Next Article in Journal
Ursodeoxycholic Acid Modulates the Interaction of miR-21 and Farnesoid X Receptor and NF-κB Signaling
Next Article in Special Issue
Post-Acute Sequelae of SARS-CoV-2 Infection (PASC) for Patients—3-Year Follow-Up of Patients with Chronic Kidney Disease
Previous Article in Journal
Perceived Stress and Life Stressors in Adults with and without Fibromyalgia
Previous Article in Special Issue
Special Issue “Latest Research in Post-COVID (Long COVID): Pathological and Treatment Studies of Sequelae and Complications”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Hemoglobin and Its Relationship with Fatigue in Long-COVID Patients Three to Six Months after SARS-CoV-2 Infection

by
Somayeh Bazdar
1,2,3,
Lizan D. Bloemsma
1,2,3,
Nadia Baalbaki
1,2,3,
Jelle M. Blankestijn
1,2,3,
Merel E. B. Cornelissen
1,2,3,
Rosanne J. H. C. G. Beijers
4,
Brigitte M. Sondermeijer
5,
Yolanda van Wijck
1,2,3,
George S. Downward
6,7 and
Anke H. Maitland-van der Zee
1,2,3,*,† on behalf of the P4O2 Consortium
1
Department of Pulmonary Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
2
Amsterdam Institute for Infection and Immunity, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
3
Amsterdam Public Health Research Institute, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands
4
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
5
Department of Pulmonology, Spaarne Gasthuis, 2035 RC Haarlem, The Netherlands
6
Department of Environmental Epidemiology, Institute for Risk Assessment Sciences (IRAS), Utrecht University, 3584 CL Utrecht, The Netherlands
7
Department of Global Public Health & Bioethics, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
*
Author to whom correspondence should be addressed.
Current address: Department of Respiratory Medicine, Amsterdam UMC, 1105 AZ Amsterdam, The Netherlands.
Biomedicines 2024, 12(6), 1234; https://doi.org/10.3390/biomedicines12061234
Submission received: 11 February 2024 / Revised: 23 May 2024 / Accepted: 24 May 2024 / Published: 1 June 2024

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.

1. Introduction

Over four years ago, the World Health Organization (WHO) declared COVID-19 a global pandemic. However, infections from SARS-CoV-2 and their long-term consequences remain a global challenge [1,2]. Of particular concern are the many aspects of the long-term outcomes that remain unknown, especially among those who have survived a SARS-CoV-2 infection [3,4]. The term “Long-COVID” refers to the condition in which patients experience a variety of persisting symptoms long after the acute phase of the infection has resolved but often typified by fatigue, dyspnea, brain fog, sleep problems, muscle stiffness, and mental problems [1,5,6]. These symptoms are not restricted to pulmonary manifestations; and just like the acute phase of the disease, organs other than the lungs may be impacted [7,8]. However, many long-term symptoms may not have been present in the acute phase.
Long-COVID is causing widespread disability worldwide, with many potentially developing lifelong disabilities. Nevertheless, current diagnostics and treatments are insufficient, necessitating urgent clinical investigations to address hypothesized underlying biological mechanisms [1]. Whilst the mechanism of long-COVID continues to be unclear due to the wide clinical spectrum and organ involvement, only a thorough understanding of its pathophysiology will allow us to assess and treat the disease’s side effects [9]. While persistence of respiratory symptoms, particularly dyspnea and cough, appear to be common in Long-COVID, some of the other persistent symptoms of Long-COVID may be part of a multisystem disease, and interdisciplinary treatment of these chronic sequelae is necessary [10].
A decrease in hemoglobin may be one of the non-respiratory complications of a SARS-CoV-2 infection. In the acute phase of infection, a reduction in hemoglobin is likely mediated via interactions between viral agents and host cells and has previously been found to be a predictive factor for severe respiratory failure in pneumonic COVID-19 patients [11,12]. Based on the results of multivariate regression of a study comprising 1147 COVID-19 patients across five tertiary pediatric hospitals in Asia, higher levels of hemoglobin and platelets were protective against severe/critical COVID-19 infection [11]. A retrospective investigation of 23 COVID-19 hospitalized patients with pneumonia also revealed a significant association between higher levels of hemoglobin and a reduced need for mechanical ventilation administration, with an odds ratio of 0.313 [12]. Various mechanisms for decreased hemoglobin have been proposed, including the conversion of Hb into other forms such as methemoglobin (MetHb) and carboxyhemoglobin (COHb) [13]. While oxidative stress, which is linked to infectious diseases, may contribute [14], medications given during treatment may also be relevant, given that certain medicines such as hydroxychloroquine used to treat COVID-19 have a high potential for oxidation, which can lead to MetHb and COHb [15]. An increase in COHb during SARS-CoV-2 infection may also potentially be related to hemolytic anemia, and an increase in MetHb is a result of hemoglobin oxidation that could also be driven by nitric oxide (NO) [13]. Besides oxidative stress, hyperinflammation may be another reason for developing new-onset anemia during SARS-CoV-2 infection [16]. Investigations of hospitalized patients with COVID-19 disease revealed that increased inflammatory indices such as Interleukin 6 (IL-6) and C-reactive protein (CRP) were increased in those with COVID-19-induced anemia [17].
Anemia is defined as a decrease in hemoglobin (Hb) concentrations and/or red blood cell (RBC) absolute counts leading to an inadequate supply of physiological requirements, and its common diagnostic method is to assess the Hb level [18]. This hematologic disease is typically a multifaceted medical condition, and different factors such as the patient’s medical history and the underlying pathological mechanism(s) should be considered while diagnosing and categorizing it [19]. Common manifestations of anemia include a wide range of symptoms such as pale skin, shortness of breath, palpitations, chest pain, dizziness, loss of strength, weakness, and fatigue [20]. According to studies on the prevalence of fatigue and its associated factors, anemia is one of the most frequently found risk factors linked to fatigue in patients with chronic kidney disease (CKD) [21].
Fatigue is the most common symptom in Long-COVID patients [22]. While anemia, psychosocial problems, sleep disorders, and sleep-related respiratory disorders are considered common causes of fatigue in general, cancer, thyroid dysfunction, and other somatic diseases can also be reasons [23]. In a study of 42 men and 33 women, Pasini et.al. reported a mean hemoglobin of 11.5 g/dL (normal range: 14–18 g/dL) among men and 10.9 g/dL (normal range: 12–16 g/dL) among women 60 days following their hospital discharge after COVID-19 [24]. Furthermore, in a 6-month follow-up of 412 ex-COVID-19 patients, it was reported that the average hemoglobin level was significantly lower in patients suffering from fatigue compared to those who were not (13.8 vs. 14.3 g/dL). However, after performing multivariate regression, no statistically significant associations were observed between hemoglobin, other cell blood count measurements (including inflammatory biomarkers), and Long-COVID symptoms [25].
A better understanding of the mechanisms involved in Long-COVID and hemoglobin status/anemia will allow better treatment of those suffering from this condition. While there is some evidence that Long-COVID patients have decreased hemoglobin levels, its link with inflammatory indices as well as its role in developing fatigue symptoms remain unclear. Finding biomarkers that can help to direct optimized care to patients most at risk is crucial, and since hemoglobin is easily assessed by assay and has been associated with Long-COVID, we believe that it is a biomarker worthy of further investigation. The purpose of the current study is therefore to examine the correlations between hemoglobin levels and inflammatory biomarkers and to evaluate the potential role of the inflammatory process in decreasing hemoglobin levels in Long-COVID patients. Furthermore, this study investigates the association between hemoglobin and fatigue in a cohort of Long-COVID patients to evaluate whether hemoglobin levels contribute to developing fatigue in these patients.

