1
|
Casarin F, Mascolo R, Motta I, Wu MA, Bizzi E, Pedroli A, Dieguez G, Iacomelli G, Serati L, Duca L, Maestroni S, Tombetti E, Cappellini MD, Brucato A. Decrease in Hemoglobin Levels during Acute Attacks in Patients with Idiopathic Recurrent Pericarditis: A Model of Anemia in Acute Disease. J Clin Med 2024; 13:5944. [PMID: 39408004 PMCID: PMC11478241 DOI: 10.3390/jcm13195944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/28/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Anemia during acute inflammation is not well described in the literature. We aimed to study whether patients develop a transient hemoglobin decrease during an acute attack of recurrent pericarditis. Methods: We retrospectively analyzed patients with recurrent pericarditis. The primary endpoint was the difference in hemoglobin levels during an acute attack and in the following remission. As secondary endpoints, we correlated this variation with laboratory and clinical features; we also evaluated the available baseline hemoglobin values. Results: Sixty-two patients, including thirty females (48.4%), with a median age of 39 years, were observed during an acute attack and remission. The attack indexed was the first in 21 patients and the second or the third in 41, with pre-attack hemoglobin levels available for the latter group. Median hemoglobin levels (IQR) were 13.8 (12.8-15.1) g/dL at baseline, 12.0 (11.2-13.4) during attacks and 13.6 (13.1-14.0) during remission (p < 0.001). The median hemoglobin reduction between an acute attack and remission was 1.4 g/dL. Their mean corpuscular volume remained in the normal range. Hb reduction significantly correlated with C-reactive protein (CRP) elevation, neutrophilia and the neutrophil-to-lymphocyte ratio, but not serosal involvement. Only CRP elevation remained associated with the variation of Hb in a multivariate analysis (p = 0.007). Conclusions: This study is a proof of concept: hemoglobin levels may decline rapidly during acute inflammation in correlation with CRP elevation, with transient normocytic anemia, followed by a rapid rebound. In this regard, idiopathic pericarditis may represent a pathogenetic model of this type of anemia.
Collapse
Affiliation(s)
- Francesca Casarin
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli-Sacco, University of Milan, 20157 Milan, Italy
| | - Ruggiero Mascolo
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli-Sacco, University of Milan, 20157 Milan, Italy
| | - Irene Motta
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- SC Medicina ad Indirizzo Metabolico, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.D.); (M.D.C.)
| | - Maddalena Alessandra Wu
- Division of Internal Medicine, ASST Fatebenefratelli-Sacco, Luigi Sacco Hospital, University of Milan, 20157 Milan, Italy;
| | - Emanuele Bizzi
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
| | - Alice Pedroli
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli-Sacco, University of Milan, 20157 Milan, Italy
| | - Giulia Dieguez
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli-Sacco, University of Milan, 20157 Milan, Italy
| | - Giacomo Iacomelli
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli-Sacco, University of Milan, 20157 Milan, Italy
| | - Lisa Serati
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli-Sacco, University of Milan, 20157 Milan, Italy
| | - Lorena Duca
- SC Medicina ad Indirizzo Metabolico, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.D.); (M.D.C.)
| | - Silvia Maestroni
- Department of Internal Medicine, Ospedale Papa Giovanni XXIII, 24127 Bergamo, Italy;
| | - Enrico Tombetti
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli-Sacco, University of Milan, 20157 Milan, Italy
| | - Maria Domenica Cappellini
- SC Medicina ad Indirizzo Metabolico, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.D.); (M.D.C.)
| | - Antonio Brucato
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, Fatebenefratelli Hospital, 20121 Milan, Italy; (F.C.); (E.B.); (A.P.); (G.D.); (G.I.); (L.S.); (E.T.); (A.B.)
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli-Sacco, University of Milan, 20157 Milan, Italy
| |
Collapse
|
2
|
Carolin A, Frazer D, Yan K, Bishop CR, Tang B, Nguyen W, Helman SL, Horvat J, Larcher T, Rawle DJ, Suhrbier A. The effects of iron deficient and high iron diets on SARS-CoV-2 lung infection and disease. Front Microbiol 2024; 15:1441495. [PMID: 39296289 PMCID: PMC11408339 DOI: 10.3389/fmicb.2024.1441495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/22/2024] [Indexed: 09/21/2024] Open
Abstract
Introduction The severity of Coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is often dictated by a range of comorbidities. A considerable literature suggests iron deficiency and iron overload may contribute to increased infection, inflammation and disease severity, although direct causal relationships have been difficult to establish. Methods Here we generate iron deficient and iron loaded C57BL/6 J mice by feeding standard low and high iron diets, with mice on a normal iron diet representing controls. All mice were infected with a primary SARS-CoV-2 omicron XBB isolate and lung inflammatory responses were analyzed by histology, immunohistochemistry and RNA-Seq. Results Compared with controls, iron deficient mice showed no significant changes in lung viral loads or histopathology, whereas, iron loaded mice showed slightly, but significantly, reduced lung viral loads and histopathology. Transcriptional changes were modest, but illustrated widespread dysregulation of inflammation signatures for both iron deficient vs. controls, and iron loaded vs. controls. Some of these changes could be associated with detrimental outcomes, whereas others would be viewed as beneficial. Discussion Diet-associated iron deficiency or overload thus induced modest modulations of inflammatory signatures, but no significant histopathologically detectable disease exacerbations.
Collapse
Affiliation(s)
- Agnes Carolin
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - David Frazer
- Molecular Nutrition, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kexin Yan
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Cameron R Bishop
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Bing Tang
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Wilson Nguyen
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Sheridan L Helman
- Molecular Nutrition, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jay Horvat
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, Australia
| | | | - Daniel J Rawle
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Suhrbier
- Inflammation Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- GVN Centre of Excellence, Australian Infectious Disease Research Centre, Brisbane, QLD, Australia
| |
Collapse
|
3
|
Ameri A, Pourseyedi F, Davoodian P, Safa O, Hassanipour S, Fathalipour M. Efficacy and safety of deferoxamine in moderately ill COVID-19 patients: An open label, randomized controlled trial. Medicine (Baltimore) 2024; 103:e39142. [PMID: 39183421 PMCID: PMC11346869 DOI: 10.1097/md.0000000000039142] [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: 10/17/2023] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Deferoxamine is a potent iron chelator that could remove iron from the virus, and severe acute respiratory syndrome coronavirus 2 requires iron to replication. Also, deferoxamine has antioxidant and cytokine-modulating effects. Therefore, we evaluated the efficacy and safety of deferoxamine in patients with moderate coronavirus disease 2019 (COVID-19). METHODS In this randomized controlled trial, patients with moderate COVID-19 were randomly assigned in a 1:1 ratio to the deferoxamine group (received a solution of 500 mg deferoxamine divided into 4 doses a day through a nebulizer for 7 days) and the control group. The main outcomes were viral clearance, oxygen saturation (SPO2), body temperature, and respiratory rate (RR). Intensive care unit admission, hospital length of stay, and hospital mortality were also assessed. RESULTS A total of 62 patients, with 30 in the deferoxamine group and 32 in the control group, were randomly assigned. There was no statistically significant improvement in viral clearance after the intervention ended in the deferoxamine group (36.7%) compared to the control group (34.4%). The results showed there was no significant difference between the analyzed groups in terms of SPO2, body temperature, RR, and the number of patients with a worse prognosis (SPO2 < 96%, temperature ≥ 37.5 °C, or RR ≥ 16/min) at the end of the study. There were no significant differences seen between the groups in terms of intensive care unit admission, hospital length of stay, hospital mortality, and the occurrence of adverse medication events during the follow-up period. CONCLUSION Deferoxamine had no significant impact on improving moderately ill patients with COVID-19. However, it was well-tolerated in the patients, and this intervention demonstrated a safe profile of adverse events.
