resuscitation. Eur J Clin Invest 2014; 44:965-71. [PMID: 25186018 DOI: 10.1111/eci.12330] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/29/2014] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The postcardiac arrest syndrome occurs after global hypoxia leading to microcirculatory impairment. Nitric oxide (NO) is a key molecule regulating microvascular function. The enzyme arginase has been suggested to modulate microvascular function by regulating NO metabolism. Therefore, we investigated whether arginase increases following global hypoxia and resuscitation and tested whether arginase inhibition influences altered microcirculation in resuscitated patients. METHODS To determine the effect of global hypoxia on circulating arginase levels, fourteen healthy subjects were exposed to hypoxia in a normobaric hypoxia chamber (FiO² = 9·9%). In addition, 31 resuscitated patients were characterized clinically, and arginase 1 was measured on days 1 and 3. In eight resuscitated patients, a microcirculatory analysis was performed using a sidestream darkfield microcirculation camera. Perfused capillary density (PCD) was recorded before and after sublingual incubation of N-omega-hydroxy-nor-l-arginine (nor-NOHA) alone or together with the NOS inhibitor NG-monomethyl-l-arginine (l-NMMA). RESULTS Circulating arginase 1 levels increased in healthy volunteers following global hypoxia in the hypoxic chamber (P < 0·01). In addition, arginase 1 levels were higher on day 1 (69·1 ± 83·3 ng/mL) and on day 3 (44·2 ± 65·6 ng/mL) after resuscitation than in control subjects (P < 0·001). Incubation of the sublingual mucosa with nor-NOHA increased microcirculatory perfusion (P < 0·001). This effect was inhibited by co-incubation with K-NMMA. CONCLUSIONS Circulating arginase 1 levels are increased following exposure to global hypoxia and in patients who have been successfully resuscitated after cardiac arrest. Topical arginase inhibition improves microcirculatory perfusion following resuscitation. This is of potential therapeutic importance for the postcardiac arrest syndrome.
Collapse
Affiliation(s)
- Christian Jung
- Department of Medicine, Karolinska Institute, Stockholm, Sweden; Clinic of Internal Medicine I, Friedrich-Schiller-University, Jena, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Keely S, Campbell EL, Baird AW, Hansbro PM, Shalwitz RA, Kotsakis A, McNamee EN, Eltzschig HK, Kominsky DJ, Colgan SP. Contribution of epithelial innate immunity to systemic protection afforded by prolyl hydroxylase inhibition in murine colitis. Mucosal Immunol 2014; 7:114-23. [PMID: 23695513 PMCID: PMC3772994 DOI: 10.1038/mi.2013.29] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/16/2013] [Indexed: 02/04/2023]
Abstract
Pharmacological stabilization of hypoxia-inducible factor (HIF) through prolyl hydroxylase (PHD) inhibition limits mucosal damage associated with models of murine colitis. However, little is known about how PHD inhibitors (PHDi) influence systemic immune function during mucosal inflammation or the relative importance of immunological changes to mucosal protection. We hypothesized that PHDi enhances systemic innate immune responses to colitis-associated bacteremia. Mice with colitis induced by trinitrobenzene sulfonic acid were treated with AKB-4924, a new HIF-1 isoform-predominant PHDi, and clinical, immunological, and biochemical endpoints were assessed. Administration of AKB-4924 led to significantly reduced weight loss and disease activity compared with vehicle controls. Treated groups were pyrexic but did not become subsequently hypothermic. PHDi treatment augmented epithelial barrier function and led to an approximately 50-fold reduction in serum endotoxin during colitis. AKB-4924 also decreased cytokines involved in pyrogenesis and hypothermia, significantly reducing serum levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α while increasing IL-10. Treatment offered no protection against colitis in epithelial-specific HIF-1α-deficient mice, strongly implicating epithelial HIF-1α as the tissue target for AKB-4924-mediated protection. Taken together, these results indicate that inhibition of prolyl hydroxylase with AKB-4924 enhances innate immunity and identifies that the epithelium is a central site of inflammatory protection afforded by PHDi in murine colitis.
Collapse
Affiliation(s)
- Simon Keely
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia,Hunter Medical Research Institute, John Hunter Hospital, NSW, Australia,Correspondence to: Simon Keely, Ph.D., School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia. Office phone: (02) 40420229 Fax: (02) 4042 0024
| | - Eric L. Campbell
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Alan W. Baird
- School of Veterinary Medicine, University College Dublin, Ireland
| | - Philip M. Hansbro
- School of Biomedical Sciences & Pharmacy, University of Newcastle, NSW, Australia,Hunter Medical Research Institute, John Hunter Hospital, NSW, Australia
| | | | | | - Eoin N. McNamee
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Holger K. Eltzschig
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Douglas J. Kominsky
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Sean P. Colgan
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| |
Collapse
|