2. Methods and Materials

2.1. Study Setting and Participants

The population for the current study consists of participants of the Precision Medicine for More Oxygen (P4O2) COVID-19 cohort. Details on the study design of this cohort have been published elsewhere [26]. In brief, P4O2 COVID-19 is a prospective cohort study that recruited 95 Long-COVID patients, aged 40–65 years, from outpatient Long-COVID clinics in five hospitals in the Netherlands. Two research visits were performed for each patient between months 3–6 and months 12–18 after SARS-CoV-2 infection, during which questionnaires were administered, and physical examinations and biological sampling were performed. There were no specific exclusion criteria for the FU study visit of this cohort, and nine months following the initial study visit, all participants received invitations for a follow-up appointment, but a portion was lost to follow-up. Information about the acute phase of the disease was extracted from patients’ medical charts. Participants for whom a hemoglobin measurement was available, either during SARS-CoV-2 infection or their first follow-up (FU) visit 3–6 months after SARS-CoV-2 infection, or both, were eligible for analysis in the current paper. The second study visit was conducted 12–18 months after infection, which included a fairly high number of lost to FU among whom only 11 provided blood samples for hemoglobin.

2.2. Ethical Considerations

The study protocol for the P4O2 COVID-19 cohort was approved by the ethical committee of Amsterdam UMC (reference number NL74701.018.20), and all participants provided written informed consent prior to their enrolment.

2.3. Variables in This Study

Measurements during the acute phase of the SARS-CoV-2 infection (baseline) and FU laboratory measurements of hemoglobin were assessed via venipuncture, performed under aseptic conditions, and analyzed following standard laboratory procedures in each participating hospital. To be eligible for the current study, a hemoglobin level either at baseline or FU (n = 93) was required. A subset of patients (n = 27) did not provide standard venipuncture at the FU visit but most of them did provide a hemoglobin value measured via finger prick test during pulmonary function testing (generally occurring concurrently with the FU visit), which was used instead, and this way all 95 participants were eligible for the current analysis. As this method was expected to provide results somewhat different from those of venipuncture, the hemoglobin values from these participants were only used to examine whether their hemoglobin levels changed by >10% (i.e., clinically significant difference, as described below). Anemia was defined, as per the reference range at the measuring laboratory, as a hemoglobin level less than 7.5 mmol/L in women and less than 8.5 mmol/L in men according to the reference laboratory range. Absolute changes in hemoglobin were calculated as the difference between FU and baseline hemoglobin and were classified into increased, decreased, or unchanged hemoglobin. In addition, “clinically relevant” changes in hemoglobin were determined by an increase or decrease in hemoglobin of 10% or more. As there is no universal definition of a clinically significant change in hemoglobin levels, we utilized the determination of obstetrics and gynecology references which usually recognize a decrease in hemoglobin level if it is more than 10% decrease [27,28,29].
The Fatigue Severity Scale (FSS) standard questionnaire was used to categorize patients into those with or without fatigue. The total FSS was the sum of the nine questions and the average FSS was calculated by dividing this sum by nine [30]. An average score equal to or more than four was considered as a positive indicator of fatigue, similar to previous research in the P4O2 COVID study [26]. The fatigue severity scale originally utilized a cut-off of 4 or more to indicate severe fatigue. We acknowledge that different cut-offs have been developed, including low (<4), medium/borderline (4–5), and high/severe (>5) [31]. However, for simplicity, and consistency with our other work in this population [26] we have chosen to consistently use 4 as a cut-off point.
Blood samples were also examined for a variety of biomarkers, and markers that indicated the presence of an inflammatory process were included as inflammatory indices in the current study. Identified markers were D-Dimer, Neutrophil/Lymphocyte ratio (NLR), Immature Granulocyte (IG), C-reactive protein (CRP), Creatine phosphokinase (CPK), and Lactate Dehydrogenase (LDH) [32,33,34]. Normal ranges and analytical approaches were as per the standard operating procedures at participating hospitals.
Additional variables assessed in this paper were: general demographics (i.e., age, sex, and ethnicity), past medical history of underlying diseases (e.g., hypertension, diabetes, cardiovascular disease, kidney diseases, asthma, etc.), history of hospital stay (including length and any ICU admission), dominant SARS-CoV-2 variant during admission, body mass index (BMI), vaccination status, and glomerular filtration rate (GFR—based on serum creatinine).

2.4. Statistical Analysis

Continuous variables were described by mean (SD) for variables with a normal distribution (age, BMI, baseline hemoglobin, FU hemoglobin, hemoglobin change) or median (25th percentile, 75th percentile) for those that were not. Categorical data were described by frequency (percent). Characteristics were also stratified by sex since the reference range for anemia differs between males and females. The Paired-Samples T-test was utilized to compare parametric variables between two timepoints (baseline and first FU visit) and the Independent Samples Test was used to compare parametric variables between two different groups (fatigue patients vs. non-fatigue patients, male vs. female). Chi-square testing was employed while comparing categorical variables between these groups, and for comparing non-parametric variables between them, the Mann–Whitney U test was used. The correlations between hemoglobin levels and inflammatory biomarkers were evaluated via Pearson’s correlation test for parametric values and Spearman’s correlation for non-parametric ones. A false discovery rate (FDR) was not used as it can be overly conservative especially when validating a smaller number of previously described features or in cases where the effective number of tests is smaller (due to redundancy in similar features measuring the same latent variables) [35,36]. Finally, logistic regression models were developed to assess the relationships between fatigue at 3–6 months after infection and hemoglobin levels (1) in the acute phase, (2) 3–6 months after SARS-CoV-2 infection, and (3) the difference in hemoglobin levels between infection and follow-up. Fatigue was defined as a binary (yes/no) variable based on the pre-defined FSS cutoff of equal or greater than four (yes) or less than four (no). Unadjusted models and models adjusted for age and sex were generated. Covariates for adjustment were identified through the generation of a Directed Acyclic Graph (DAG) (Figure 1) [37]. According to this DAG, age, sex, ethnicity, BMI, underlying diseases, hospital admission, ICU admission, inflammation, and lung function were all identified confounders for the potential pathway of developing fatigue due to hemoglobin alteration. However, considering the sample size and a target of 20 events per covariate, only age and sex, identified as the most important potential confounders, were included in the regression analysis.
Since renal function has the potential to affect both hemoglobin and fatigue, we performed a sensitivity analysis by restricting the regression analysis to those with GFR at follow-up greater than 60 mL/min/1.73 m2 (n = 56). This cut-off was chosen to differentiate those with normal/mild kidney dysfunction from more severe cases where the prevalence of anemia is higher [38,39,40]. Furthermore, since a case was detected among the participants with an extreme outlier value for hemoglobin (both baseline and FU), another sensitivity analysis was also performed after excluding this single case. All statistical analyses were performed by SPSS version 28 and a p-value ≤ 0.05 was considered statistically significant for all statistical tests.

3. Results

In total, 1 or more hemoglobin results were available from all 95 participants in the P4O2 COVID cohort, with 59 having data on the Hb level at both the acute phase and at FU (11 participants had missing data during the acute phase and 27 at FU). The mean age of our study population was 54.2 years, and 50.5% of them were men. Most of the participants (89.5%) were suffering from some form of underlying disease, and comorbidities were more common in men than in women (93.8% vs. 85.1%). According to the WHO severity index, most of the infections were categorized as moderate (61.3%) or severe (28.0%). The characteristics that were significantly different between men and women were fatigue average score during the Long-COVID phase (83.3% in females and 68.9% in males, p-value = 0.026 according to Independent Samples Test), ICU admission (19.1% in women and 38.3% in men, p-value = 0.044 according to Chi-Square test), ICU stay (median 0 (0–0) days in women and median 0 (0–8) days in men, p-value = 0.044 according to Mann–Whitney U test), and BMI (32.2 in women and 28.9 in men, p-value = 0.002 according to Independent Samples Test) (Table 1).