Collapse
Affiliation(s)
- Ali Ameri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Farnaz Pourseyedi
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parivash Davoodian
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Omid Safa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Fathalipour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Endocrinology and Metabolic Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
4
|
Stoffel NU, Drakesmith H. Effects of Iron Status on Adaptive Immunity and Vaccine Efficacy: A Review. Adv Nutr 2024; 15:100238. [PMID: 38729263 PMCID: PMC11251406 DOI: 10.1016/j.advnut.2024.100238] [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: 01/24/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
Vaccines can prevent infectious diseases, but their efficacy varies, and factors impacting vaccine effectiveness remain unclear. Iron deficiency is the most common nutrient deficiency, affecting >2 billion individuals. It is particularly common in areas with high infectious disease burden and in groups that are routinely vaccinated, such as infants, pregnant women, and the elderly. Recent evidence suggests that iron deficiency and low serum iron (hypoferremia) not only cause anemia but also may impair adaptive immunity and vaccine efficacy. A report of human immunodeficiency caused by defective iron transport underscored the necessity of iron for adaptive immune responses and spurred research in this area. Sufficient iron is essential for optimal production of plasmablasts and IgG responses by human B-cells in vitro and in vivo. The increased metabolism of activated lymphocytes depends on the high-iron acquisition, and hypoferremia, especially when occurring during lymphocyte expansion, adversely affects multiple facets of adaptive immunity, and may lead to prolonged inhibition of T-cell memory. In mice, hypoferremia suppresses the adaptive immune response to influenza infection, resulting in more severe pulmonary disease. In African infants, anemia and/or iron deficiency at the time of vaccination predict decreased response to diphtheria, pertussis, and pneumococcal vaccines, and response to measles vaccine may be increased by iron supplementation. In this review, we examine the emerging evidence that iron deficiency may limit adaptive immunity and vaccine responses. We discuss the molecular mechanisms and evidence from animal and human studies, highlight important unknowns, and propose a framework of key research questions to better understand iron-vaccine interactions.
Collapse
Affiliation(s)
- Nicole U Stoffel
- Medical Research Council Translational Immune Discovery Unit, Medical Research Council Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom.
| | - Hal Drakesmith
- Medical Research Council Translational Immune Discovery Unit, Medical Research Council Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
5
|
Bazdar S, Bloemsma LD, Baalbaki N, Blankestijn JM, Cornelissen MEB, Beijers RJHCG, Sondermeijer BM, van Wijck Y, Downward GS, Maitland-van der Zee AH. Hemoglobin and Its Relationship with Fatigue in Long-COVID Patients Three to Six Months after SARS-CoV-2 Infection. Biomedicines 2024; 12:1234. [PMID: 38927441 PMCID: PMC11201257 DOI: 10.3390/biomedicines12061234] [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: 02/11/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Background: While some long-term effects of COVID-19 are respiratory in nature, a non-respiratory effect gaining attention has been a decline in hemoglobin, potentially mediated by inflammatory processes. In this study, we examined the correlations between hemoglobin levels and inflammatory biomarkers and evaluated the association between hemoglobin and fatigue in a cohort of Long-COVID patients. Methods: This prospective cohort study in the Netherlands evaluated 95 (mostly hospitalized) patients, aged 40-65 years, 3-6 months post SARS-CoV-2 infection, examining their venous hemoglobin concentration, anemia (hemoglobin < 7.5 mmol/L in women and <8.5 mmol/L in men), inflammatory blood biomarkers, average FSS (Fatigue Severity Score), demographics, and clinical features. Follow-up hemoglobin was compared against hemoglobin during acute infection. Spearman correlation was used for assessing the relationship between hemoglobin concentrations and inflammatory biomarkers, and the association between hemoglobin and fatigue was examined using logistic regression. Results: In total, 11 (16.4%) participants were suffering from anemia 3-6 months after SARS-CoV-2 infection. The mean hemoglobin value increased by 0.3 mmol/L 3-6 months after infection compared to the hemoglobin during the acute phase (p-value = 0.003). Whilst logistic regression showed that a 1 mmol/L greater increase in hemoglobin is related to a decrease in experiencing fatigue in Long-COVID patients (adjusted OR 0.38 [95%CI 0.13-1.09]), we observed no correlations between hemoglobin and any of the inflammatory biomarkers examined. Conclusion: Our results indicate that hemoglobin impairment might play a role in developing Long-COVID fatigue. Further investigation is necessary to identify the precise mechanism causing hemoglobin alteration in these patients.
Collapse
Affiliation(s)
- Somayeh Bazdar
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.B.); (L.D.B.); (J.M.B.); (M.E.B.C.)
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| | - Lizan D. Bloemsma
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.B.); (L.D.B.); (J.M.B.); (M.E.B.C.)
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| | - Nadia Baalbaki
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.B.); (L.D.B.); (J.M.B.); (M.E.B.C.)
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| | - Jelle M. Blankestijn
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.B.); (L.D.B.); (J.M.B.); (M.E.B.C.)
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| | - Merel E. B. Cornelissen
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.B.); (L.D.B.); (J.M.B.); (M.E.B.C.)
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| | - Rosanne J. H. C. G. Beijers
- Department of Respiratory Medicine, Nutrim Institute of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6202 AZ Maastricht, The Netherlands;
| | | | - Yolanda van Wijck
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.B.); (L.D.B.); (J.M.B.); (M.E.B.C.)
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| | - George S. Downward
- Department of Environmental Epidemiology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3584 CL Utrecht, The Netherlands;
- Department of Global Public Health & Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Anke H. Maitland-van der Zee
- Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; (S.B.); (L.D.B.); (J.M.B.); (M.E.B.C.)
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
6
|
Gietl M, Burkert F, Hofer S, Gostner JM, Sonnweber T, Tancevski I, Pizzini A, Sahanic S, Schroll A, Brigo N, Egger A, Bellmann-Weiler R, Löffler-Ragg J, Weiss G, Kurz K. Laboratory parameters related to disease severity and physical performance after reconvalescence of acute COVID-19 infection. Sci Rep 2024; 14:10388. [PMID: 38710760 DOI: 10.1038/s41598-024-57448-6] [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] [Received: 09/28/2023] [Accepted: 03/18/2024] [Indexed: 05/08/2024] Open
Abstract
Research into the molecular basis of disease trajectory and Long-COVID is important to get insights toward underlying pathophysiological processes. The objective of this study was to investigate inflammation-mediated changes of metabolism in patients with acute COVID-19 infection and throughout a one-year follow up period. The study enrolled 34 patients with moderate to severe COVID-19 infection admitted to the University Clinic of Innsbruck in early 2020. The dynamics of multiple laboratory parameters (including inflammatory markers [C-reactive protein (CRP), interleukin-6 (IL-6), neopterin] as well as amino acids [tryptophan (Trp), phenylalanine (Phe) and tyrosine (Tyr)], and parameters of iron and vitamin B metabolism) was related to disease severity and patients' physical performance. Also, symptom load during acute illness and at approximately 60 days (FU1), and one year after symptom onset (FU2) were monitored and related with changes of the investigated laboratory parameters: During acute infection many investigated laboratory parameters were elevated (e.g., inflammatory markers, ferritin, kynurenine, phenylalanine) and enhanced tryptophan catabolism and phenylalanine accumulation were found. At FU2 nearly all laboratory markers had declined back to reference ranges. However, kynurenine/tryptophan ratio (Kyn/Trp) and the phenylalanine/tyrosine ratio (Phe/Tyr) were still exceeding the 95th percentile of healthy controls in about two thirds of our cohort at FU2. Lower tryptophan concentrations were associated with B vitamin availability (during acute infection and at FU1), patients with lower vitamin B12 levels at FU1 had a prolonged and more severe impairment of their physical functioning ability. Patients who had fully recovered (ECOG 0) presented with higher concentrations of iron parameters (ferritin, hepcidin, transferrin) and amino acids (phenylalanine, tyrosine) at FU2 compared to patients with restricted ability to work. Persistent symptoms at FU2 were tendentially associated with IFN-γ related parameters. Women were affected by long-term symptoms more frequently. Conclusively, inflammation-mediated biochemical changes appear to be related to symptoms of patients with acute and Long Covid.
Collapse
Affiliation(s)
- Mario Gietl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Francesco Burkert
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Stefanie Hofer
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Johanna M Gostner
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80, 6020, Innsbruck, Austria
| | - Thomas Sonnweber
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Alex Pizzini
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Sabina Sahanic
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Natascha Brigo
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Alexander Egger
- Central Institute for Medical and Chemical Laboratory Diagnostics (ZIMCL), Tirol Kliniken GmbH, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Rosa Bellmann-Weiler
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Judith Löffler-Ragg
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Katharina Kurz
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
7
|
Naidu AS, Wang CK, Rao P, Mancini F, Clemens RA, Wirakartakusumah A, Chiu HF, Yen CH, Porretta S, Mathai I, Naidu SAG. Precision nutrition to reset virus-induced human metabolic reprogramming and dysregulation (HMRD) in long-COVID. NPJ Sci Food 2024; 8:19. [PMID: 38555403 PMCID: PMC10981760 DOI: 10.1038/s41538-024-00261-2] [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: 10/12/2023] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
SARS-CoV-2, the etiological agent of COVID-19, is devoid of any metabolic capacity; therefore, it is critical for the viral pathogen to hijack host cellular metabolic machinery for its replication and propagation. This single-stranded RNA virus with a 29.9 kb genome encodes 14 open reading frames (ORFs) and initiates a plethora of virus-host protein-protein interactions in the human body. These extensive viral protein interactions with host-specific cellular targets could trigger severe human metabolic reprogramming/dysregulation (HMRD), a rewiring of sugar-, amino acid-, lipid-, and nucleotide-metabolism(s), as well as altered or impaired bioenergetics, immune dysfunction, and redox imbalance in the body. In the infectious process, the viral pathogen hijacks two major human receptors, angiotensin-converting enzyme (ACE)-2 and/or neuropilin (NRP)-1, for initial adhesion to cell surface; then utilizes two major host proteases, TMPRSS2 and/or furin, to gain cellular entry; and finally employs an endosomal enzyme, cathepsin L (CTSL) for fusogenic release of its viral genome. The virus-induced HMRD results in 5 possible infectious outcomes: asymptomatic, mild, moderate, severe to fatal episodes; while the symptomatic acute COVID-19 condition could manifest into 3 clinical phases: (i) hypoxia and hypoxemia (Warburg effect), (ii) hyperferritinemia ('cytokine storm'), and (iii) thrombocytosis (coagulopathy). The mean incubation period for COVID-19 onset was estimated to be 5.1 days, and most cases develop symptoms after 14 days. The mean viral clearance times were 24, 30, and 39 days for acute, severe, and ICU-admitted COVID-19 patients, respectively. However, about 25-70% of virus-free COVID-19 survivors continue to sustain virus-induced HMRD and exhibit a wide range of symptoms that are persistent, exacerbated, or new 'onset' clinical incidents, collectively termed as post-acute sequelae of COVID-19 (PASC) or long COVID. PASC patients experience several debilitating clinical condition(s) with >200 different and overlapping symptoms that may last for weeks to months. Chronic PASC is a cumulative outcome of at least 10 different HMRD-related pathophysiological mechanisms involving both virus-derived virulence factors and a multitude of innate host responses. Based on HMRD and virus-free clinical impairments of different human organs/systems, PASC patients can be categorized into 4 different clusters or sub-phenotypes: sub-phenotype-1 (33.8%) with cardiac and renal manifestations; sub-phenotype-2 (32.8%) with respiratory, sleep and anxiety disorders; sub-phenotype-3 (23.4%) with skeleto-muscular and nervous disorders; and sub-phenotype-4 (10.1%) with digestive and pulmonary dysfunctions. This narrative review elucidates the effects of viral hijack on host cellular machinery during SARS-CoV-2 infection, ensuing detrimental effect(s) of virus-induced HMRD on human metabolism, consequential symptomatic clinical implications, and damage to multiple organ systems; as well as chronic pathophysiological sequelae in virus-free PASC patients. We have also provided a few evidence-based, human randomized controlled trial (RCT)-tested, precision nutrients to reset HMRD for health recovery of PASC patients.
Collapse
Affiliation(s)
- A Satyanarayan Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA.
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA.
| | - Chin-Kun Wang
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- School of Nutrition, Chung Shan Medical University, 110, Section 1, Jianguo North Road, Taichung, 40201, Taiwan
| | - Pingfan Rao
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- College of Food and Bioengineering, Fujian Polytechnic Normal University, No.1, Campus New Village, Longjiang Street, Fuqing City, Fujian, China
| | - Fabrizio Mancini
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President-Emeritus, Parker University, 2540 Walnut Hill Lane, Dallas, TX, 75229, USA
| | - Roger A Clemens
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- University of Southern California, Alfred E. Mann School of Pharmacy/D. K. Kim International Center for Regulatory & Quality Sciences, 1540 Alcazar St., CHP 140, Los Angeles, CA, 90089, USA
| | - Aman Wirakartakusumah
- International Union of Food Science and Technology (IUFoST), Guelph, ON, Canada
- IPMI International Business School Jakarta; South East Asian Food and Agriculture Science and Technology, IPB University, Bogor, Indonesia
| | - Hui-Fang Chiu
- Department of Chinese Medicine, Taichung Hospital, Ministry of Health & Well-being, Taichung, Taiwan
| | - Chi-Hua Yen
- Department of Family and Community Medicine, Chung Shan Medical University Hospital; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Sebastiano Porretta
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- President, Italian Association of Food Technology (AITA), Milan, Italy
- Experimental Station for the Food Preserving Industry, Department of Consumer Science, Viale Tanara 31/a, I-43121, Parma, Italy
| | - Issac Mathai
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- Soukya International Holistic Health Center, Whitefield, Bengaluru, India
| | - Sreus A G Naidu
- Global Nutrition Healthcare Council (GNHC) Mission-COVID, Yorba Linda, CA, USA
- N-terminus Research Laboratory, 232659 Via del Rio, Yorba Linda, CA, 92887, USA
| |
Collapse
|
8
|
Hanson AL, Mulè MP, Ruffieux H, Mescia F, Bergamaschi L, Pelly VS, Turner L, Kotagiri P, Göttgens B, Hess C, Gleadall N, Bradley JR, Nathan JA, Lyons PA, Drakesmith H, Smith KGC. Iron dysregulation and inflammatory stress erythropoiesis associates with long-term outcome of COVID-19. Nat Immunol 2024; 25:471-482. [PMID: 38429458 PMCID: PMC10907301 DOI: 10.1038/s41590-024-01754-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 01/12/2024] [Indexed: 03/03/2024]
Abstract
Persistent symptoms following SARS-CoV-2 infection are increasingly reported, although the drivers of post-acute sequelae (PASC) of COVID-19 are unclear. Here we assessed 214 individuals infected with SARS-CoV-2, with varying disease severity, for one year from COVID-19 symptom onset to determine the early correlates of PASC. A multivariate signature detected beyond two weeks of disease, encompassing unresolving inflammation, anemia, low serum iron, altered iron-homeostasis gene expression and emerging stress erythropoiesis; differentiated those who reported PASC months later, irrespective of COVID-19 severity. A whole-blood heme-metabolism signature, enriched in hospitalized patients at month 1-3 post onset, coincided with pronounced iron-deficient reticulocytosis. Lymphopenia and low numbers of dendritic cells persisted in those with PASC, and single-cell analysis reported iron maldistribution, suggesting monocyte iron loading and increased iron demand in proliferating lymphocytes. Thus, defects in iron homeostasis, dysregulated erythropoiesis and immune dysfunction due to COVID-19 possibly contribute to inefficient oxygen transport, inflammatory disequilibrium and persisting symptomatology, and may be therapeutically tractable.
Collapse
Affiliation(s)
- Aimee L Hanson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Matthew P Mulè
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NIH-Oxford-Cambridge Scholars Program, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hélène Ruffieux
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Federica Mescia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Victoria S Pelly
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Lorinda Turner
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Berthold Göttgens
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Christoph Hess
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Nicholas Gleadall
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Botnar Research Centre for Child Health (BRCCH), University of Basel and ETH Zurich, Basel, Switzerland
| | - John R Bradley
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - James A Nathan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Hal Drakesmith
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
9
|
Abstract
COVID-19 can cause detrimental effects on health. Vaccines have helped in reducing disease severity and transmission but their long-term effects on health and effectiveness against future viral variants remain unknown. COVID-19 pathogenesis involves alteration in iron homeostasis. Thus, a contextual understanding of iron-related parameters would be very valuable for disease prognosis and therapeutics.Accordingly, we reviewed the status of iron and iron-related proteins in COVID-19. Iron-associated alterations in COVID-19 reported hitherto include anemia of inflammation, low levels of serum iron (hypoferremia), transferrin and transferrin saturation, and high levels of serum ferritin (hyperferritinemia), hepcidin, lipocalin-2, catalytic iron, and soluble transferrin receptor (in ICU patients). Hemoglobin levels can be low or normal, and compromised hemoglobin function has been proposed. Membrane-bound transferrin receptor may facilitate viral entry, so it acts as a potential target for antiviral therapy. Lactoferrin can provide natural defense by preventing viral entry and/or inhibiting viral replication. Serum iron and ferritin levels can predict COVID-19-related hospitalization, severity, and mortality. Serum hepcidin and ferritin/transferrin ratio can predict COVID-19 severity. Here, serum levels of these iron-related parameters are provided, caveats of iron chelation for therapy are discussed and the interplay of these iron-related parameters in COVID-19 is explained.This synopsis is crucial as it clearly presents the iron picture of COVID-19. The information may assist in disease prognosis and/or in formulating iron-related adjunctive strategies that can help reduce infection/inflammation and better manage COVID-19 caused by future variants. Indeed, the current picture will augment as more is revealed about these iron-related parameters in COVID-19.