3.1. Hemoglobin Levels

The mean hemoglobin level during SARS-CoV-2 infection for our entire study population was 8.6 mmol/L (8.35 mmol/L in women and 8.84 mmol/L in men) (Table 2), which is within the normal laboratory reference range (>7.5 in women and >8.5 in males). During the acute phase, the average hemoglobin concentration was 8.61 mmol/L and 8.70 mmol/L 3–6 months after infection (p-value for difference: 0.003 according to Paired-Samples T-test). Hemoglobin values were significantly different (via Independent Samples Test) between men and women both during the acute phase (8.84 vs. 8.35 mmol/L, respectively, p-value = 0.023) and at follow-up (9.04 vs. 8.36 mmol/L p-value = 0.003,) (Table 2). In patients with hemoglobin levels available at both time points (n = 59), the hemoglobin levels increased on average by 0.3 (from 8.41 mmol/L during the acute phase and 8.71 mmol/L 3–6 months after infection). This was more pronounced in men where the average hemoglobin increased from 8.89 mmol/L to 9.24 mmol/L (mean difference = 0.42 mmol/L). For female participants, the mean hemoglobin value remained the same at baseline and FU (8.35 mmol/L and 8.36 mmol/L, respectively, mean difference = 0.18 mmol/L). However, men and women’s differences in hemoglobin change did not reach statistical significance (p-value = 0.222 according to Independent Samples Test). Consistent with this overall increase, hemoglobin levels at FU were higher for 66.1% of our participants (N = 39), with 14 having an increase of greater than 10%. However, 28.8% (N = 17) of the participants had lower hemoglobin levels at FU, with 8 showing a decrease of greater than 10%. Also, in three cases the hemoglobin level during FU remained the same as the baseline value.
The prevalence of anemia during the acute phase of SARS-CoV-2 infection was 22.6% (19 cases, among whom 7 continued experiencing anemia during follow-up, 8 did not, and data were unavailable for four). This reduced to 16.4% at follow-up (n = 11, 3 were new-onset and 7 were persistent from the acute phase, and data were missing for 1, Table S1). Suffering from anemia was more common in men compared to women during both phases of COVID-19 disease (31.1% versus 12.8% during the acute phase (p-value = 0.046 according to Chi-Square Test) and 18.8% versus 13.9% during the Long-COVID phase (p-value = 0.587 according to Chi-Square Test).
We also examined demographic and clinical characteristics, by dividing the population into notable hemoglobin change (notable decrease versus notable increase or no change). Of note, the group with a notable decrease in hemoglobin had a longer hospital stay than those without (17 days vs. 8 days, Table S2).
When comparing hemoglobin levels between those with and without fatigue 3–6 months after infection, we observed that those experiencing fatigue had lower levels of hemoglobin both at baseline (8.53 vs. 8.89 mmol/L, respectively, p-value = 0.171 according to Independent Samples Test) and at FU (8.54 vs. 9.24 mmol/L, p-value = 0.046 according to Independent Samples Test) than those without fatigue (Table 2). Boxplots displaying the difference in hemoglobin level between those with fatigue versus those without are shown in Figure 2.

3.2. Correlations between Hemoglobin and Inflammatory Biomarkers

Results of Spearman correlation tests demonstrated that there were no strong or statistically significant correlations between hemoglobin levels (at baseline, follow-up, and the change in hemoglobin levels) and inflammatory biomarkers (Table 3). Among other variables that were found to be linked with hemoglobin level according to the DAG (Figure 1), the duration of hospital stay, as well as ICU stay had a positive correlation with hemoglobin change (rs = 0.268, and rs = 0.360, respectively) (Table S3).

3.3. Associations between Hemoglobin Levels and Fatigue

Results of logistic regression analysis indicated that higher venous hemoglobin concentrations were associated with lower odds of fatigue 3–6 months after infection (Table 4). While findings were non-significant, an association between increased Hb, and reduced fatigue was consistently observed. The strongest associations were observed with hemoglobin levels 3–6 months after infection where increased hemoglobin was associated with a reduced likelihood of fatigue. A 1 mmol/L increase in hemoglobin was associated with an adjusted odds ratio of 0.35 for fatigue (95%CI 0.12–1.02). Another strong relationship between fatigue and hemoglobin indices was observed for the change in hemoglobin levels, where a 1 mmol/L positive increase in Hb (i.e., greater in visit 2 than visit 1) resulted in an adjusted OR of 0.38 (95% CI 0.13–1.09). There was also a positive association between fatigue and anemia at both baseline and at follow-up (adjusted OR 1.56 [95% CI 0.42–5.78] and OR 2.25 [95% CI 0.25–20.20], respectively).
Results of the sensitivity analysis where patients with impaired renal function were excluded showed associations that were directionally consistent and comparable to those in the total population. For example, the adjusted ORs of the associations between fatigue and FU hemoglobin and hemoglobin change were, respectively, 0.475 (95% CI 0.159–1.419) per 1 mmol/L increase in hemoglobin, and 0.467 (95% CI 0.150–1.452) per 1 mmol/L increase in hemoglobin (Table S4). In addition, the results of the other sensitivity analysis, which excluded the case with the extreme outlier data for the hemoglobin variable, showed barely any variation from the findings of the regression analysis in the total population (Table S5).