Collapse
Affiliation(s)
- Erin Suriawinata
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Kosha J Mehta
- Centre for Education, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
10
|
Nkansah C, Owusu M, Appiah SK, Mensah K, Bani SB, Osei-Boakye F, Agyemang LD, Ackah EB, Abbam G, Daud S, Quansah Y, Derigubah CA, Apodola FA, Ayangba V, Afrifa DA, Eshun CP, Iddrisu AW, Mintaah S, Twum B, Mohammed A, Agyare EM, Gyasi WA, Agbadza PE, Wilson CAE, Anane S, Antwi P, Antwi RAY. Effects of COVID-19 disease on PAI-1 antigen and haematological parameters during disease management: A prospective cross-sectional study in a regional Hospital in Ghana. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001866. [PMID: 37347738 DOI: 10.1371/journal.pgph.0001866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/29/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Individuals with COVID-19 experience thrombotic events probably due to the associated hypofibrinolysis resulting from the upregulation of plasminogen activator inhibitor-1 (PAI-1) antigen. This study evaluated plasma PAI-1 antigen levels and haematological parameters before treatment and after recovery from severe COVID-19 in Ghana. MATERIALS AND METHODS This cross-sectional study was conducted at Sunyani Regional Hospital, and recruited 51 patients who had RT-PCR-confirmed SARS-CoV-2. Participants' sociodemographic data and clinical characteristics were taken from the hospital records. Venous blood was taken before COVID-19 treatment commenced for FBC, PAI-1 and ferritin assays. FBC was assessed using an automated haematology analyzer, whilst plasma PAI-1 Ag and serum ferritin levels were assessed with sandwich ELISA. All the tests were repeated immediately after participants recovered from COVID-19. RESULTS Of the 51 participants recruited into the study, 78.4% (40) had non-severe COVID-19 whiles 21.6% (11) experienced a severe form of the disease. Severe COVID-19 participants had significantly lower haemoglobin (g/dL): 8.1 (7.3-8.4) vs 11.8 (11.0-12.5), p<0.001; RBC x 1012/L: 2.9 (2.6-3.1) vs 3.4 (3.1-4.3), p = 0.001; HCT%: 24.8 ± 2.6 vs 35.3 ± 6.7, p<0.001 and platelet x 109/L: 86.4 (62.2-91.8) vs 165.5 (115.1-210.3), p<0.001, compared with the non-severe COVID-19 group. But WBC x 109/L: 11.6 (9.9-14.2) vs 5.4 (3.7-6.6), p<0.001 and ferritin (ng/mL): 473.1 (428.3-496.0) vs 336.2 (249.9-386.5), p<0.001, were relatively higher in the participants with severe COVID-19 than the non-severe COVID-19 counterparts. Also, the severely ill SARS-CoV-2-infected participants had relatively higher plasma PAI-1 Ag levels (ng/mL): 131.1 (128.7-131.9) vs 101.3 (92.0-116.8), p<0.001, than those with the non-severe form of the disease. Participants had lower haemoglobin (g/dL): 11.4 (8.8-12.3 vs 12.4 (11.5-13.6), p<0.001; RBC x 1012/L: 3.3 (2.9-4.0) vs 4.3 (3.4-4.6), p = 0.001; absolute granulocyte count x 109/L: 2.3 ± 1.0 vs 4.6 ± 1.8, p<0.001, and platelet x 109/L: 135.0 (107.0-193.0) vs 229.0 (166.0-270.0), p<0.001 values at admission before treatment commenced, compared to when they recovered from the disease. Additionally, the median PAI-1 Ag (ng/mL): 89.6 (74.9-100.8) vs 103.1 (93.2-128.7), p<0.001 and ferritin (ng/mL): 242.2 (197.1-302.1) vs 362.3 (273.1-399.9), p<0.001 levels were reduced after a successful recovery from COVID-19 compared to the values at admission. CONCLUSION Plasma PAI-1 Ag level was higher among severe COVID-19 participants. The COVID-19-associated inflammation could affect red blood cell parameters and platelets. Successful recovery from COVID-19, with reduced inflammatory response as observed in the decline of serum ferritin levels restores the haematological parameters. Plasma levels of PAI-1 should be assessed during the management of severe COVID-19 in Ghana. This will enhance the early detection of probable thrombotic events and prompts Physicians to provide interventions to prevent thrombotic complications associated with COVID-19.
Collapse
Affiliation(s)
- Charles Nkansah
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Michael Owusu
- Department of Medical Laboratory Sciences, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
- Kumasi Centre for Collaborative Research, Kumasi, Ghana
| | - Samuel Kwasi Appiah
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Kofi Mensah
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Simon Bannison Bani
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Felix Osei-Boakye
- Department of Medical Laboratory Technology, Faculty of Applied Science and Technology, Sunyani Technical University, Sunyani, Ghana
| | - Lawrence Duah Agyemang
- Department of Medical Laboratory Sciences, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
- Department of Clinical Microbiology, Laboratory Service Directorate, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - Ezekiel Bonwin Ackah
- Department of Medical Laboratory Sciences, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
| | - Gabriel Abbam
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Samira Daud
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Yeduah Quansah
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Charles Angnataa Derigubah
- Department of Medical Laboratory Technology, School of Applied Science and Arts, Bolgatanga Technical University, Bolgatanga, Ghana
| | - Francis Atoroba Apodola
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Nursing and Allied Health Sciences, Nalerigu, Ghana
| | - Valentine Ayangba
- Department of Nursing and Midwifery, Faculty of Nursing and Midwifery, College of Nursing and Allied Health Sciences, Nalerigu, Ghana
| | - David Amoah Afrifa
- Department of Medical Laboratory Sciences, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
- Department of Medical Laboratory, Ankaase Methodist Hospital, Kumasi, Ghana
| | - Caleb Paul Eshun
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Abdul-Waliu Iddrisu
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Selina Mintaah
- Department of Medical Laboratory Sciences, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
- Department of Haematology, Laboratory Service Directorate, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - Benjamin Twum
- Department of Medical Laboratory, Sunyani Regional Hospital, Sunyani, Ghana
| | - Abidatu Mohammed
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Eugene Mensah Agyare
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Wendy Akomeah Gyasi
- Department of Clinical Microbiology, Laboratory Service Directorate, Komfo Anokye Teaching Hospital, Kumasi, Ghana
- Department of Theoretical and Applied Biology, Faculty of Biosciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Peace Esenam Agbadza
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Candy Adwoa Ewusiwaa Wilson
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Seth Anane
- Department of Clinical Microbiology, Laboratory Service Directorate, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - Prince Antwi
- Department of Microbiology, School of Health and Life Sciences, TEESSIDE University, Middlesbrough, United Kingdom
| | | |
Collapse
|
11
|
Eder J, Schumm L, Armann JP, Puhan MA, Beuschlein F, Kirschbaum C, Berner R, Toepfner N. Increased red blood cell deformation in children and adolescents after SARS-CoV-2 infection. Sci Rep 2023; 13:9823. [PMID: 37330522 PMCID: PMC10276822 DOI: 10.1038/s41598-023-35692-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 05/22/2023] [Indexed: 06/19/2023] Open
Abstract
Severe coronavirus disease 2019 (COVID-19) is associated with hyperinflammation, hypercoagulability and hypoxia. Red blood cells (RBCs) play a key role in microcirculation and hypoxemia and are therefore of special interest in COVID-19 pathophysiology. While this novel disease has claimed the lives of many older patients, it often goes unnoticed or with mild symptoms in children. This study aimed to investigate morphological and mechanical characteristics of RBCs after SARS-CoV-2 infection in children and adolescents by real-time deformability-cytometry (RT-DC), to investigate the relationship between alterations of RBCs and clinical course of COVID-19. Full blood of 121 students from secondary schools in Saxony, Germany, was analyzed. SARS-CoV-2-serostatus was acquired at the same time. Median RBC deformation was significantly increased in SARS-CoV-2-seropositive compared to seronegative children and adolescents, but no difference could be detected when the infection dated back more than 6 months. Median RBC area was the same in seropositive and seronegative adolescents. Our findings of increased median RBC deformation in SARS-CoV-2 seropositive children and adolescents until 6 months post COVID-19 could potentially serve as a progression parameter in the clinical course of the disease with an increased RBC deformation pointing towards a mild course of COVID-19.
Collapse
Affiliation(s)
- Julian Eder
- Biopsychology, Technische Universität Dresden, Dresden, Germany
| | - Leonie Schumm
- Department of Paediatrics, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jakob P Armann
- Department of Paediatrics, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Milo A Puhan
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Zurich, Switzerland
| | | | - Reinhard Berner
- Department of Paediatrics, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nicole Toepfner
- Department of Paediatrics, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
12
|
Tene L, Karasik A, Chodick G, Pereira DIA, Schou H, Waechter S, Göhring UM, Drakesmith H. Iron deficiency and the effectiveness of the BNT162b2 vaccine for SARS-CoV-2 infection: A retrospective, longitudinal analysis of real-world data. PLoS One 2023; 18:e0285606. [PMID: 37216375 PMCID: PMC10202294 DOI: 10.1371/journal.pone.0285606] [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] [Received: 10/23/2022] [Accepted: 04/26/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Iron plays a key role in human immune responses; however, the influence of iron deficiency on the coronavirus disease 2019 (COVID-19) vaccine effectiveness is unclear. AIM To assess the effectiveness of the BNT162b2 messenger RNA COVID-19 vaccine in preventing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and COVID-19-related hospitalization and death in individuals with or without iron deficiency. METHODS This large retrospective, longitudinal cohort study analyzed real-world data from the Maccabi Healthcare Services database (covering 25% of Israeli residents). Eligible adults (aged ≥16 years) received a first BNT162b2 vaccine dose between December 19, 2020, and February 28, 2021, followed by a second dose as per approved vaccine label. Individuals were excluded if they had SARS-CoV-2 infection before vaccination, had hemoglobinopathy, received a cancer diagnosis since January 2020, had been treated with immunosuppressants, or were pregnant at the time of vaccination. Vaccine effectiveness was assessed in terms of incidence rates of SARS-CoV-2 infection confirmed by real-time polymerase chain reaction assay, relative risks of COVID-19-related hospitalization, and mortality in individuals with iron deficiency (ferritin <30 ng/mL or transferrin saturation <20%). The two-dose protection period was Days 7 to 28 after the second vaccination. RESULTS Data from 184,171 individuals with (mean [standard deviation; SD] age 46.2 [19.6] years; 81.2% female) versus 1,072,019 without (mean [SD] age 46.9 [18.0] years; 46.2% female) known iron deficiency were analyzed. Vaccine effectiveness in the two-dose protection period was 91.9% (95% confidence interval [CI] 83.7-96.0%) and 92.1% (95% CI 84.2-96.1%) for those with versus without iron deficiency (P = 0.96). Of patients with versus without iron deficiency, hospitalizations occurred in 28 and 19 per 100,000 during the reference period (Days 1-7 after the first dose), and in 19 and 7 per 100,000 during the two-dose protection period, respectively. Mortality rates were comparable between study groups: 2.2 per 100,000 (4/181,012) in the population with iron deficiency and 1.8 per 100,000 (19/1,055,298) in those without known iron deficiency. CONCLUSIONS Results suggest that the BNT162b2 COVID-19 vaccine is >90% effective in preventing SARS-CoV-2 infection in the 3 weeks after the second vaccination, irrespective of iron-deficiency status. These findings support the use of the vaccine in populations with iron deficiency.
Collapse
Affiliation(s)
- Lilac Tene
- Maccabi Institute for Research & Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Avraham Karasik
- Maccabi Institute for Research & Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| | - Gabriel Chodick
- Maccabi Institute for Research & Innovation, Maccabi Healthcare Services, Tel Aviv, Israel
| | | | | | | | | | - Hal Drakesmith
- Medical Research Council Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Lack of Evidence on Association between Iron Deficiency and COVID-19 Vaccine-Induced Neutralizing Humoral Immunity. Vaccines (Basel) 2023; 11:vaccines11020327. [PMID: 36851205 PMCID: PMC9965425 DOI: 10.3390/vaccines11020327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Iron is a crucial micronutrient for immunity induction in response to infections and vaccinations. This study aimed to investigate the effect of iron deficiency on COVID-19-vaccine-induced humoral immunity. We investigated the effectiveness of COVID-19 vaccines (BNT162b2, mRNA-1273, and ChAdOx nCov-2019) in iron-deficient individuals (n = 63) and provide a side-by-side comparison to healthy controls (n = 67). The presence of anti-SARS-CoV-2 spike (S) and anti-nucleocapsid (NP) IgG were assessed using in-house S- and NP-based ELISA followed by serum neutralization test (SNT). High concordance between S-based ELISA and SNT results was observed. The prevalence of neutralizing antibodies was 95.24% (60/63) in the study group and 95.52% (64/67) in the controls with no significant difference. The presence/absence of past infection, period since vaccination, vaccine type, and being iron-deficient or having iron-deficiency anemia did not exert any significant effect on the prevalence or titer of anti-SARS-CoV-2 neutralizing antibodies. NP-based ELISA identified individuals unaware of exposure to SARS-CoV-2. Moreover, absence of anti-NP IgG was noted in participants who were previously diagnosed with COVID-19 suggesting the unpredictability of after-infection immunity. To sum up, this study demonstrated an initial lack of evidence on the association between iron deficiency and the effectiveness of COVID-19-vaccine-induced neutralizing humoral immunity. Similar studies with larger sample size remain necessary to obtain comprehensive conclusions about the effect or lack of effect of iron on COVID-19-vaccine effectiveness.
Collapse
|
14
|
Naidu SAG, Clemens RA, Naidu AS. SARS-CoV-2 Infection Dysregulates Host Iron (Fe)-Redox Homeostasis (Fe-R-H): Role of Fe-Redox Regulators, Ferroptosis Inhibitors, Anticoagulants, and Iron-Chelators in COVID-19 Control. J Diet Suppl 2023; 20:312-371. [PMID: 35603834 DOI: 10.1080/19390211.2022.2075072] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Severe imbalance in iron metabolism among SARS-CoV-2 infected patients is prominent in every symptomatic (mild, moderate to severe) clinical phase of COVID-19. Phase-I - Hypoxia correlates with reduced O2 transport by erythrocytes, overexpression of HIF-1α, altered mitochondrial bioenergetics with host metabolic reprogramming (HMR). Phase-II - Hyperferritinemia results from an increased iron overload, which triggers a fulminant proinflammatory response - the acute cytokine release syndrome (CRS). Elevated cytokine levels (i.e. IL6, TNFα and CRP) strongly correlates with altered ferritin/TF ratios in COVID-19 patients. Phase-III - Thromboembolism is consequential to erythrocyte dysfunction with heme release, increased prothrombin time and elevated D-dimers, cumulatively linked to severe coagulopathies with life-threatening outcomes such as ARDS, and multi-organ failure. Taken together, Fe-R-H dysregulation is implicated in every symptomatic phase of COVID-19. Fe-R-H regulators such as lactoferrin (LF), hemoxygenase-1 (HO-1), erythropoietin (EPO) and hepcidin modulators are innate bio-replenishments that sequester iron, neutralize iron-mediated free radicals, reduce oxidative stress, and improve host defense by optimizing iron metabolism. Due to its pivotal role in 'cytokine storm', ferroptosis is a potential intervention target. Ferroptosis inhibitors such as ferrostatin-1, liproxstatin-1, quercetin, and melatonin could prevent mitochondrial lipid peroxidation, up-regulate antioxidant/GSH levels and abrogate iron overload-induced apoptosis through activation of Nrf2 and HO-1 signaling pathways. Iron chelators such as heparin, deferoxamine, caffeic acid, curcumin, α-lipoic acid, and phytic acid could protect against ferroptosis and restore mitochondrial function, iron-redox potential, and rebalance Fe-R-H status. Therefore, Fe-R-H restoration is a host biomarker-driven potential combat strategy for an effective clinical and post-recovery management of COVID-19.
Collapse
Affiliation(s)
| | - Roger A Clemens
- Department of International Regulatory Science, University of Southern California School of Pharmacy, Los Angeles, CA, USA
| | | |
Collapse
|
15
|
The role of iron in chronic inflammatory diseases: from mechanisms to treatment options in anemia of inflammation. Blood 2022; 140:2011-2023. [PMID: 35994752 DOI: 10.1182/blood.2021013472] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/09/2022] [Indexed: 11/20/2022] Open
Abstract
Anemia of inflammation (AI) is a highly prevalent comorbidity in patients affected by chronic inflammatory disorders, such as chronic kidney disease, inflammatory bowel disease, or cancer, that negatively affect disease outcome and quality of life. The pathophysiology of AI is multifactorial, with inflammatory hypoferremia and iron-restricted erythropoiesis playing a major role in the context of disease-specific factors. Here, we review the recent progress in our understanding of the molecular mechanisms contributing to iron dysregulation in AI, the impact of hypoferremia and anemia on the course of the underlying disease, and (novel) therapeutic strategies applied to treat AI.