4. Discussion

In the current study, we aimed to assess the role of hemoglobin levels during the acute phase of SARS-CoV-2 infection and 3–6 months after follow-up against inflammatory biomarkers and fatigue in the P4O2 COVID-19 study. Overall, we found that hemoglobin levels increased between the acute and follow-up phases and that the average hemoglobin concentrations in our study population were within the normal range. The proportion of anemic patients also decreased, from 22.6% to 16.4%. However, hemoglobin levels were significantly lower, both at baseline and at follow-up, in patients with fatigue compared to patients without fatigue. No correlations were detected between hemoglobin and inflammatory biomarkers.
Other work examining anemic COVID-19 patients has reported that the SARS-CoV-2 virus is a contributor to both causing and exacerbating anemia [39]. Further, anemia itself has also been shown to play a role in prolonging COVID-19 symptoms [41]. It is therefore possible that anemia, either pre-existing or caused by acute infection, may contribute to some (but not all) of the fatigue symptoms in those who suffer from Long-COVID [42]. In a similar study of 145 former COVID-19 patients, it was reported that while the prevalence of anemia, hyperferritinemia, and systemic thrombo-inflammation all gradually decreased, a sizable portion of the post-COVID population still showed persistent anemia (4.65–11.1%) or iron deficiency (16–35%) at different FU evaluation time points, which may have contributed to their ongoing symptom burden, including fatigue [43].
Fatigue is a multifaceted condition and although its specific etiology can be uncertain, it is usually identified as the main manifestation of anemic patients [44,45] Our regression analysis demonstrated that hemoglobin levels are lower in Long-COVID patients who suffer from fatigue in both the acute phase and 3–6 months afterward. This suggests that hemoglobin may play a role in the fatigue symptoms of Long-COVID patients. Nevertheless, as the current study is observational in nature, further investigation is necessary to establish causation as the link between anemia and fatigue is not always evident. For example, a review of the contributors to persistent fatigue in Myelodysplastic syndrome patients revealed that no clear association has been found between fatigue and anemia. [46]. Therefore, other potential factors including inflammation, sleep-related problems, and oxidative stress may provide alternate explanations.
Another finding in our study suggesting a role of hemoglobin in fatigue is that we also observed a higher percentage of women who reported fatigue (83.3% in women vs. 68.9% in men), while their mean hemoglobin level in the Long-COVID phase was lower than in men (8.36 vs. 9.04). However, BMI was also significantly different between men and women, which can also be associated with fatigue [47,48]. Another characteristic that varied between males and females was hemoglobin change, where men showed a higher rate of increase than women. Previous research has reported that increased erythropoiesis is a secondary outcome of higher testosterone concentrations in men [49], which may explain the difference observed here.
These findings may have potential clinical implications, providing evidence for physicians to consider monitoring the hemoglobin levels in their Long-COVID patients, particularly in those suffering from fatigue as their primary symptom. Whether fatigue can be managed by treating any underlying hemoglobin impairment is outside the scope of the current work; however, it is worth closer evaluation.
We did not observe any correlation between inflammatory markers and hemoglobin concentrations. This stands in contrast to other work such as that of Lanser et al., who examined inflammation-mediated anemia, reporting the impact of immune system stimulation and cytokine-mediated alterations on iron homeostasis [16]. The three inflammatory variables that they assessed in their study were ferritin, IL-6, and CRP—of which we only studied CRP. This difference may illustrate that only extremely severe inflammation can affect hemoglobin values, that our sample size was insufficient to identify an effect, or that the inflammatory biomarkers available for use in the current study were not optimal for the evaluation of inflammatory-driven Hb disruption. For example, the erythropoiesis-suppressing effects of specific inflammatory cytokines, such as TNFα, IL-1, and interferon-γ, have been reported in various investigations. Therefore, a broader assessment of inflammatory markers alongside other diagnostic tests for anemia related to inflammation (serum iron, ferritin, and hepcidin), may be a more accurate way to assess the correlation between anemia and inflammation [50]. For example, in an analysis of 214 ex-COVID-19 patients, where all aforementioned variables were included, patients who reported Long-COVID Syndrome months after the acute phase were distinguished by inflammation and abnormal iron homeostasis that lasted longer than two weeks after the onset of symptoms. This led the researchers to conclude that the effects of inflammatory iron dysregulation on erythropoiesis and blood oxygen transportation may be partially responsible for several of the characteristics of Long-COVID [51]. Alternately this might suggest that factors other than inflammation have a more significant role. For example, there is evidence that SARS-CoV-2 infection can produce hemolytic anemia by either generating structural changes in host cells (RBCs) via viral invasion or activating autoantibodies [52]. The fact that we could not validate the same link between inflammation and Hb that was reported by Hanson et al. emphasizes the need to further test the robustness of biomarker analyses [51]. Furthermore, a meta-analysis reviewing the studies evaluating the COVID-19 biomarkers revealed a number of findings, including different results per population, which highlights the significance of validation studies [53]. Especially, since long COVID is a relatively young disease correlations with biomarkers in one population should be validated in other populations. Additional work is therefore needed to further understand the mechanism of hemoglobin decrease in SARS-CoV-2 infection, and how (or if) these factors persist long-term.
An alternative to the proposed hemoglobin and inflammatory biomarkers explored here is the evidence-based association with oxidative stress [54]. Experiments have shown that oxidative stress significantly impacts the structure of hemoporphyrin, which in turn affects hemoglobin function and reduces the efficiency with which hemoglobin binds to oxygen. This process is caused by lipid peroxidation (LPO), which is followed by the formation of LPO’s primary and end products [55]. Understanding the potential role of oxidative stress in the development of hemoglobin decrease would be a significant finding that may lead to preventative and therapeutic approaches [56,57].
Among this study’s strengths is its well-defined population of Long-COVID patients, who have comprehensive information, including hospital and personal data. Additionally, these patients have an extensive list of fatigue symptoms obtained through standard fatigue questionnaires. However, this study also has some limitations. First, while SARS-CoV-2 infection is the exclusive focus of this investigation, it has been revealed that other viral infections, such as seasonal influenza, may have long-term effects similar to those following SARS-CoV-2 infection [58]. Thus, it is uncertain whether our findings are specific to this particular viral infectious disease, or indicative of post-viral syndromes more broadly. Our limited sample size restricts the overall statistical power, including for regression analysis, and limits the external generalizability of our findings. Further compounding this was missing values for several variables of interest, particularly the baseline variables of inflammatory biomarkers. Such data were only available if they had been ordered during the original presentation and admission in different hospitals. Imputation was considered but ultimately decided against as these were outcome variables. Towards the later part of the pandemic, follow-up visits were more often occurring remotely, meaning that biological sampling at this point was reduced. Furthermore, not having pre-infection hemoglobin data makes evaluating acute changes impossible. In addition, it has been shown that vaccination against SARS-CoV-2 might enhance the likelihood of hematologic problems, such as anemia [59] and on the other hand, that receiving the vaccine prior to SARS-CoV-2 infection might be associated with a lower risk of developing Long-COVID [60]. However, since the vaccination pattern varied greatly (Tables S6 and S7) and our sample size was not large enough, we were unable to analyze the association between anemia and vaccination status in our population. Finally, this cohort was started at a point in time when not much was known about Long-COVID and therefore no specific age group was known to be important at that time, and it seemed that it was mainly affecting individuals of 40 years and older. In addition, because older patients (above 65) are more likely to have more comorbidities that may interfere with the analysis we decided to limit our population to below that age. Thus, the age range of our study population (40 to 65 years) makes us more cautious in generalizing our findings to other age ranges.

5. Conclusions

In our study of Long-COVID patients, we observed that those with fatigue tended to have lower hemoglobin levels than those without. These findings suggest a role of hemoglobin in Long-COVID fatigue. To determine the causes of low hemoglobin levels in Long-COVID patients and in turn, the best course of action for treatment, further studies on the mechanisms influencing hemoglobin levels in Long-COVID are necessary. Furthermore, it is important to perform validation studies to examine whether our findings are reproducible and robust across different cohorts.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/biomedicines12061234/s1, Table S1: Frequency and persistence of anemia in the study population; Table S2: Characteristics of study population according to hemoglobin level change; Table S3: Correlations between hemoglobin levels and demographic and clinical characteristics; Table S4: Results of logistic regression model for Hemoglobin and fatigue (average FSS ≥ 4) for cases with GFR of mild renal dysfunction or better (≥60 mL/min/1.73m2, N = 56); Table S5: Results of logistic regression model for Hemoglobin and fatigue (average FSS ≥ 4) after excluding the case with extreme outlier for hemoglobin; Table S6: Frequency of SARS-CoV-2 vaccination (main dosage) in the study population; Table S7: Type of SARS-CoV-2 vaccination in the study population.

Author Contributions

Conceptualization, S.B., A.H.M.-v.d.Z., G.S.D., L.D.B., Y.v.W. and R.J.H.C.G.B.; methodology, S.B., A.H.M.-v.d.Z., L.D.B., G.S.D. and Y.v.W.; software, S.B., L.D.B. and J.M.B.; validation, S.B., A.H.M.-v.d.Z., G.S.D., L.D.B., Y.v.W., R.J.H.C.G.B., N.B., B.M.S., M.E.B.C. and J.M.B.; formal analysis, S.B. and J.M.B.; investigation, A.H.M.-v.d.Z., Y.v.W., R.J.H.C.G.B., B.M.S., N.B., S.B., M.E.B.C. and L.D.B.; resources, A.H.M.-v.d.Z., Y.v.W., B.M.S., R.J.H.C.G.B., N.B., S.B., M.E.B.C., L.D.B. and J.M.B.; data curation, A.H.M.-v.d.Z., Y.v.W., G.S.D., N.B., S.B., M.E.B.C., L.D.B. and J.M.B.; writing—original draft preparation, S.B.; writing—review and editing, all authors; visualization, S.B.; supervision, A.H.M.-v.d.Z., G.S.D., L.D.B. and Y.v.W.; project administration, A.H.M.-v.d.Z., L.D.B., Y.v.W., N.B. and M.E.B.C.; funding acquisition, A.H.M.-v.d.Z. All authors have read and agreed to the published version of the manuscript.