Collapse
|
16
|
Maira D, Duca L, Busti F, Consonni D, Salvatici M, Vianello A, Milani A, Guzzardella A, Di Pierro E, Aliberti S, Baldini IM, Bandera A, Blasi F, Cassinerio E, Cesari M, Fracanzani AL, Grasselli G, Graziadei G, Lombardi R, Marchi G, Montano N, Monzani V, Peyvandi F, Proietti M, Sandri M, Valenti L, Cappellini MD, Girelli D, Protti A, Motta I. The role of hypoxia and inflammation in the regulation of iron metabolism and erythropoiesis in COVID-19: The IRONCOVID study. Am J Hematol 2022; 97:1404-1412. [PMID: 36215667 PMCID: PMC9538950 DOI: 10.1002/ajh.26679] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/15/2022] [Accepted: 08/02/2022] [Indexed: 01/28/2023]
Abstract
Coronavirus Disease (COVID-19) can be considered as a human pathological model of inflammation combined with hypoxia. In this setting, both erythropoiesis and iron metabolism appear to be profoundly affected by inflammatory and hypoxic stimuli, which act in the opposite direction on hepcidin regulation. The impact of low blood oxygen levels on erythropoiesis and iron metabolism in the context of human hypoxic disease (e.g., pneumonia) has not been fully elucidated. This multicentric observational study was aimed at investigating the prevalence of anemia, the alterations of iron homeostasis, and the relationship between inflammation, hypoxia, and erythropoietic parameters in a cohort of 481 COVID-19 patients admitted both to medical wards and intensive care units (ICU). Data were collected on admission and after 7 days of hospitalization. On admission, nearly half of the patients were anemic, displaying mild-to-moderate anemia. We found that hepcidin levels were increased during the whole period of observation. The patients with a higher burden of disease (i.e., those who needed intensive care treatment or had a more severe degree of hypoxia) showed lower hepcidin levels, despite having a more marked inflammatory pattern. Erythropoietin (EPO) levels were also lower in the ICU group on admission. After 7 days, EPO levels rose in the ICU group while they remained stable in the non-ICU group, reflecting that the initial hypoxic stimulus was stronger in the first group. These findings strengthen the hypothesis that, at least in the early phases, hypoxia-driven stimuli prevail over inflammation in the regulation of hepcidin and, finally, of erythropoiesis.
Collapse
Affiliation(s)
- Diletta Maira
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoGeneral Medicine UnitMilanItaly
| | - Lorena Duca
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoGeneral Medicine UnitMilanItaly
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine, EuroBloodNet CenterUniversity of Verona and Azienda Ospedaliera Universitaria Integrata VeronaVeronaItaly
| | - Dario Consonni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoEpidemiology UnitMilanItaly
| | | | - Alice Vianello
- Department of Medicine, Section of Internal Medicine, EuroBloodNet CenterUniversity of Verona and Azienda Ospedaliera Universitaria Integrata VeronaVeronaItaly
| | - Angelo Milani
- Department of Biomedical SciencesHumanitas UniversityPieve Emanuele (Milan)Italy
- Department of Anesthesia and Intensive Care UnitsIRCCS‐Humanitas Research HospitalRozzano (Milan)Italy
| | - Amedeo Guzzardella
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of AnesthesiaIntensive Care and EmergencyMilanItaly
| | - Elena Di Pierro
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoGeneral Medicine UnitMilanItaly
| | - Stefano Aliberti
- Respiratory Unit, Humanitas Clinical and Research CenterIRCCSRozzano (Milan)Italy
| | - Itala Marina Baldini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoGeneral Medicine UnitMilanItaly
| | - Alessandra Bandera
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoInfectious Diseases UnitMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Francesco Blasi
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Respiratory UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Elena Cassinerio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoGeneral Medicine UnitMilanItaly
| | - Matteo Cesari
- Geriatric UnitIRCCS Istituti Clinici Scientifici MaugeriMilanItaly
- Department of Clinical Sciences and Community HealthUniversity of MilanMilanItaly
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoInternal Medicine and Metabolic Disease UnitMilanItaly
| | - Giacomo Grasselli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of AnesthesiaIntensive Care and EmergencyMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Giovanna Graziadei
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoGeneral Medicine UnitMilanItaly
| | - Rosa Lombardi
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoInternal Medicine and Metabolic Disease UnitMilanItaly
| | - Giacomo Marchi
- Department of Medicine, Section of Internal Medicine, EuroBloodNet CenterUniversity of Verona and Azienda Ospedaliera Universitaria Integrata VeronaVeronaItaly
| | - Nicola Montano
- Department of Clinical Sciences and Community HealthUniversity of MilanMilanItaly
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoInternal Medicine, Immunology and Allergology UnitMilanItaly
| | - Valter Monzani
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoHigh Care Internal Medicin UnitMilanItaly
| | - Flora Peyvandi
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoUOC Medicina Generale Emostasi e TrombosiMilanItaly
| | - Marco Proietti
- Geriatric UnitIRCCS Istituti Clinici Scientifici MaugeriMilanItaly
- Department of Clinical Sciences and Community HealthUniversity of MilanMilanItaly
- Liverpool Centre for Cardiovascular ScienceUniversity of Liverpool and Liverpool Heart & Chest HospitalLiverpoolUK
| | | | - Luca Valenti
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoDepartment of Transfusion Medicine and Hematology, Biological Resource Center and Precision Medicine LabMilanItaly
| | | | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine, EuroBloodNet CenterUniversity of Verona and Azienda Ospedaliera Universitaria Integrata VeronaVeronaItaly
| | - Alessandro Protti
- Department of Biomedical SciencesHumanitas UniversityPieve Emanuele (Milan)Italy
- Department of Anesthesia and Intensive Care UnitsIRCCS‐Humanitas Research HospitalRozzano (Milan)Italy
| | - Irene Motta
- Fondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoGeneral Medicine UnitMilanItaly
- Department of Clinical Sciences and Community HealthUniversity of MilanMilanItaly
| |
Collapse
|
17
|
Juanes‐Velasco P, García‐Vaquero ML, Landeira‐Viñuela A, Lopez‐Campos JL, Marín C, Lecrevisse Q, Arias‐Hidalgo C, Montalvillo E, Góngora R, Hernández Á, Fuentes M. Systematic evaluation of plasma signaling cascades by functional proteomics approaches: SARS-CoV-2 infection as model. Proteomics Clin Appl 2022; 16:e2100100. [PMID: 36168869 PMCID: PMC9537801 DOI: 10.1002/prca.202100100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 09/09/2022] [Accepted: 09/21/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE Acute phase reactants (APRs) play a critical role in inflammation. The difference in their physiological functions or the different dynamic ranges of these proteins in plasma makes it difficult to detect them simultaneously and to use several of these proteins as a tool in clinical practice. EXPERIMENTAL DESIGN A novel multiplex assay has been designed and optimized to carry out a high-throughput and simultaneous screening of APRs, allowing the detection of each of them at the same time and in their corresponding dynamic range. RESULTS Using Sars-CoV-2 infection as a model, it has been possible to profile different patterns of acute phase proteins that vary significantly between healthy and infected patients. In addition, severity profiles (acute respiratory distress syndrome and sepsis) have been established. CONCLUSIONS AND CLINICAL RELEVANCE Differential profiles in acute phase proteins can serve as a diagnostic and prognostic tool, among patient stratification. The design of this new platform for their simultaneous detection paves the way for them to be more extensive use in clinical practice.
Collapse
Affiliation(s)
- Pablo Juanes‐Velasco
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Marina L. García‐Vaquero
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Alicia Landeira‐Viñuela
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - José Luis Lopez‐Campos
- Unidad Médico‐Quirúrgica de Enfermedades Respiratorias. Instituto de Biomedicina de Sevilla (IBiS)Hospital Universitario Virgen del Rocío/Universidad de SevillaSpain,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)Instituto de Salud Carlos IIIMadridSpain
| | - Carmen Marín
- Instituto de Biomedicina de Sevilla (IBiS)Hospital Universitario Virgen del RocíoSevillaSpain
| | - Quentin Lecrevisse
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Carlota Arias‐Hidalgo
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Enrique Montalvillo
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Rafael Góngora
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain
| | - Ángela‐Patricia Hernández
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain,Department of Pharmaceutical Sciences: Organic Chemistry; Faculty of PharmacyUniversity of Salamanca, CIETUS, IBSALSalamanca37007Spain
| | - Manuel Fuentes
- Department of Medicine and Cytometry General Service‐Nucleus, CIBERONCCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)SalamancaSpain,Proteomics UnitCancer Research Centre (IBMCC/CSIC/USAL/IBSAL)Salamanca37007Spain
| |
Collapse
|
18
|
Craina M, Iacob D, Dima M, Bernad S, Silaghi C, Moza A, Pantea M, Gluhovschi A, Bernad E. Clinical, Laboratory, and Imaging Findings of Pregnant Women with Possible Vertical Transmission of SARS-CoV-2-Case Series. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10916. [PMID: 36078626 PMCID: PMC9518023 DOI: 10.3390/ijerph191710916] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 06/15/2023]
Abstract
The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic significantly impacted the general population's health. At times, the infection has unfavorably influenced pregnancy evolution and the result of birth. However, vertical transmission of the virus is rare and generates controversial discussions. The study aimed to highlight the clinical, laboratory, and imaging findings of pregnant women with confirmed Coronavirus Disease 2019 (COVID-19) with possible vertical transmission and identify possible factors that encourage vertical transmission. Between 1 April 2020 and 31 December 2021, 281 pregnant women diagnosed with COVID-19 gave birth in the Obstetrics and Gynecology Departments of the tertiary unit of County Emergency Clinical Hospital from Timisoara. Three newborns (1.06%) tested positive. The characteristic of these three cases was described as a short series. In two cases, the patients were asymptomatic. In one case, the patient developed a mild form of COVID-19 with a favorable evolution in all cases. We did not identify the presence of smoking history, vaccine before admission, atypical presentation, fever, or chest X-ray abnormalities. We note possible factors that encourage vertical transmission: Pregnancy-induced hypertension, thrombophilia, asymptomatic cough, an asymptomatic or mild form of the disease, a ruptured membrane, and cesarean. The laboratory results highlight the inconstant presence of some changes found in the list of potential predictors of the severity of the infection: Lymphopenia, high values of C-reactive protein, D-dimer, fibrinogen, platelets, Aspartate Aminotransferase, Lactate dehydrogenase, and ferritin. The study's conclusion of this small group suggests that there may have been an intrauterine infection in late pregnancy and described characteristics of the pregnant women. Possible risk factors that could encourage vertical transmission have been identified.
Collapse
Affiliation(s)
- Marius Craina
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Clinic of Obstetrics and Gynecology, County Clinical Emergency Hospital “Pius Brinzeu”, 300723 Timisoara, Romania
| | - Daniela Iacob
- Department of Neonatology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Mirabela Dima
- Department of Neonatology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Sandor Bernad
- Romanian Academy Timisoara Branch, Mihai Viteazul Avenue, 24, 300275 Timisoara, Romania
| | - Carmen Silaghi
- Clinic of Obstetrics and Gynecology, County Clinical Emergency Hospital “Pius Brinzeu”, 300723 Timisoara, Romania
| | - Andreea Moza
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Manuela Pantea
- Department of Neonatology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Adrian Gluhovschi
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Elena Bernad
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Clinic of Obstetrics and Gynecology, County Clinical Emergency Hospital “Pius Brinzeu”, 300723 Timisoara, Romania
| |
Collapse
|
19
|
Li Y, Luo W, Liang B. Circulating trace elements status in COVID-19 disease: A meta-analysis. Front Nutr 2022; 9:982032. [PMID: 36034929 PMCID: PMC9411985 DOI: 10.3389/fnut.2022.982032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/27/2022] [Indexed: 12/18/2022] Open
Abstract
Trace elements are a group of essential metals or metalloids, which are necessary for life, and present in minute amounts. Despite substantial researches highlighting the importance of trace elements in Coronavirus disease 2019 (COVID-19) diseases, a thorough evaluation of the levels of circulating trace elements is lacking. Therefore, we conducted a systematic review and meta-analysis to evaluate the trace element status (Zn, Fe, Cu, Mg, and Se) in COVID-19 disease. We also assessed the relationship between circulating trace elements and COVID-19 disease severity and survival status during follow-up. We searched comprehensively MEDLINE, Web of Science, CNKI, and WangFang databases without language restriction, between November 1, 2019 and April 1, 2022. The search identified 1,566 preliminary references. A total of 49 studies met the eligibility criteria and were included in the review, and 42 studies were included in the final meta-analysis. Meta-analysis showed that COVID-19 patients had significantly lower circulating Zn (SMD: -0.83, 95% CI: -1.19 to -0.46, P < 0.001), Fe (SMD: -1.56, 95% CI: -2.90 to -0.21, P = 0.023), and Se (SMD: -0.75, 95% CI: -0.94 to -0.56, P < 0.001) levels than healthy controls, and circulating Zn (SMD: -0.47, 95% CI: -0.75 to -0.18, P = 0.002), Fe (SMD: -0.45, 95% CI: -0.79 to -0.12, P = 0.008), and Se (SMD: -0.27, 95% CI: -0.49 to -0.04, P = 0.020) levels were associated with the presence of severity status in COVID-19 patients. Moreover, circulating Fe levels in non-survivors were significantly lower than survivors in COVID-19 (SMD: -0.28, 95% CI: -0.44 to -0.12, P = 0.001). However, there was no significant difference in Cu and Mg levels between COVID-19 patients and controls, severity and non-severity status, and survivors and non-survivors (all P > 0.05). Taken together, COVID-19 patients displayed lower circulating levels of Zn, Fe, and Se, and their levels were associated with severity status. Moreover, circulating Fe levels may provide part of the explanation for the unfavorable survival status. Therefore, we presumed optimistically that supplements of trace elements might provide an adjutant treatment in the early stages of COVID-19. Systematic review registration [https://www.crd.york.ac.uk/prospero], identifier [CRD42022348599].
Collapse
Affiliation(s)
- Yunhui Li
- Clinical Laboratory, PLA North Military Command Region General Hospital, Shenyang, China
| | - Weihe Luo
- Department of Medical Engineering, PLA North Military Command Region General Hospital, Shenyang, China
| | - Bin Liang
- Department of Bioinformatics, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, China
| |
Collapse
|
20
|
COVID-19 Severity and Mortality in Two Pandemic Waves in Poland and Predictors of Poor Outcomes of SARS-CoV-2 Infection in Hospitalized Young Adults. Viruses 2022; 14:v14081700. [PMID: 36016322 PMCID: PMC9413321 DOI: 10.3390/v14081700] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023] Open
Abstract
SARS-CoV-2 variants pose a significant threat to global public health. However, their influence on disease severity, especially among young adults who may exhibit different clinical characteristics, is debatable. In this retrospective study of 229 young adults hospitalized with COVID-19, we investigated the differences between Poland's second and third waves of the pandemic. To identify potential predictors of severe COVID-19 in young adults, we analyzed patient characteristics and laboratory findings between survivors and non-survivors and we performed logistic regression to assess the risk of death, mechanical ventilation, and intensive care unit treatment. We found no increase in COVID-19 severity comparing the third and second waves of the pandemic, indicating that the alpha variant had no influence on disease severity. In addition, we found that factors, such as obesity, comorbidities, lung involvement, leukocytosis, neutrophilia, lymphopenia, higher IG count, the neutrophil-to-lymphocyte ratio, C-reactive protein, procalcitonin, interleukin-6, D-Dimer, lactate dehydrogenase, high-sensitive troponin I, creatine kinase-myocardial band, myoglobin, N-terminal-pro-B-type natriuretic peptide, creatinine, urea and gamma-glutamyl transferase, lower estimated glomerular filtration rate, albumin, calcium and vitamin D3, possibly a decrease in red blood cell counts, hemoglobin and hematocrit, and an increase in creatine kinase during hospitalization may be associated with poor outcomes of COVID-19.
Collapse
|
21
|
The Impact of Iron Dyshomeostasis and Anaemia on Long-Term Pulmonary Recovery and Persisting Symptom Burden after COVID-19: A Prospective Observational Cohort Study. Metabolites 2022; 12:metabo12060546. [PMID: 35736479 PMCID: PMC9228477 DOI: 10.3390/metabo12060546] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is frequently associated with iron dyshomeostasis. The latter is related to acute disease severity and COVID-19 convalescence. We herein describe iron dyshomeostasis at COVID-19 follow-up and its association with long-term pulmonary and symptomatic recovery. The prospective, multicentre, observational cohort study “Development of Interstitial Lung Disease (ILD) in Patients With Severe SARS-CoV-2 Infection (CovILD)” encompasses serial extensive clinical, laboratory, functional and imaging evaluations at 60, 100, 180 and 360 days after COVID-19 onset. We included 108 individuals with mild-to-critical acute COVID-19, whereas 75% presented with severe acute disease. At 60 days post-COVID-19 follow-up, hyperferritinaemia (35% of patients), iron deficiency (24% of the cohort) and anaemia (9% of the patients) were frequently found. Anaemia of inflammation (AI) was the predominant feature at early post-acute follow-up, whereas the anaemia phenotype shifted towards iron deficiency anaemia (IDA) and combinations of IDA and AI until the 360 days follow-up. The prevalence of anaemia significantly decreased over time, but iron dyshomeostasis remained a frequent finding throughout the study. Neither iron dyshomeostasis nor anaemia were related to persisting structural lung impairment, but both were associated with impaired stress resilience at long-term COVID-19 follow-up. To conclude, iron dyshomeostasis and anaemia are frequent findings after COVID-19 and may contribute to its long-term symptomatic outcome.