Funding

Partners in the Precision Medicine for more Oxygen (P4O2) consortium are the Amsterdam UMC, Leiden University Medical Center, Maastricht UMC+, Maastricht University, UMC Groningen, UMC Utrecht, Utrecht University, TNO, Abbvie, Aparito, Boehringer Ingelheim, Breathomix, Clear, Danone Nutricia Research, Fluidda, Ncardia, Olive, Ortec Logiqcare, Philips, Proefdiervrij, Quantib-U, RespiQ, Roche, Smartfish, SODAQ, Thirona, TopMD, Lung Alliance Netherlands (LAN), and the Lung Foundation Netherlands (Longfonds). The consortium is additionally funded by the PPP Allowance made available by Health~Holland, Top Sector Life Sciences & Health (LSHM20104; LSHM20068), to stimulate public-private partnerships and by Novartis. Prof. Dr. Anke-Hilse Maitland-van der Zee is the PI of P4O2.

Institutional Review Board Statement

This study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the ethical committee of Amsterdam UMC (reference number NL74701.018.20), and all participants provided written informed consent prior to their enrolment.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available upon request to the corresponding author. The data are not publicly available due to agreements made by the consortium, that only allow access by each consortium partner to specific data that answer their pre-specified research questions. A request for access to data by organizations outside of the consortium can be submitted to the P4O2 Data Committee (via [email protected]) and the research will need to be performed in collaboration with one of the P4O2 consortium partners.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Davis, H.E.; McCorkell, L.; Vogel, J.M.; Topol, E.J. Long COVID: Major findings, mechanisms and recommendations. Nat. Rev. Microbiol. 2023, 21, 133–146. [Google Scholar] [CrossRef] [PubMed]
  2. Saunders, C.; Sperling, S.; Bendstrup, E. A new paradigm is needed to explain long COVID. The Lancet. Respir. Med. 2023, 11, e12–e13. [Google Scholar]
  3. Wahlgren, C.; Forsberg, G.; Divanoglou, A.; Balkhed Å, Ö.; Niward, K.; Berg, S.; Levi, R. Two-year follow-up of patients with post-COVID-19 condition in Sweden: A prospective cohort study. Lancet Reg. Health–Eur. 2023, 28, 100595. [Google Scholar] [CrossRef] [PubMed]
  4. COVID, Gemelli Against, and Post-Acute Care Study Group. Post-COVID-19 global health strategies: The need for an interdisciplinary approach. Aging Clin. Exp. Res. 2020, 1, 1613–1620. [Google Scholar]
  5. NICE. COVID-19 Rapid Guideline: Managing the Long-Term Effects of COVID-19; National Institute for Health and Care Excellence (NICE): London, UK, 2020. [Google Scholar]
  6. Crook, H.; Raza, S.; Nowell, J.; Young, M.; Edison, P. Long covid—Mechanisms, risk factors, and management. BMJ 2021, 26, 374. [Google Scholar] [CrossRef] [PubMed]
  7. Vaes, A.W.; Goërtz, Y.M.; Van Herck, M.; Machado, F.V.; Meys, R.; Delbressine, J.M.; Houben-Wilke, S.; Gaffron, S.; Maier, D.; Burtin, C.; et al. Recovery from COVID-19: A sprint or marathon? 6-month follow-up data from online long COVID-19 support group members. ERJ Open Res. 2021, 7, 00141-2021. [Google Scholar] [CrossRef] [PubMed]
  8. Ayoubkhani, D.; Khunti, K.; Nafilyan, N.; Maddox, T.; Humberstone, B.; Diamond, S.I.; Banerjee, A. Epidemiology of post-COVID syndrome following hospitalisation with coronavirus: A retrospective cohort study. MedRxiv 2021. [Google Scholar] [CrossRef]
  9. Castanares-Zapatero, D.; Chalon, P.; Kohn, L.; Dauvrin, M.; Detollenaere, J.; de Noordhout, C.M.; Jong, C.P.-D.; Cleemput, I.; Heede, K.V.D. Pathophysiology and mechanism of long COVID: A comprehensive review. Ann. Med. 2022, 54, 1473–1487. [Google Scholar] [CrossRef]
  10. Montani, D.; Savale, L.; Noel, N.; Meyrignac, O.; Colle, R.; Gasnier, M.; Corruble, E.; Beurnier, A.; Jutant, E.M.; Pham, T.; et al. Post-acute COVID-19 syndrome. Eur. Respir. Rev. 2022, 31, 163. [Google Scholar] [CrossRef]
  11. Wong, J.J.M.; Abbas, Q.; Liauw, F.; Malisie, R.F.; Gan, C.S.; Abid, M.; Efar, P.; Gloriana, J.; Chuah, S.L.; Sultana, R.; et al. Development and validation of a clinical predictive model for severe and critical pediatric COVID-19 infection. PLoS ONE 2022, 17, e0275761. [Google Scholar] [CrossRef]
  12. Anai, M.; Akaike, K.; Iwagoe, H.; Akasaka, T.; Higuchi, T.; Miyazaki, A.; Naito, D.; Tajima, Y.; Takahashi, H.; Komatsu, T.; et al. Decrease in hemoglobin level predicts increased risk for severe respiratory failure in COVID-19 patients with pneumonia. Respir. Investig. 2021, 59, 187–193. [Google Scholar] [CrossRef]
  13. Russo, A.; Tellone, E.; Barreca, D.; Ficarra, S.; Laganà, G. Implication of COVID-19 on Erythrocytes Functionality: Red Blood Cell Biochemical Implications and Morpho-Functional Aspects. Int. J. Mol. Sci. 2022, 23, 2171. [Google Scholar] [CrossRef]
  14. Lopes, D.V.; Neto, F.L.; Marques, L.C.; Lima, R.B.; Brandão, A.A.G.S. Methemoglobinemia and hemolytic anemia after COVID-19 infection without identifiable eliciting drug: A case-report. IDCases 2021, 23, e01013. [Google Scholar] [CrossRef] [PubMed]
  15. Scholkmann, F.; Restin, T.; Ferrari, M.; Quaresima, V. The role of methemoglobin and carboxyhemoglobin in COVID-19: A review. J. Clin. Med. 2020, 10, 50. [Google Scholar] [CrossRef]
  16. Lanser, L.; Burkert, F.R.; Bellmann-Weiler, R.; Schroll, A.; Wildner, S.; Fritsche, G.; Weiss, G. Dynamics in anemia development and dysregulation of iron homeostasis in hospitalized patients with COVID-19. Metabolites 2021, 11, 653. [Google Scholar] [CrossRef]
  17. Bellmann-Weiler, R.; Lanser, L.; Barket, R.; Rangger, L.; Schapfl, A.; Schaber, M.; Fritsche, G.; Wöll, E.; Weiss, G. Prevalence and predictive value of anemia and dysregulated iron homeostasis in patients with COVID-19 infection. J. Clin. Med. 2020, 9, 2429. [Google Scholar] [CrossRef]
  18. Chaparro, C.M.; Parminder, S.S. Anemia epidemiology, pathophysiology, and etiology in low-and middle-income countries. Ann. N. Y. Acad. Sci. 2019, 1450, 15–31. [Google Scholar] [CrossRef] [PubMed]
  19. Cappellini, M.D.; Irene, M. Anemia in Clinical Practice-Definition and Classification: Does Hemoglobin Change with Aging? Semin. Hematol. 2015, 52, 261–269. [Google Scholar] [CrossRef] [PubMed]
  20. Vieth, J.T.; Lane, D.R. Anemia. Emerg. Med. Clin. 2014, 32, 613–628. [Google Scholar] [CrossRef]
  21. Gregg, L.P.; Bossola, M.; Ostrosky-Frid, M.; Hedayati, S.S. Fatigue in CKD: Epidemiology, pathophysiology, and treatment. Clin. J. Am. Soc. Nephrol. 2021, 16, 1445–1455. [Google Scholar] [CrossRef]
  22. Sandler, C.X.; Wyller VB, B.; Moss-Morris, R.; Buchwald, D.; Crawley, E.; Hautvast, J.; Katz, B.Z.; Knoop, H.; Little, P.; Taylor, R.; et al. Long COVID and post-infective fatigue syndrome: A review. In Open Forum Infectious Diseases; Oxford University Press: Oxford, UK, 2021; Volume 8. [Google Scholar]
  23. Maisel, P.; Erika, B.; Norbert, D.-B. Fatigue as the Chief Complaint: Epidemiology, Causes, Diagnosis, and Treatment. Dtsch. Ärzteblatt Int. 2021, 118, 566. [Google Scholar]
  24. Pasini, E.; Corsetti, G.; Romano, C.; Scarabelli, T.M.; Chen-Scarabelli, C.; Saravolatz, L.; Dioguardi, F.S. Serum metabolic profile in patients with long-Covid (PASC) syndrome: Clinical implications. Front. Med. 2021, 1182, 714426. [Google Scholar] [CrossRef]
  25. Fernández-De-Las-Peñas, C.; Ryan-Murua, P.; Rodríguez-Jiménez, J.; Palacios-Ceña, M.; Arendt-Nielsen, L.; Torres-Macho, J. Serological biomarkers at hospital admission are not related to long-term post-COVID fatigue and dyspnea in COVID-19 survivors. Respiration 2022, 101, 658–665. [Google Scholar] [CrossRef] [PubMed]
  26. Baalbaki, N.; Blankestijn, J.M.; Abdel-Aziz, M.I.; de Backer, J.; Bazdar, S.; Beekers, I.; Beijers, R.J.H.C.G.; Bergh, J.P.v.D.; Bloemsma, L.D.; Bogaard, H.J.; et al. Precision Medicine for More Oxygen (P4O2)—Study Design and First Results of the Long COVID-19 Extension. J. Pers. Med. 2023, 13, 1060. [Google Scholar] [CrossRef] [PubMed]
  27. Cardoso, P.; Nielsen, B.B.; Hvidman, L.; Nielsen, J.; Aaby, P. Effect of sublingual misoprostol on severe postpartum haemorrhage in a primary health centre in Guinea-Bissau: Randomised double blind clinical trial. BMJ 2005, 331, 723. [Google Scholar]
  28. Atukunda, E.C.; Mugyenyi, G.R.; Obua, C.; Atuhumuza, E.B.; Musinguzi, N.; Tornes, Y.F.; Agaba, A.G.; Siedner, M.J. Measuring post-partum haemorrhage in low-resource settings: The diagnostic validity of weighed blood loss versus quantitative changes in hemoglobin. PLoS ONE 2016, 11, e0152408. [Google Scholar] [CrossRef] [PubMed]
  29. Hamm, R.F.; Wang, E.; Romanos, A.; O’Rourke, K.; Srinivas, S.K. Implementation of quantification of blood loss does not improve prediction of hemoglobin drop in deliveries with average blood loss. Am. J. Perinatol. 2018, 35, 134–139. [Google Scholar] [CrossRef]
  30. Jerković, A.; Proroković, A.; Matijaca, M.; Katić, A.Ć.; Košta, V.; Mihalj, M.; Dolić, K.; Đogaš, Z.; Vidaković, M.R. Validation of the fatigue severity scale in Croatian population of patients with multiple sclerosis disease: Factor structure, internal consistency, and correlates. Mult. Scler. Relat. Disord. 2022, 58, 103397. [Google Scholar] [CrossRef]
  31. Lerdal, A. Fatigue severity scale. Encyclopedia of Quality of Life and Well-Being Research; Springer International Publishing: Cham, Switzerland, 2021; pp. 1–5. [Google Scholar]
  32. Samprathi, M.; Muralidharan, J. Biomarkers in COVID-19: An up-to-date review. Front. Pediatr. 2021, 8, 607647. [Google Scholar] [CrossRef]
  33. Budhiraja, S.; Indrayan, A.; Das, P.; Dewan, A.; Singh, O.; Nangia, V.; Singh, Y.P.; Pandey, R.; Gupta, A.K.; Gupta, M.; et al. Relative Importance of Various Inflammatory Markers and Their Critical Thresholds for COVID-19 Mortality. medRxiv 2021. [Google Scholar] [CrossRef]
  34. Alisik, M.; Erdogan, U.G.; Ates, M.; Sert, M.A.; Yis, O.M.; Bugdayci, G. Predictive value of immature granulocyte count and other inflammatory parameters for disease severity in COVID-19 patients. Int. J. Med. Biochem. 2021, 4, 143–149. [Google Scholar]
  35. Li, J.; Ji, L. Adjusting multiple testing in multilocus analyses using the eigenvalues of a correlation matrix. Heredity 2005, 95, 221–227. [Google Scholar] [CrossRef] [PubMed]
  36. Wen, S.-H.; Lu, Z.-S. Factors affecting the effective number of tests in genetic association studies: A comparative study of three PCA-based methods. J. Hum. 2011, 56, 6. [Google Scholar] [CrossRef]
  37. Textor, J.; Van der Zander, B.; Gilthorpe, M.S.; Liśkiewicz, M.; Ellison, G.T. Robust causal inference using directed acyclic graphs: The R package ‘dagitty’. Int. J. Epidemiol. 2016, 45, 1887–1894. [Google Scholar] [CrossRef] [PubMed]
  38. Gluba-Brzózka, A.; Franczyk, B.; Olszewski, R.; Rysz, J. The influence of inflammation on anemia in CKD patients. Int. J. Mol. Sci. 2020, 21, 725. [Google Scholar] [CrossRef] [PubMed]
  39. Kim, B.; Kim, G.; Kim, E.; Park, J.; Isobe, T.; Sakae, T.; Oh, S. The a body shape index might be a stronger predictor of chronic kidney disease than BMI in a senior population. Int. J. Environ. Res. Public Health 2021, 18, 12874. [Google Scholar] [CrossRef] [PubMed]
  40. National Kidney Foundation. eGFR Calculator. Available online: https://www.kidney.org/professionals/kdoqi/gfr_calculator (accessed on 27 November 2023).
  41. Abu-Ismail, L.; Taha MJ, J.; Abuawwad, M.T.; Al-Bustanji, Y.; Al-Shami, K.; Nashwan, A.; Yassin, M. COVID-19 and anemia: What do we know so far? Hemoglobin 2023, 47, 122–129. [Google Scholar] [CrossRef] [PubMed]
  42. Lechuga, G.C.; Carlos, M.M.; Salvatore, G.D.-S. Hematological alterations associated with long COVID-19. Front. Physiol. 2023, 14, 1203472. [Google Scholar] [CrossRef]
  43. Sonnweber, T.; Grubwieser, P.; Sahanic, S.; Böhm, A.K.; Pizzini, A.; Luger, A.; Schwabl, C.; Koppelstätter, S.; Kurz, K.; Puchner, B.; et al. 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, 546. [Google Scholar] [CrossRef]
  44. Sobrero, A.; Puglisi, F.; Grossi, F. Fatigue: A main component of anemia symptomatology. In Seminars in Oncology; WB Saunders: Philadelphia, PE, USA, 2001; Volume 28. [Google Scholar]
  45. Prochaska, M.T.; Zhang, H.; Alavi, C.; Meltzer, D.O. Fatigability: A new perspective on and patient-centered outcome measure for patients with anemia. Am. J. Hematol. 2020, 95, E166. [Google Scholar] [CrossRef]
  46. Brownstein, C.G.; Daguenet, E.; Guyotat, D.; Millet, G.Y. Chronic fatigue in myelodysplastic syndromes: Looking beyond anemia. Crit. Rev. Oncol./Hematol. 2020, 154, 103067. [Google Scholar] [CrossRef] [PubMed]