Collapse
|
22
|
Roth-Walter F. Iron-Deficiency in Atopic Diseases: Innate Immune Priming by Allergens and Siderophores. FRONTIERS IN ALLERGY 2022; 3:859922. [PMID: 35769558 PMCID: PMC9234869 DOI: 10.3389/falgy.2022.859922] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Although iron is one of the most abundant elements on earth, about a third of the world's population are affected by iron deficiency. Main drivers of iron deficiency are beside the chronic lack of dietary iron, a hampered uptake machinery as a result of immune activation. Macrophages are the principal cells distributing iron in the human body with their iron restriction skewing these cells to a more pro-inflammatory state. Consequently, iron deficiency has a pronounced impact on immune cells, favoring Th2-cell survival, immunoglobulin class switching and primes mast cells for degranulation. Iron deficiency during pregnancy increases the risk of atopic diseases in children, while both children and adults with allergy are more likely to have anemia. In contrast, an improved iron status seems to protect against allergy development. Here, the most important interconnections between iron metabolism and allergies, the effect of iron deprivation on distinct immune cell types, as well as the pathophysiology in atopic diseases are summarized. Although the main focus will be humans, we also compare them with innate defense and iron sequestration strategies of microbes, given, particularly, attention to catechol-siderophores. Similarly, the defense and nutritional strategies in plants with their inducible systemic acquired resistance by salicylic acid, which further leads to synthesis of flavonoids as well as pathogenesis-related proteins, will be elaborated as both are very important for understanding the etiology of allergic diseases. Many allergens, such as lipocalins and the pathogenesis-related proteins, are able to bind iron and either deprive or supply iron to immune cells. Thus, a locally induced iron deficiency will result in immune activation and allergic sensitization. However, the same proteins such as the whey protein beta-lactoglobulin can also transport this precious micronutrient to the host immune cells (holoBLG) and hinder their activation, promoting tolerance and protecting against allergy. Since 2019, several clinical trials have also been conducted in allergic subjects using holoBLG as a food for special medical purposes, leading to a reduction in the allergic symptom burden. Supplementation with nutrient-carrying lipocalin proteins can circumvent the mucosal block and nourish selectively immune cells, therefore representing a new dietary and causative approach to compensate for functional iron deficiency in allergy sufferers.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, University of Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- *Correspondence: Franziska Roth-Walter ;
| |
Collapse
|
23
|
Mitochondrial Respiration in Response to Iron Deficiency Anemia: Comparison of Peripheral Blood Mononuclear Cells and Liver. Metabolites 2022; 12:metabo12030270. [PMID: 35323713 PMCID: PMC8950937 DOI: 10.3390/metabo12030270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/14/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Iron is an essential component for metabolic processes, including oxygen transport within hemoglobin, tricarboxylic acid (TCA) cycle activity, and mitochondrial energy transformation. Iron deficiency can thus lead to metabolic dysfunction and eventually result in iron deficiency anemia (IDA), which affects approximately 1.5 billion people worldwide. Using a rat model of IDA induced by phlebotomy, we studied the effects of IDA on mitochondrial respiration in peripheral blood mononuclear cells (PBMCs) and the liver. Furthermore, we evaluated whether the mitochondrial function evaluated by high-resolution respirometry in PBMCs reflects corresponding alterations in the liver. Surprisingly, mitochondrial respiratory capacity was increased in PBMCs from rats with IDA compared to the controls. In contrast, mitochondrial respiration remained unaffected in livers from IDA rats. Of note, citrate synthase activity indicated an increased mitochondrial density in PBMCs, whereas it remained unchanged in the liver, partly explaining the different responses of mitochondrial respiration in PBMCs and the liver. Taken together, these results indicate that mitochondrial function determined in PBMCs cannot serve as a valid surrogate for respiration in the liver. Metabolic adaptions to iron deficiency resulted in different metabolic reprogramming in the blood cells and liver tissue.
Collapse
|
24
|
Implication of COVID-19 on Erythrocytes Functionality: Red Blood Cell Biochemical Implications and Morpho-Functional Aspects. Int J Mol Sci 2022; 23:ijms23042171. [PMID: 35216286 PMCID: PMC8878454 DOI: 10.3390/ijms23042171] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
Several diseases (such as diabetes, cancer, and neurodegenerative disorders) affect the morpho-functional aspects of red blood cells, sometimes altering their normal metabolism. In this review, the hematological changes are evaluated, with particular focus on the morphology and metabolic aspects of erythrocytes. Changes in the functionality of such cells may, in fact, help provide important information about disease severity and progression. The viral infection causes significant damage to the blood cells that are altered in size, rigidity, and distribution width. Lower levels of hemoglobin and anemia have been reported in several studies, and an alteration in the concentration of antioxidant enzymes has been shown to promote a dangerous state of oxidative stress in red blood cells. Patients with severe COVID-19 showed an increase in hematological changes, indicating a progressive worsening as COVID-19 severity progressed. Therefore, monitored hematological alterations in patients with COVID-19 may play an important role in the management of the disease and prevent the risk of a severe course of the disease. Finally, monitored changes in erythrocytes and blood, in general, may be one of the causes of the condition known as Long COVID.
Collapse
|
25
|
Burkert FR, Lanser L, Bellmann-Weiler R, Weiss G. Coronavirus Disease 2019: Clinics, Treatment, and Prevention. Front Microbiol 2021; 12:761887. [PMID: 34858373 PMCID: PMC8631905 DOI: 10.3389/fmicb.2021.761887] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/21/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by a novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), emerged at the end of 2019 in China and affected the entire world population, either by infection and its health consequences, or by restrictions in daily life as a consequence of hygiene measures and containment strategies. As of September 2021, more than 231,000.000 infections and 4,740.000 deaths due to COVID-19 have been reported. The infections present with varied clinical symptoms and severity, ranging from asymptomatic course to fatal outcome. Several risk factors for a severe course of the disease have been identified, the most important being age, gender, comorbidities, lifestyle, and genetics. While most patients recover within several weeks, some report persistent symptoms restricting their daily lives and activities, termed as post-COVID. Over the past 18months, we have acquired significant knowledge as reflected by an almost uncountable number of publications on the nature of the underlying virus and its evolution, host responses to infection, modes of transmission, and different clinical presentations of the disease. Along this line, new diagnostic tests and algorithms have been developed paralleled by the search for and clinical evaluation of specific treatments for the different stages of the disease. In addition, preventive non-pharmacological measures have been implemented to control the spread of infection in the community. While an effective antiviral therapy is not yet available, numerous vaccines including novel vaccine technologies have been developed, which show high protection from infection and specifically from a severe course or death from COVID-19. In this review, we tried to provide an up-to-date schematic of COVID-19, including aspects of epidemiology, virology, clinical presentation, diagnostics, therapy, and prevention.
Collapse
Affiliation(s)
- Francesco Robert Burkert
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Innsbruck Medical University, Innsbruck, Austria
| | - Lukas Lanser
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Innsbruck Medical University, Innsbruck, Austria
| | - Rosa Bellmann-Weiler
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Innsbruck Medical University, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
26
|
Lanser L, Fuchs D, Kurz K, Weiss G. Physiology and Inflammation Driven Pathophysiology of Iron Homeostasis-Mechanistic Insights into Anemia of Inflammation and Its Treatment. Nutrients 2021; 13:3732. [PMID: 34835988 PMCID: PMC8619077 DOI: 10.3390/nu13113732] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023] Open
Abstract
Anemia is very common in patients with inflammatory disorders. Its prevalence is associated with severity of the underlying disease, and it negatively affects quality of life and cardio-vascular performance of patients. Anemia of inflammation (AI) is caused by disturbances of iron metabolism resulting in iron retention within macrophages, a reduced erythrocyte half-life, and cytokine mediated inhibition of erythropoietin function and erythroid progenitor cell differentiation. AI is mostly mild to moderate, normochromic and normocytic, and characterized by low circulating iron, but normal and increased levels of the storage protein ferritin and the iron hormone hepcidin. The primary therapeutic approach for AI is treatment of the underlying inflammatory disease which mostly results in normalization of hemoglobin levels over time unless other pathologies such as vitamin deficiencies, true iron deficiency on the basis of bleeding episodes, or renal insufficiency are present. If the underlying disease and/or anemia are not resolved, iron supplementation therapy and/or treatment with erythropoietin stimulating agents may be considered whereas blood transfusions are an emergency treatment for life-threatening anemia. New treatments with hepcidin-modifying strategies and stabilizers of hypoxia inducible factors emerge but their therapeutic efficacy for treatment of AI in ill patients needs to be evaluated in clinical trials.
Collapse
Affiliation(s)
- Lukas Lanser
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Katharina Kurz
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|