  47. Lim, W.; Hong, S.; Nelesen, R.; Dimsdale, J.E. The association of obesity, cytokine levels, and depressive symptoms with diverse measures of fatigue in healthy subjects. Arch. Intern. Med. 2005, 165, 910–915. [Google Scholar] [CrossRef] [PubMed]
  48. Resnick, H.E.; Carter, E.A.; Aloia, M.; Phillips, B. Cross-sectional relationship of reported fatigue to obesity, diet, and physical activity: Results from the third national health and nutrition examination survey. J. Clin. Sleep Med. 2006, 2, 163–169. [Google Scholar] [CrossRef] [PubMed]
  49. Garcia-Casal, M.N.; Dary, O.; Jefferds, M.E.; Pasricha, S.R. Diagnosing anemia: Challenges selecting methods, addressing underlying causes, and implementing actions at the public health level. Ann. N. Y. Acad. Sci. 2023, 1524, 37–50. [Google Scholar] [CrossRef] [PubMed]
  50. Nemeth, E.; Tomas, G. Anemia of inflammation. Hematol./Oncol. Clin. N. Am. 2014, 28, 671–681. [Google Scholar] [CrossRef] [PubMed]
  51. Hanson, A.L.; Mulè, M.P.; Ruffieux, H.; Mescia, F.; Bergamaschi, L.; Pelly, V.S.; Turner, L.; Kotagiri, P.; Göttgens, B.; Hess, C.; et al. Iron dysregulation and inflammatory stress erythropoiesis associates with long-term outcome of COVID-19. Nat. Immunol. 2024, 25, 471–482. [Google Scholar] [CrossRef] [PubMed]
  52. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Kaushik, A.; Kujawska, M.; Batiha, G.E.-S. Hemolytic anemia in COVID-19. Ann. Hematol. 2022, 101, 1887–1895. [Google Scholar] [CrossRef] [PubMed]
  53. Onoja, A.; von Gerichten, J.; Lewis, H.-M.; Bailey, M.J.; Skene, D.J.; Geifman, N.; Spick, M. Meta-Analysis of COVID-19 Metabolomics Identifies Variations in Robustness of Biomarkers. Int. J. Mol. Sci. 2023, 24, 14371. [Google Scholar] [CrossRef]
  54. Mironova, G.D.; Belosludtseva, N.V.; Ananyan, M.A. Prospects for the use of regulators of oxidative stress in the comprehensive treatment of the novel Coronavirus Disease 2019 (COVID-19) and its complications. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 8585–8591. [Google Scholar]
  55. Revin, V.V.; Gromova, N.V.; Revina, E.S.; Samonova, A.Y.; Tychkov, A.Y.; Bochkareva, S.S.; Moskovkin, A.A.; Kuzmenko, T.P. The Influence of Oxidative Stress and Natural Antioxidants on Morphometric Parameters of Red Blood Cells, the Hemoglobin Oxygen Binding Capacity, and the Activity of Antioxidant Enzymes. BioMed Res. Int. 2019, 2019, 2109269. [Google Scholar] [CrossRef]
  56. Chudow, M.; Beatrice, A. ABC’s of vitamin supplementation in critical illness. J. Pharm. Pract. 2021, 34, 934–942. [Google Scholar] [CrossRef] [PubMed]
  57. Atanasovska, E.; Petrusevska, M.; Zendelovska, D.; Spasovska, K.; Stevanovikj, M.; Kasapinova, K.; Gjorgjievska, K.; Labachevski, N. Vitamin D levels and oxidative stress markers in patients hospitalized with COVID-19. Redox Rep. 2021, 26, 184–189. [Google Scholar] [CrossRef] [PubMed]
  58. Xie, Y.; Choi, T.; Al-Aly, Z. Long-term outcomes following hospital admission for COVID-19 versus seasonal influenza: A cohort study. Lancet Infect. Dis. 2024, 24, 239–255. [Google Scholar] [CrossRef] [PubMed]
  59. Choi, H.S.; Kim, M.H.; Choi, M.G.; Park, J.H.; Chun, E.M. Hematologic abnormalities after COVID-19 vaccination: A large Korean population-based cohort study. medRxiv 2023. [Google Scholar] [CrossRef]
  60. Watanabe, A.; Iwagami, M.; Yasuhara, J.; Takagi, H.; Kuno, T. Protective effect of COVID-19 vaccination against long COVID syndrome: A systematic review and meta-analysis. Vaccine 2023, 41, 1783–1790. [Google Scholar] [CrossRef] [PubMed]
Figure 1. DAG for determining the confounders and effect modifiers in the relationship between hemoglobin levels and fatigue. Figure legend; Biomedicines 12 01234 i001 exposure, Biomedicines 12 01234 i002 outcome, Biomedicines 12 01234 i003 ancestor of outcome, Biomedicines 12 01234 i004 ancestor of exposure and outcome, Biomedicines 12 01234 i005 (potentially) causal path, Biomedicines 12 01234 i006 biasing path.
Figure 1. DAG for determining the confounders and effect modifiers in the relationship between hemoglobin levels and fatigue. Figure legend; Biomedicines 12 01234 i001 exposure, Biomedicines 12 01234 i002 outcome, Biomedicines 12 01234 i003 ancestor of outcome, Biomedicines 12 01234 i004 ancestor of exposure and outcome, Biomedicines 12 01234 i005 (potentially) causal path, Biomedicines 12 01234 i006 biasing path.
Biomedicines 12 01234 g001
Figure 2. Boxplots for assessing the difference in hemoglobin levels between fatigue and non-fatigue cases. Figure legend: O: mild outlier, *: extreme outlier. Footnotes: 1. fatigue status data were missing for 8 cases, and considering the missing data for Hb variables, this graph includes information from 52 cases. 2. The extreme outlier in both baseline and FU figure belongs to the same case.
Figure 2. Boxplots for assessing the difference in hemoglobin levels between fatigue and non-fatigue cases. Figure legend: O: mild outlier, *: extreme outlier. Footnotes: 1. fatigue status data were missing for 8 cases, and considering the missing data for Hb variables, this graph includes information from 52 cases. 2. The extreme outlier in both baseline and FU figure belongs to the same case.
Biomedicines 12 01234 g002
Table 1. Characteristics of the study population.
Table 1. Characteristics of the study population.
Population CharacteristicsTotal (N = 95)p-Value £Female (N = 47)Male (N = 48)
Demographic characteristics
Age (mean, SD)54.2 (6.2)0.76554.3 (6.1)54.0 (6.3)
Ethnicity (N, %)African 8 (9.4%)0.0775 (1.6%)3 (7.0%)
Asian 3 (3.5%) 2 (4.7%)1 (2.3%)
Caucasian66 (76.7%) 33 (76.7%)33 (76.7%)
Latin-American 3 (3.5%) 3 (7.0%)0 (0.0%)
Others6 (7.0%) 0 (0.0%)6 (14.0%)
Clinical characteristics
Suffering from any comorbidities (N, %)85 (89.5%)0.17040 (85.1%)45 (93.8%)
BMI (mean, SD)30.39 (5.3)0.00232.15 (5.4%)28.85 (4.7)
Dominant SARS-CoV-2 variant at the time of infection (N, %)Alpha 43 (45.3%)0.27823 (48.9%)20 (41.7%)
Delta41 (43.2%) 21 (44.7%)20 (41.7%)
Omicron11 (11.6%) 3 (6.4%)8 (16.7%)
Hospitalized (N, %)85 (89.5%)0.17040 (85.1%)45 (93.8%)
Hospital stay (days) (median, percentile 25–75) *8.00 (4.0–15.2)0.0838.00 (3.0–13.0)10.00 (5.0–21.0)
ICU admission (N, %)27 (28.7%)0.409 (19.1%)18 (38.3%)
ICU stay (days) (median, percentile 25–75) *0.00 (0.0–4.0)0.0440.00 (0.0–0.0)0.00 (0.0–8.0)
One dosage of SARS-CoV-2 vaccination66 (69.5%)0.77132 (68.1%)34 (70.8%)
Two dosages of SARS-CoV-2 vaccination41 (43.2%)0.26123 (48.9%)18 (37.5%)
WHO severity index (N, %)Mild10 (10.8%)0.2407 (15.2%)3 (6.4%)
Moderate57 (61.3%) 30 (65.2%)27 (57.4%)
Severe 26 (28.0%) 9 (19.6%)17 (36.2%)
Average FSS during the 3–6 months visit (median, percentile 25–75) ¥5.6 (4.1–6.3)0.0265.8 (4.7–6.4)5.3 (3.0–6.3)
Fatigue (Average FSS ≥ 4) during the 3–6 months visit (N, %) ¥ 0.116
Yes66 (69.5%) 35 (74.5%)31 (68.9%)
No21 (22.1%) 7 (14.9%)14 (29.2%)
Missing8 (8.4%) 5 (10.6%)3 (63%)
* The number of days spent in the ICU or hospital for the participants who were not admitted were counted as 0. ¥ Fatigue data were missing for eight (five female and three male) participants. £ p-value for comparing variables between men and women.
Table 2. Hemoglobin level status and the prevalence of anemia in the P4O2 COVID-19 study.
Table 2. Hemoglobin level status and the prevalence of anemia in the P4O2 COVID-19 study.
Hb and Anemia StatusTotalFemaleMalep-ValueExperiencing Fatigue 3–6 Months after Infection ¶No Fatigue 3–6 Months after Infection p-Value π
Total number of participants954748---6621---
Hb ◊ level during the acute phase (mmol/L) (mean, SD) (n = 84)8.61 (0.99)8.35 (0.86)8.84 (1.05)0.0238.53 (0.99)8.89 (1.06)0.171
Hb level during the 3–6 months visit (mmol/L) (mean, SD) (n = 68)8.70 (0.98)8.36 (0.87)9.04 (1.00)0.0048.54 (0.99)9.24 (0.99)0.046
Hb level change (mmol/L) (mean, SD) ¥ (n = 59)0.30 (0.74)0.18 (0.80)0.42 (0.67)0.2220.19 (0.73)0.68 (0.69)0.069
Clinically notable (10%) Hb change (N, %)Hb decrease ‡8 (8.4%)4 (10.50%)4 (9.50%)0.8816 (10.90%)1 (5.90%)1.000
Hb increase Ⱡ15 (23.7%)6 (15.80%)9 (21.40%)0.51910 (18.20%)3 (17.6%)1.000
Hb level change µ (N, %)Hb decrease *17 (28.8%)10 (34.50%)7 (23.30%)0.34414 (32.20%)1 (10.00%)0.249
Hb increase €39 (66.1%)19 (65.50%)20 (66.70%)0.92626 (60.50%)9 (90.00%)0.137
Anemia during the acute phase (N, %) £ (n = 84)19 (22.61%)5 (12.80%)14 (31.10%)0.04613 (23.2%)4 (20%)1.000
Anemia during the 3–6 months visit (N, %) £ (n = 68)11 (16.41%)5 (13.90%)6 (18.80%)0.58710 (19.2%)1 (10%)0.674
Footnotes: ¶ fatigue data were missing for eight cases. π the p-values are related to the comparisons that have been performed for Hb levels/anemia status, between patients experiencing fatigue 3–6 months after infection and those who did not. ◊ normal Hb range is >7.5 in women and >8.5 in males. ¥ follow-up Hb–baseline Hb (difference between baseline and follow-up Hb; there were missing data for 36 (37.90%) cases. ‡ cases with follow-up Hb–baseline Hb < 0.10 × baseline Hb. Ⱡ cases with baseline Hb–follow-up Hb < 0.10 × baseline Hb. * cases with follow-up Hb < baseline Hb. € cases with follow-up Hb > baseline Hb. £ Hb level less than 7.5 mmol/L in women and less than 8.5 mmol/L in men. µ For three cases the baseline Hb was exactly the same as the FH Hb and they had no Hb change. p-values showing statistically significant difference between compared groups have been marked in bold font.
Table 3. Correlations between hemoglobin measures and inflammatory indices.
Table 3. Correlations between hemoglobin measures and inflammatory indices.
VariableMedian (Percentile 25–75)cc * with Hb ChangeN Ⱡcc with Baseline HbNcc with FU HbN
Baseline CRP91.00 (31.40–152.00)0.031590.01884−0.01250
FU CRP2.40 (1.85–6.30)−0.00249NA ‡50−0.26554
Baseline LDH365.00 (272.00–516.00)0.082460.012640.08946
FU LDH200.00 (170.00–244.75)0.00251NA51−0.12655
Baseline IG0.03 (0.01–0.08)0.072300.108360.33130
FU IG0.02 (0.01–0.03)0.07525NA25−0.00628
Baseline CPK102.00 (57.00–298.00)0.070340.05352−0.00334
FU CPK72.00 (66.00–100.00)0.03331NA310.08434
Baseline NLR5.05 (3.07–7.14)0.24246−0.071550.13346
FU NLR1.63 (1.29–2.13)0.29436NA45−0.01849
Baseline D-dimer0.75 (0.54–2.20)0.20655−0.09380−0.03055
Abbreviations: CRP: C-reactive protein, LDH: Lactate Dehydrogenase, IG: Immature Granulocyte, CPK: creatine phosphokinase, NLR: Neutrophil/Lymphocyte ratio. * Correlation coefficient. Ⱡ number of cases in which correlation analysis between them and Hb change has been performed. ‡ NA: not applicable; our research hypothesis did not consider the relationship between FU inflammatory biomarkers and baseline Hb, since the outcome (Hb concentrations) would precede the exposures in this analysis.
Table 4. Associations of hemoglobin levels and anemia with fatigue 3–6 months after SARS-CoV-2 infection (average FSS ≥ 4).
Table 4. Associations of hemoglobin levels and anemia with fatigue 3–6 months after SARS-CoV-2 infection (average FSS ≥ 4).
UnadjustedAdjusted *
OR ‡ (95% CI)OR (95% CI)
Baseline Hb levels0.676 (0.386–1.184)0.760 (0.430–1.342)
Follow-up Hb levels0.370 (0.141–0.969)0.350 (0.120–1.015)
Hb change0.380 (0.133–1.081)0.377 (0.130–1.091)
Baseline anemia1.209 (0.343–4.259)1.557 (0.420–5.779)
Follow-up anemia2.143 (0.243–18.919)2.246 (0.250–20.203)
* Adjusted for age and sex. ‡ Per 1 mmol/L increase in Hb levels.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bazdar, S.; Bloemsma, L.D.; Baalbaki, N.; Blankestijn, J.M.; Cornelissen, M.E.B.; Beijers, R.J.H.C.G.; Sondermeijer, B.M.; van Wijck, Y.; Downward, G.S.; Maitland-van der Zee, A.H., on behalf of the P4O2 Consortium. Hemoglobin and Its Relationship with Fatigue in Long-COVID Patients Three to Six Months after SARS-CoV-2 Infection. Biomedicines 2024, 12, 1234. https://doi.org/10.3390/biomedicines12061234

AMA Style

Bazdar S, Bloemsma LD, Baalbaki N, Blankestijn JM, Cornelissen MEB, Beijers RJHCG, Sondermeijer BM, van Wijck Y, Downward GS, Maitland-van der Zee AH on behalf of the P4O2 Consortium. Hemoglobin and Its Relationship with Fatigue in Long-COVID Patients Three to Six Months after SARS-CoV-2 Infection. Biomedicines. 2024; 12(6):1234. https://doi.org/10.3390/biomedicines12061234

Chicago/Turabian Style

Bazdar, Somayeh, Lizan D. Bloemsma, Nadia Baalbaki, Jelle M. Blankestijn, Merel E. B. Cornelissen, Rosanne J. H. C. G. Beijers, Brigitte M. Sondermeijer, Yolanda van Wijck, George S. Downward, and Anke H. Maitland-van der Zee on behalf of the P4O2 Consortium. 2024. "Hemoglobin and Its Relationship with Fatigue in Long-COVID Patients Three to Six Months after SARS-CoV-2 Infection" Biomedicines 12, no. 6: 1234. https://doi.org/10.3390/biomedicines12061234

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop