1
|
Tizazu AM, Gize A, Ali S. Age influences blood cell-based immune deregulation antibody response and unfavorable clinical outcomes in COVID-19 patients. Sci Rep 2025; 15:17431. [PMID: 40394243 PMCID: PMC12092784 DOI: 10.1038/s41598-025-95722-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/24/2025] [Indexed: 05/22/2025] Open
Abstract
COVID-19 is caused by SARS-CoV-2 and has a diverse spectrum of clinical presentations ranging from asymptomatic cases to severe and critical cases that result in the death of the patient. Alongside the viral factors host factors like Age, deregulation of the immune response and presence of comorbidity determine the patient's outcome. Here we sought to assess the impact of age on natural antibody response, CBC-based inflammatory markers, and outcome of COVID-19 patients. We divided the participants into three groups, young (≤ 35 years), middle-aged (40-60 years), and old (≥ 65 years) patients and collected and analyzed sociodemographic, clinical, and laboratory parameters. We found that elderly patients showed higher (P < 0.05) levels of inflammation like increased neutrophil percentages, NLR, lymphopenia, and low Hgb levels, compared to middle-aged and young patients. Interestingly these markers were also associated with mortality of COVID-19 patients. On the other hand, no significant difference was observed in ion concentration, lipid profile, and coagulation test between the three age groups. We also found that elderly patients showed significantly (P < 0.05) decreased levels of natural antibody response to SARS-CoV-2 infection compared with the two groups. Lastly, we assessed the effect of dexamethasone treatment, even if statistically not significant (P > 0.05) we observed a positive trend among patients under dexamethasone in the aspect of decreasing inflammatory markers. To conclude we showed that SARS-CoV-2 is characterized by an age-dependent deregulation of inflammatory markers that are associated with mortality among COVID-19 patients.
Collapse
Affiliation(s)
- Anteneh Mehari Tizazu
- Department of Microbiology, Immunology and Parasitology, School of Medicine, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia.
| | - Addisu Gize
- Department of Microbiology, Immunology and Parasitology, School of Medicine, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
- CIHLMU Center for International Health, LMU University Hospital, LMU Munich, Germany
| | - Solomon Ali
- Department of Microbiology, Immunology and Parasitology, School of Medicine, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia
| |
Collapse
|
2
|
Ayyad M, Abu Alya W, Barabrah AM, Darawish SM, AlHabil Y, MohammedAli M, Nabilsi MZ, Asad D, Ayasa LA, Matassa D. Autoimmune hemolytic anemia in COVID-19 patients: A systematic review of 105 cases on clinical characteristics and outcomes. Clin Immunol 2025; 277:110512. [PMID: 40348041 DOI: 10.1016/j.clim.2025.110512] [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: 12/11/2024] [Revised: 03/29/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND COVID-19 has been linked to autoimmune hemolytic anemia (AIHA), a rare but serious condition causing red blood cell destruction. This systematic review examines the clinical characteristics, management, and outcomes of AIHA in COVID-19 patients. METHODS A systematic search of PubMed, CINAHL, and Scopus identified 85 studies encompassing 105 patients. Data on demographics, clinical features, and treatment outcomes were extracted. RESULTS Of 1402 articles, 85 met inclusion criteria. Most patients were male (54.3 %) with a mean age of 50.6 years, predominantly from Asia (83.5 %). Cold agglutinin AIHA was most common (48.2 %). Presenting symptoms included fatigue, dyspnea, and fever. Steroids were the most effective treatment, used in 95 % of recovered cases. Mortality was 14.3 %, with 26.7 % of deaths directly related to AIHA. CONCLUSIONS COVID-19 is associated with AIHA, often presenting with non-specific symptoms. Early recognition and prompt steroid therapy are critical for improving outcomes. Further research is needed to guide management.
Collapse
Affiliation(s)
- Mohammed Ayyad
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.
| | - Walaa Abu Alya
- Department of Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH, USA
| | | | | | - Yazan AlHabil
- Department of Medicine, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestine
| | | | | | - Diya Asad
- Faculty of Medicine, Al-Quds University, Jerusalem, Palestine
| | - Laith A Ayasa
- Faculty of Medicine, Al-Quds University, Jerusalem, Palestine
| | - Daniel Matassa
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
3
|
Rashid W, Paduchuri V, Chandy J, Hodgson J, Camporesi E. Evaluating Predictive Value of Plasma Free Hemoglobin (PFH) in ECMO for COVID-19, Non-COVID-19 Pulmonary, and Cardiac Patients. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:801. [PMID: 40428759 PMCID: PMC12112789 DOI: 10.3390/medicina61050801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/31/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Extracorporeal membrane oxygenation (ECMO) can support patients with severe cardiopulmonary failure, but it poses risks such as hemolysis, leading to complications. Plasma-free hemoglobin (PFH) is a hemolysis biomarker, with elevated levels linked to mortality. This study evaluates PFH and ECMO survival in COVID-19, non-COVID-19 pulmonary, and cardiac patients, focusing on late PFH spikes. Materials and Methods: We retrospectively analyzed 122 ECMO patients treated at our tertiary hospital (January 2020-December 2021). Patients were categorized by indication: post-COVID-19, non-COVID-19 pulmonary, or cardiac. We classified patients as Expired (died during ECMO or ≤30 days post-ECMO) or Survived (>30 days post-ECMO). Data included demographics, ECMO duration, and PFH values at 24 h and during the last 3 and 5 ECMO days. Groups were compared using two-tailed t-tests, with p < 0.05 indicating significance. Results: COVID-19 patients survived after significantly longer ECMO duration than non-COVID-19 pulmonary and cardiac patients. Expired COVID-19 patients had higher PFH values during the last 3 and 5 days of ECMO compared to survivors. Cardiac patients had the highest overall PFH levels regardless of mortality. No significant differences in PFH trends were observed between non-COVID-19 pulmonary and cardiac patients. Conclusions: Late PFH spikes correlated with mortality in COVID-19 patients, suggesting the utility of measuring late PFH spikes in ECMO management. Additionally, COVID-19 pulmonary patients survived when undergoing ECMO significantly longer than both groups, while VA ECMO was more prone to hemolysis. However, technical cannulation differences and frequent use of an Impella pump in cardiac patients may increase blood stress and PFH values.
Collapse
Affiliation(s)
- Wasiq Rashid
- Morsani College of Medicine, University of South Florida, Tampa, FL 33606, USA; (W.R.); (V.P.)
| | - Varshith Paduchuri
- Morsani College of Medicine, University of South Florida, Tampa, FL 33606, USA; (W.R.); (V.P.)
| | - Joby Chandy
- Department of Anesthesiology and Perioperative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33606, USA; (J.C.); (J.H.)
- TeamHEALTH Anesthesia, Tampa General Hospital, Tampa, FL 33606, USA
| | - John Hodgson
- Department of Anesthesiology and Perioperative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33606, USA; (J.C.); (J.H.)
- TeamHEALTH Anesthesia, Tampa General Hospital, Tampa, FL 33606, USA
| | - Enrico Camporesi
- Department of Anesthesiology and Perioperative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33606, USA; (J.C.); (J.H.)
- TeamHEALTH Anesthesia, Tampa General Hospital, Tampa, FL 33606, USA
| |
Collapse
|
4
|
Cahuapaza-Gutierrez NL, Calderon-Hernandez CC, Pajuelo-Vasquez R, Coronado-Quispe HY, Altamirano-Molina M, Runzer-Colmenares FM, Villavicencio-Escudero TV. New-onset hematologic disorders following COVID-19 vaccination: a systematic review. Clin Exp Vaccine Res 2025; 14:169-184. [PMID: 40321788 PMCID: PMC12046088 DOI: 10.7774/cevr.2025.14.e20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 02/25/2025] [Indexed: 05/08/2025] Open
Abstract
Purpose Coronavirus disease 2019 (COVID-19) vaccination reduced morbimortality rates due to severe acute respiratory syndrome coronavirus 2 infection worldwide. However, various complications have been reported, including hematologic disorders. Materials and Methods We conducted a systematic review to synthesize and analyze the current available evidence on the development of hematological disorders associated with COVID-19 vaccination. Results A total of 227 patients were reported in the papers that were selected to be included. There was a slight predominance of females (n=114, 50.22%) compared to males (n=113, 49.78%), and the calculated mean age was 54.86±18.94 years. The most frequently reported hematological disorders were Immune thrombocytopenic purpura (n=58, 25.55%), followed by thrombotic thrombocytopenic purpura (n=38, 16.74%). The less frequently recorded cases were acquired factor XIII/13 deficiency (n=2, 0.88%) and pernicious anemia (n=2, 0.88%). Messenger RNA (mRNA)-based COVID-19 vaccines, including Pfizer BioNTech 162b2 (n=106, 46.70%), Moderna mRNA 127-3 (n = 42, 18.50%), and the Bivalent vaccine (n = 1, 0.44%), were the most prevalent (n=150, 66.08%). Most cases developed after the first dose (n=120, 52.86%). In most cases, patient outcomes were favorable (n=175, 77.09%), but there were significant mortality cases (n=23, 10.13%). Conclusion Our findings suggest close monitoring of patients who receive the first dose with mRNA technology vaccines, regardless of sex, especially in adults, as they appear more vulnerable to developing hematologic disorders. Trial Registration PROSPERO Identifier: CRD42023452589.
Collapse
Affiliation(s)
| | | | - Renzo Pajuelo-Vasquez
- Universidad Científica del Sur, Lima, Perú
- CHANGE Research Working Group, Universidad Científica del Sur, Lima, Perú
| | | | - Milagros Altamirano-Molina
- Facultad de Medicina, Universidad Nacional Mayor de San Marcos, Lima, Perú
- Guillermo Almenara Irigoyen Hospital, EsSalud, Lima, Perú
| | | | | |
Collapse
|
5
|
Kim DW, Jeong IH, Son YK, Rha SH, Chung YS. A Case of Autoimmune Hemolytic Anemia Following COVID-19 Infection Accompanied by Acute Tubulointerstitial Nephritis in a Kidney Transplant Recipient. Transplant Proc 2025; 57:109-115. [PMID: 39638714 DOI: 10.1016/j.transproceed.2024.10.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/16/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Acute tubular injury is one of the main causes of acute tubular injury (acute kidney injury ) in patients with COVID-19 infection. Autoimmune hemolytic anemia (AIHA) is also one of the autoimmune complications of COVID-19. However, AIHA accompanied by acute tubulointerstitial nephritis (ATIN) caused by SARS-CoV-2 is rarely reported. Here, we report a kidney transplant recipient who underwent graftectomy owing to ATIN accompanied by AIHA, possibly exacerbated by COVID-19 infection. CASE PRESENTATION A 32-year-old male renal allograft recipient owing to immunoglobulin A nephropathy visited the emergency department owing to dyspnea and general weakness. Three weeks earlier, the patient had been transplanted with deceased-donor kidney with full HLA-A, -B, -DR match, and had been on tacrolimus, prednisolone, and mycophenolate since then. At the time of the visit, laboratory findings revealed hemoglobin of 2.4 g/dL, reticulocyte of 21.7%, total bilirubin of 1.9 mg/dL, direct bilirubin of 0.3 mg/dL, lactate dehydrogenase of 946 U/L, haptoglobin of <10 mg/dL, and severe red cell agglutination on peripheral blood smear, which suggested AIHA. In addition, his SARS-CoV-2 real-time polymerase chain reaction test was positive. During steroid treatment for AIHA, a sudden decrease in urine volume, estimated glomerular filtration rate (from 64.9 to 35.1 mL/min/1.73 m2) and increase of creatinine (from 1.42 to 2.36 mg/dL) indicated renal function deterioration, so steroid was increased to 500 mg. On the third day of renal function deterioration, dialysis was started owing to anuria and fluid retention. On renal biopsy, C4d was absent; however, ATIN with eosinophilic infiltration was observed. On renal ultrasound examination, a severely enlarged kidney with edema was observed. At the same time, the patient had a high fever with increased C-reactive protein and procalcitonin. Graftectomy was performed to prevent secondary infection. The postgraftectomy renal biopsy showed renal parenchymal and hilar inflammatory change, endotheliitis, and lymphocytic infiltration of peripheral nerve fibers. After graftectomy, dialysis was maintained and AIHA had ameliorated. CONCLUSION The patient had to have his allografted kidney removed owing to ATIN possibly caused by COVID-19 infection. Acute kidney injury caused by SARS-CoV-2 can be either by direct viral infection or as consequence of immunological response. The exact immunological mechanism of AIHA secondary to COVID-19 infection remains to be elucidated.
Collapse
Affiliation(s)
- Dong Wook Kim
- Division of Nephrology, Department of Internal Medicine, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Republic of Korea
| | - In Hwa Jeong
- Department of Laboratory Medicine, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Young Ki Son
- Division of Nephrology, Department of Internal Medicine, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Seo Hee Rha
- Department of Pathology, Dong-A University Hospital Dong-A University College of Medicine, Busan, Republic of Korea
| | - Young Soo Chung
- Department of Surgery, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Republic of Korea.
| |
Collapse
|
6
|
Sato K, Nakamura Y, Hara R, Kayama R, Sunohara S, Okajima S, Kazama S, Komaba W, Ishikawa R, Sumi M, Kobayashi H. Adult-onset severe paroxysmal cold hemoglobinuria after COVID-19 successfully treated with sutimlimab. Int J Hematol 2024; 120:735-742. [PMID: 39304597 DOI: 10.1007/s12185-024-03851-8] [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: 02/22/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Paroxysmal cold hemoglobinuria (PCH) is a form of cold autoimmune hemolytic anemia characterized by the presence of the Donath-Landsteiner antibody, which triggers complement-mediated intravascular hemolysis when the body temperature changes from cold to warm. PCH occurs primarily in children as a rare, self-limiting disease following viral infections. In contrast, adult-onset PCH is very rare and associated with a diverse range of underlying conditions, which complicates its management and treatment. We describe a case of adult-onset PCH following COVID-19, effectively managed with a single dose of sutimlimab, a selective classical complement pathway inhibitor. This intervention was performed during a life-threatening hemolytic crisis, at a time requiring swift decision-making when specific tests to differentiate from other hemolytic anemias were not readily available. This case illustrates the potential of using a single dose of sutimlimab to manage life-threatening hemolytic crises in PCH, highlighting the significance of inhibiting the classical complement pathway.
Collapse
Affiliation(s)
- Keijiro Sato
- Department of Hematology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano City, Nagano, 380-8582, Japan.
| | - Yusuke Nakamura
- Department of Hematology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano City, Nagano, 380-8582, Japan
| | - Ryosuke Hara
- Department of Rheumatology, Minami Nagano Medical Center, Shinonoi General Hospital, Nagano, Japan
| | - Ryuhei Kayama
- Department of Rheumatology, Minami Nagano Medical Center, Shinonoi General Hospital, Nagano, Japan
| | - Shunya Sunohara
- Department of Clinical Laboratory, Nagano Red Cross Hospital, Nagano, Japan
| | - Sayaka Okajima
- Laboratory Department, 1st Laboratory Division, Kanto-Koshinetsu Block Blood Center, Japanese Red Cross Society, Tokyo, Japan
| | - Shintaro Kazama
- Department of Hematology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano City, Nagano, 380-8582, Japan
| | - Wataru Komaba
- Department of Hematology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano City, Nagano, 380-8582, Japan
| | - Ryuto Ishikawa
- Department of Hematology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano City, Nagano, 380-8582, Japan
| | - Masahiko Sumi
- Department of Hematology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano City, Nagano, 380-8582, Japan
| | - Hikaru Kobayashi
- Department of Hematology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano City, Nagano, 380-8582, Japan
| |
Collapse
|
7
|
Toro A, Arévalo AP, Pereira-Gómez M, Sabater A, Zizzi EA, Perbolianachis P, Pascual G, Lage-Vickers S, Pórfido JL, Achinelli I, Seniuk R, Bizzotto J, Sanchis P, Olivera A, Leyva A, Moreno P, Costábile A, Fajardo A, Carrión F, Fló M, Olivero-Deibe N, Rodriguez F, Nin N, Anselmino N, Labanca E, Vazquez E, Cotignola J, Alonso DF, Valacco MP, Marti M, Gentile F, Cherkasov A, Crispo M, Moratorio G, Gueron G. Blood matters: the hematological signatures of Coronavirus infection. Cell Death Dis 2024; 15:863. [PMID: 39609423 PMCID: PMC11605097 DOI: 10.1038/s41419-024-07247-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024]
Abstract
Recent developments have broadened our perception of SARS-CoV-2, indicating its capability to affect the body systemically beyond its initial recognition as a mere respiratory pathogen. However, the pathways of its widespread are not well understood. Employing a dual-modality approach, we integrated findings from a Murine Hepatitis Virus (MHV) infection model with corroborative clinical data to investigate the pervasive reach of Coronaviruses. The novel presence of viral particles within red blood cells (RBCs) was demonstrated via high-resolution transmission electron microscopy, with computational modeling elucidating a potential heme-mediated viral entry mechanism via Spike protein affinity. Our data affirm viral localization in RBCs, suggesting heme moieties as facilitators for cellular invasion. Exacerbation of MHV pathology upon hemin administration, contrasted with chloroquine-mediated amelioration, underscoring a heme-centric pathway in disease progression. These observations extend the paradigm of Coronavirus pathogenicity to include hemoprotein interactions. This study casts new light on the systemic invasion capabilities of Coronaviruses, linking RBC hemoproteins with viral virulence. The modulation of disease severity through heme-interacting agents heralds a promising avenue for COVID-19 therapeutics. Our findings propose a paradigm shift in the treatment approach, leveraging the virus-heme interplay as a strategic hinge for intervention.
Collapse
Affiliation(s)
- Ayelen Toro
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Ana P Arévalo
- Unidad de Biotecnología en Animales de Laboratorio, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Marianoel Pereira-Gómez
- Laboratorio de Virología Molecular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Evolución Experimental de Virus, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Agustina Sabater
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires, Argentina
| | - Eric A Zizzi
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Paula Perbolianachis
- Laboratorio de Virología Molecular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Evolución Experimental de Virus, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Gaston Pascual
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sofia Lage-Vickers
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge L Pórfido
- Unidad de Biotecnología en Animales de Laboratorio, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Ines Achinelli
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rocio Seniuk
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Bizzotto
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires, Argentina
| | - Pablo Sanchis
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires, Argentina
| | - Alvaro Olivera
- Laboratorio de Alta Resolución, Departamento de Desarrollo Tecnológico, Centro Universitario Regional Este (CURE), Universidad de la República, Rocha, Uruguay
| | - Alejandro Leyva
- Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Pilar Moreno
- Laboratorio de Virología Molecular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Evolución Experimental de Virus, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Alicia Costábile
- Laboratorio de Virología Molecular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Evolución Experimental de Virus, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Centro de Innovación en Vigilancia Epidemiológica, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Alvaro Fajardo
- Laboratorio de Virología Molecular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Evolución Experimental de Virus, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Federico Carrión
- Laboratorio de Inmunovirología, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Martín Fló
- Laboratorio de Inmunovirología, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | | | - Fernando Rodriguez
- Unidad de Cuidados Intensivos, Hospital Español "Juan José Crottoggini", Administración de Servicios de Salud del Estado, Montevideo, Uruguay
| | - Nicolas Nin
- Unidad de Cuidados Intensivos, Hospital Español "Juan José Crottoggini", Administración de Servicios de Salud del Estado, Montevideo, Uruguay
| | - Nicolas Anselmino
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elba Vazquez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Javier Cotignola
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniel F Alonso
- Centro de Oncología Molecular y Traslacional y Plataforma de Servicios Biotecnológicos, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Maria P Valacco
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marcelo Marti
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Francesco Gentile
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, Ottawa, ON, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Martina Crispo
- Unidad de Biotecnología en Animales de Laboratorio, Institut Pasteur de Montevideo, Montevideo, Uruguay.
| | - Gonzalo Moratorio
- Laboratorio de Virología Molecular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay.
- Laboratorio de Evolución Experimental de Virus, Institut Pasteur de Montevideo, Montevideo, Uruguay.
| | - Geraldine Gueron
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Binsfeld M, Devey A, Gothot A. Transfusion support and pre-transfusion testing in autoimmune haemolytic anaemia. Vox Sang 2024; 119:1029-1038. [PMID: 38922929 DOI: 10.1111/vox.13699] [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: 01/25/2024] [Revised: 03/29/2024] [Accepted: 04/19/2024] [Indexed: 06/28/2024]
Abstract
Autoimmune haemolytic anaemia (AIHA) is characterized by an increased destruction of red blood cells due to immune dysfunction and auto-antibody production. Clinical manifestations are mainly related to anaemia, which can become life-threatening in case of acute haemolysis. Aiming at counterbalancing severe anaemia, supportive treatments for these patients frequently include transfusions. Unfortunately, free serum auto-antibodies greatly interfere in pre-transfusion testing, and the identification of compatible red blood cell units for AIHA patients can be challenging or even impossible. Problems faced in pre-transfusion testing often lead to delay or abandonment of transfusions for AIHA patients. In this review, we discuss publications concerning global transfusion management in AIHA, with a focus on pre-transfusion testing, and practical clues to manage the selection of transfusion units for these patients. Depending on the degree of transfusion emergency, we propose an algorithm for the selection and laboratory testing of units to be transfused to AIHA patients.
Collapse
Affiliation(s)
- Marilène Binsfeld
- Immuno-Hematology Laboratory and Blood Blank, Department of Clinical Biology, University Hospital of Liège (B35), Liège, Belgium
| | - Anaïs Devey
- Immuno-Hematology Laboratory and Blood Blank, Department of Clinical Biology, University Hospital of Liège (B35), Liège, Belgium
| | - André Gothot
- Immuno-Hematology Laboratory and Blood Blank, Department of Clinical Biology, University Hospital of Liège (B35), Liège, Belgium
| |
Collapse
|
9
|
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
|
10
|
Sayed AA. Back to Basics: The Diagnostic Value of a Complete Blood Count in the Clinical Management of COVID-19. Diagnostics (Basel) 2024; 14:1933. [PMID: 39272717 PMCID: PMC11393994 DOI: 10.3390/diagnostics14171933] [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: 08/02/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Since the beginning of the COVID-19 pandemic, scientists have struggled significantly to understand the complexity of COVID-19 pathophysiology. COVID-19 has demonstrated a notoriously unpredictable clinical course. This unpredictability constituted a significant obstacle to clinicians in predicting the disease course among COVID-19 patients, more specifically, in predicting who would develop severe cases and possibly die from the infection. This brief report aims to assess the diagnostic value of using a complete blood count (CBC) and applying high-dimensional analysis, i.e., principal component analysis (PCA), on it to differentiate between patients with mild and severe COVID-19 infection. The data of 855 patients were retrieved from multiple centres in Saudi Arabia. Descriptive statistics, such as counts, percentages, and medians (interquartile ranges) were used to describe patients' characteristics and CBC parameters. Analytical statistics, such as the Mann-Whitney U test, were used to compare between survivors and non-survivors. PCA was applied using the CBC parameters, and the results were compared between survivors and non-survivors. Patients in this study had a median age of 41, with an almost equal ratio of men to women. Most participants were Saudis, and non-survivors were 13.22% of the total cohort. The median values of all CBC indices were within reference ranges; however, some statistically significant differences were observed between survivors and non-survivors. Non-survivors had lower hemoglobin levels and lower hematocrit, lymphocyte, and eosinophil counts but higher WBC and neutrophil counts compared to survivors. PCA on the CBC results of survivors yielded a significantly different profile than non-survivors, indicating the possibility of its use in the context of COVID-19. The diagnostic value of CBC in the clinical management of COVID-19 should be utilized in clinical guidelines for managing COVID-19 cases.
Collapse
Affiliation(s)
- Anwar A Sayed
- Department of Basic Medical Sciences, College of Medicine, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
11
|
Podzolkov VI, Vetluzhskaya MV, Medvedev ID, Abramova AA, Kislenko GA. [Dyspnea in post-COVID-19 patients: A review]. TERAPEVT ARKH 2024; 96:706-712. [PMID: 39106515 DOI: 10.26442/00403660.2024.07.202785] [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: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 08/09/2024]
Abstract
New coronavirus infection may lead to long-term consequences, particularly to post-COVID syndrome, one of the most common manifestations of which is dyspnea. Post-COVID-19 shortness of breath may persist from one to several months and even years that results in low quality of life of patients. The review highlights possible risk factors and causes of dyspnea in post-COVID period such as lung damage, cardiovascular pathology, hyperventilation syndrome, dysfunction of the autonomic nervous system, detraining, anemia, etc. The authors present data about COVID-19-associated causes of dyspnea and severity of acute COVID-19. The review emphasizes the importance of a multidisciplinary approach to the diagnosis and treatment of patients with shortness of breath in post-COVID-19 period.
Collapse
Affiliation(s)
- V I Podzolkov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - M V Vetluzhskaya
- Sechenov First Moscow State Medical University (Sechenov University)
| | - I D Medvedev
- Sechenov First Moscow State Medical University (Sechenov University)
| | - A A Abramova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - G A Kislenko
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
12
|
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
|
13
|
Wen Q, Guo Z, Zhang XH, Xu LP, Wang Y, Yan CH, Chen H, Chen YH, Han W, Wang FR, Sun YQ, Huang XJ, Mo XD. COVID-19 was associated with the complications after allogeneic hematopoietic stem cell transplantation. Sci Rep 2024; 14:11778. [PMID: 38782966 PMCID: PMC11116404 DOI: 10.1038/s41598-024-62731-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 05/21/2024] [Indexed: 05/25/2024] Open
Abstract
We aimed to identify the severity and duration of COVID-19 infection on complications after allo-HSCT. Enrolled 179 hospitalized patients with COVID-19 were categorized into long-term infection (> 18 days, n = 90) or short-term infection group (≤ 18 days, n = 89) according to the median duration of COVID-19. The severity of COVID-19 was categorized as asymptomatic infection, mild, moderate, severe, and critical illness according to guidelines of National Institutes of Health. Particularly, severe illness and critical illness were classified as serious infection. Asymptomatic infection, mild illness and moderate illness were classified as non-serious infection. The 150-day probabilities of poor graft function (PGF), cytomegalovirus (CMV) pneumonia and non-relapse mortality (NRM) were significantly higher in long-term infection group. The 150-day probabilities of CMV pneumonia and NRM after COVID-19 were higher in serious infection group. The 150-day probabilities of overall survival (OS) was significantly lower in long-term and serious infection group. In multivariable analysis, the severity of COVID-19 was associated with NRM and OS, and the duration of COVID-19 was associated with PGF. In summary, our data reported that the severity and duration of COVID-19 were associated with several complications and contribute to poor outcomes after allo-HSCT.
Collapse
Affiliation(s)
- Qi Wen
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Ze Guo
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Xiao-Hui Zhang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Lan-Ping Xu
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Yu Wang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Chen-Hua Yan
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Huan Chen
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Yu-Hong Chen
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Wei Han
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Feng-Rong Wang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Yu-Qian Sun
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Xiao-Jun Huang
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, 2019RU029, China
| | - Xiao-Dong Mo
- Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, 2019RU029, China.
| |
Collapse
|
14
|
Sayed AA. Assessing the Diagnostic Values of the Neutrophil-to-Lymphocyte Ratio (NLR) and Systematic Immunoinflammatory Index (SII) as Biomarkers in Predicting COVID-19 Severity: A Multicentre Comparative Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:602. [PMID: 38674248 PMCID: PMC11052014 DOI: 10.3390/medicina60040602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 03/23/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
COVID-19 has been notoriously unpredictable in its clinical course. Such unpredictability poses a challenge to clinicians in predicting patients who will develop severe cases and possibly die from the infection. This study aims to assess and compare the diagnostic value of the NLR and SII as biomarkers in predicting COVID-19 severity, represented by mortality, with a multicentre comparative study including 855 patients in Saudi Arabia. Descriptive and analytical statistics were used to compare haematological indices between survivors and non-survivors. The median age of patients included was 41 years old, with an almost equal ratio of men to women. Most participants were Saudis, and the mortality rate in the study cohort was 13.22%. Non-survivors, as compared to survivors, were significantly older, had lower RBC counts, haemoglobin and haematocrit levels, as well as significantly higher WBC and neutrophil counts. Both the NLR and SII were capable of differentiating between survivors and non-survivors, with the latter having significantly higher values. However, the NLR was superior to the SII in such differentiation, as it had a larger area under the curve. This study further confirms the diagnostic values of the NLR and SII as biomarkers in predicting COVID-19 severity and mortality, with the NLR being more sensitive and specific. Clinical guidelines on managing COVID-19 cases should benefit from these findings by harnessing the value of the NLR in COVID-19 management.
Collapse
Affiliation(s)
- Anwar A Sayed
- Department of Basic Medical Sciences, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
15
|
Kronstein-Wiedemann R, Tausche K, Kolditz M, Teichert M, Thiel J, Koschel D, Tonn T, Künzel SR. Long-COVID is Associated with Impaired Red Blood Cell Function. Horm Metab Res 2024; 56:318-323. [PMID: 37890507 DOI: 10.1055/a-2186-8108] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
COVID-19 disease, caused by the severe acute respiratory syndrome virus 2 (SARS-CoV-2), induces a broad spectrum of clinical symptoms ranging from asymptomatic cases to fatal outcomes. About 10-35% of all COVID-19 patients, even those with mild COVID-19 symptoms, continue to show symptoms, i. e., fatigue, shortness of breath, cough, and cognitive dysfunction, after initial recovery. Previously, we and others identified red blood cell precursors as a direct target of SARS-CoV-2 and suggested that SARS-CoV-2 induces dysregulation in hemoglobin- and iron-metabolism contributing to the severe systemic course of COVID-19. Here, we put particular emphasis on differences in parameters of clinical blood gas analysis and hematological parameters of more than 20 healthy and Long-COVID patients, respectively. Long-COVID patients showed impaired oxygen binding to hemoglobin with concomitant increase in carbon monoxide binding. Hand in hand with decreased plasma iron concentration and transferrin saturation, mean corpuscular hemoglobin was elevated in Long-COVID patients compared to healthy donors suggesting a potential compensatory mechanism. Although blood pH was within the physiological range in both groups, base excess- and bicarbonate values were significantly lower in Long-COVID patients. Furthermore, Long-COVID patients displayed reduced lymphocyte levels. The clinical relevance of these findings, e. g., as a cause of chronic immunodeficiency, remains to be investigated in future studies. In conclusion, our data suggest impaired erythrocyte functionality in Long-COVID patients, leading to diminished oxygen supply. This in turn could be an explanation for the CFS, dyspnea and anemia. Further investigations are necessary to identify the underlying pathomechanisms.
Collapse
Affiliation(s)
- Romy Kronstein-Wiedemann
- Laboratory for Experimental Transfusion Medicine, Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Red Cross Blood Donation Service North-East, Institute for Transfusion Medicine, Dresden, Germany
| | - Kristin Tausche
- Division of Pneumology, Medical Department I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Martin Kolditz
- Division of Pneumology, Medical Department I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Madeleine Teichert
- German Red Cross Blood Donation Service North-East, Institute for Transfusion Medicine, Dresden, Germany
| | - Jessica Thiel
- Laboratory for Experimental Transfusion Medicine, Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Dirk Koschel
- Division of Pneumology, Medical Department I, University Hospital Carl Gustav Carus, Dresden, Germany
- Department of Internal Medicine and Pneumology, Fachkrankenhaus Coswig, Lung Center, Coswig, Germany
| | - Torsten Tonn
- Laboratory for Experimental Transfusion Medicine, Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Red Cross Blood Donation Service North-East, Institute for Transfusion Medicine, Dresden, Germany
| | - Stephan R Künzel
- Laboratory for Experimental Transfusion Medicine, Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Red Cross Blood Donation Service North-East, Institute for Transfusion Medicine, Dresden, Germany
| |
Collapse
|
16
|
Sanikommu DR, Nadeem A, Ponnusamy V. Coombs-Negative Hemolytic Anemia in an Elderly COVID-19 Patient. Cureus 2024; 16:e58589. [PMID: 38765388 PMCID: PMC11102707 DOI: 10.7759/cureus.58589] [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] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
COVID-19 infections are known to cause multi-organ complications. Hematological complications like autoimmune hemolytic anemia with a positive direct antiglobulin test (DAT), are commonly encountered. However, Coombs-negative hemolytic anemia is extremely rare. We report an interesting case of an elderly female with moderate-severe acute respiratory distress syndrome in the setting of COVID-19 pneumonia-causing Coombs-negative hemolytic anemia. This patient initially presented with sudden onset abdominal pain and vomiting, found to have an incarcerated inguinal hernia with small bowel obstruction (SBO) on imaging. Additionally, labs revealed positive COVID-19 antigen test and normocytic anemia. The hospital course was complicated by worsening hemolytic anemia and thrombocytopenia requiring blood products. Extensive workup for hemolysis in this patient with no prior hematological abnormalities, was negative for DAT and other conditions associated with or causative of hemolysis. At discharge, hemolytic parameters improved and on follow-up, hemoglobin returned to baseline, and repeat hemolytic parameters were normal. This case emphasizes the importance of considering SARS-CoV-2 along with other viral infections as one of the differentials for Coombs-negative hemolytic anemia.
Collapse
Affiliation(s)
| | - Ahmed Nadeem
- Oncology, New York Medical College, Landmark Medical Center, Woonsocket, USA
| | - Vignesh Ponnusamy
- Critical Care Medicine, New York Medical College, Landmark Medical Center, Woonsocket, USA
| |
Collapse
|
17
|
Fattizzo B, Röth A, Broome CM, Khan U, Wardęcki M, Cordoba M, Barcellini W. COVID-19 vaccine safety and immunogenicity in patients with cold agglutinin disease receiving concomitant sutimlimab. Am J Hematol 2024; 99:789-791. [PMID: 38385639 DOI: 10.1002/ajh.27234] [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: 10/06/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
Patients with cold agglutinin disease (CAD) are more vulnerable to infectious agents, thus the COVID-19 pandemic has posed a particular risk to this population. Sutimlimab Phase 3 studies CARDINAL and CADENZA spanned the period before and during the pandemic; investigators were advised to vaccinate enrolled patients without stopping treatment. Of 61 completers from both studies, 47 received ≥1 dose of a COVID-19 vaccine. In the immunogenicity analysis (n = 27) all patients developed an immune response post-COVID-19 vaccination, with detectable immunoglobulin G anti-spike antibodies. Analysis of six patients with booster vaccinations demonstrated increased immune responses pre- to post-booster. COVID-19 vaccines were well tolerated in patients with CAD receiving sutimlimab treatment, and no signs of hemolytic exacerbations were observed post-vaccination.
Collapse
Affiliation(s)
- Bruno Fattizzo
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alexander Röth
- University Hospital Essen, Department of Hematology and Stem Cell Transplantation, University Duisburg-Essen, Essen, Germany
| | | | | | | | | | - Wilma Barcellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
18
|
Kakavandi S, Hajikhani B, Azizi P, Aziziyan F, Nabi-Afjadi M, Farani MR, Zalpoor H, Azarian M, Saadi MI, Gharesi-Fard B, Terpos E, Zare I, Motamedifar M. COVID-19 in patients with anemia and haematological malignancies: risk factors, clinical guidelines, and emerging therapeutic approaches. Cell Commun Signal 2024; 22:126. [PMID: 38360719 PMCID: PMC10868124 DOI: 10.1186/s12964-023-01316-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/13/2023] [Indexed: 02/17/2024] Open
Abstract
Extensive research in countries with high sociodemographic indices (SDIs) to date has shown that coronavirus disease 2019 (COVID-19) may be directly associated with more severe outcomes among patients living with haematological disorders and malignancies (HDMs). Because individuals with moderate to severe immunodeficiency are likely to undergo persistent infections, shed virus particles for prolonged periods, and lack an inflammatory or abortive phase, this represents an overall risk of morbidity and mortality from COVID-19. In cases suffering from HDMs, further investigation is needed to achieve a better understanding of triviruses and a group of related variants in patients with anemia and HDMs, as well as their treatment through vaccines, drugs, and other methods. Against this background, the present study aimed to delineate the relationship between HDMs and the novel COVID-19, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Besides, effective treatment options for HDM cases were further explored to address this epidemic and its variants. Therefore, learning about how COVID-19 manifests in these patients, along with exploiting the most appropriate treatments, may lead to the development of treatment and care strategies by clinicians and researchers to help patients recover faster. Video Abstract.
Collapse
Affiliation(s)
- Sareh Kakavandi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Hajikhani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Paniz Azizi
- Psychological and Brain Science Departments, Program in Neuroscience, Indiana University, Bloomington, IN, USA
| | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Marzieh Ramezani Farani
- Department of Biological Sciences and Bioengineering, Nano Bio High-Tech Materials Research Center, Inha University, Incheon, 22212, Republic of Korea
| | - Hamidreza Zalpoor
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Maryam Azarian
- Department of Radiology, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | | | | | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co., Ltd., Shiraz, 7178795844, Iran.
| | - Mohammad Motamedifar
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Shiraz HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
19
|
Hamad RS, Al-Kuraishy HM, Alexiou A, Papadakis M, Ahmed EA, Saad HM, Batiha GES. SARS-CoV-2 infection and dysregulation of nuclear factor erythroid-2-related factor 2 (Nrf2) pathway. Cell Stress Chaperones 2023; 28:657-673. [PMID: 37796433 PMCID: PMC10746631 DOI: 10.1007/s12192-023-01379-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/19/2023] [Accepted: 09/04/2023] [Indexed: 10/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a recent pandemic caused by a novel severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) leading to pulmonary and extra-pulmonary manifestations due to the development of oxidative stress (OS) and hyperinflammation. The underlying cause for OS and hyperinflammation in COVID-19 may be related to the inhibition of nuclear factor erythroid 2-related factor 2 (Nrf2), a master regulator of antioxidative responses and cellular homeostasis. The Nrf2 pathway inhibits the expression of pro-inflammatory cytokines and the development of cytokine storm and OS in COVID-19. Nrf2 activators can attenuate endothelial dysfunction (ED), renin-angiotensin system (RAS) dysregulation, immune thrombosis, and coagulopathy. Hence, this review aimed to reveal the potential role of the Nrf2 pathway and its activators in the management of COVID-19. As well, we tried to revise the mechanistic role of the Nrf2 pathway in COVID-19.
Collapse
Affiliation(s)
- Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, 31982, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, 12411, Egypt
| | - Hayder M Al-Kuraishy
- Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Eman A Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt.
| |
Collapse
|
20
|
Khizer U, Scott J, Chitkara A, Arslan S, Shoukat S. Autoimmune Hemolytic Anemia in a Patient With COVID-19 Pneumonia: A Case Report on a Rare Presentation. Cureus 2023; 15:e49067. [PMID: 38125223 PMCID: PMC10730459 DOI: 10.7759/cureus.49067] [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] [Accepted: 11/19/2023] [Indexed: 12/23/2023] Open
Abstract
Patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)/coronavirus disease 2019 (COVID-19) pneumonia can have a range of clinical presentations ranging from being asymptomatic to having severe acute respiratory syndrome (SARS). Autoimmune hemolytic anemia (AIHA) is a very rare presentation of COVID-19. We present the case of a 67-year-old male with a past medical history of chronic obstructive pulmonary disease (COPD) who presented to the emergency department (ED) with shortness of breath and was found to be COVID-19-positive. His laboratory results demonstrated autoimmune hemolytic anemia with decreased hemoglobin (Hgb), elevated lactate dehydrogenase (LDH), decreased haptoglobin, peripheral smear showing spherocytes, and a positive direct antiglobulin (Coombs) test. The patient was started on glucocorticoids, but his hemoglobin continued to worsen. The dose of glucocorticoids was increased significantly, and his hemoglobin started improving with the resolution of hemolysis. Autoimmune hemolytic anemia is usually treated with glucocorticoids, but escalating glucocorticoid doses increases the risk of side effects. This case report highlights the importance of further research needed to establish guidelines for AIHA in the context of COVID-19 pneumonia.
Collapse
Affiliation(s)
- Umair Khizer
- Internal Medicine, University of California, Riverside School of Medicine, Riverside, USA
| | - Jonathan Scott
- Internal Medicine, University of California, Riverside School of Medicine, Riverside, USA
| | - Akshit Chitkara
- Internal Medicine, University of California, Riverside School of Medicine, Riverside, USA
| | - Shukaib Arslan
- Hematology/Hematopoietic Cell Transplant, City of Hope National Medical Center, Duarte, USA
| | - Sonia Shoukat
- Internal Medicine, University of California, Riverside School of Medicine, Riverside, USA
| |
Collapse
|
21
|
Medford S, Jalal Eldin A, Brgdar A, Obwolo L, Ojo AS, Mere C, Ali A. Beyond the Norm: A Case of Multiorgan Injury Triggered by Ibuprofen. Cureus 2023; 15:e46461. [PMID: 37927669 PMCID: PMC10623888 DOI: 10.7759/cureus.46461] [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] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
We report the case of a 71-year-old African American male with a history of chronic obstructive pulmonary disease (COPD), heart failure, vitiligo, penicillin allergy, and cocaine use, who presented with respiratory symptoms and was diagnosed with sepsis, COVID-19 pneumonia, exacerbation of COPD, and acute kidney injury (AKI). Treatment included antibiotics and high-dose steroids. The patient developed thrombocytopenia, autoimmune hemolytic anemia, acute liver failure, and interstitial nephritis associated with prolonged ibuprofen use. High-dose steroids and ibuprofen discontinuation led to significant improvement. This case highlights the rare occurrence of multiorgan injury from ibuprofen use, possibly aggravated by COVID-19, emphasizing the need for cautious non-steroidal anti-inflammatory drug (NSAID) use and close patient monitoring.
Collapse
Affiliation(s)
- Shawn Medford
- College of Medicine, Howard University College of Medicine, Washington, DC, USA
| | | | - Ahmed Brgdar
- Internal Medicine, Howard University Hospital, Washington, DC, USA
| | - Lilian Obwolo
- Internal Medicine, Howard University Hospital, Washington, DC, USA
| | - Ademola S Ojo
- Internal Medicine, Howard University Hospital, Washington, DC, USA
| | - Constance Mere
- Nephrology, Howard University Hospital, Washington, DC, USA
| | - Ahmed Ali
- Oncology, Howard University Hospital, Washington, DC, USA
| |
Collapse
|
22
|
Al-Kuraishy HM, Batiha GES, Al-Gareeb AI, Al-Harcan NAH, Welson NN. Receptor-dependent effects of sphingosine-1-phosphate (S1P) in COVID-19: the black side of the moon. Mol Cell Biochem 2023; 478:2271-2279. [PMID: 36652045 PMCID: PMC9848039 DOI: 10.1007/s11010-023-04658-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/02/2023] [Indexed: 01/19/2023]
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection leads to hyper-inflammation and amplified immune response in severe cases that may progress to cytokine storm and multi-organ injuries like acute respiratory distress syndrome and acute lung injury. In addition to pro-inflammatory cytokines, different mediators are involved in SARS-CoV-2 pathogenesis and infection, such as sphingosine-1-phosphate (S1P). S1P is a bioactive lipid found at a high level in plasma, and it is synthesized from sphingomyelin by the action of sphingosine kinase. It is involved in inflammation, immunity, angiogenesis, vascular permeability, and lymphocyte trafficking through G-protein coupled S1P receptors. Reduction of the circulating S1P level correlates with COVID-19 severity. S1P binding to sphingosine-1-phosphate receptor 1 (S1PR1) elicits endothelial protection and anti-inflammatory effects during SARS-CoV-2 infection, by limiting excessive INF-α response and hindering mitogen-activated protein kinase and nuclear factor kappa B action. However, binding to S1PR2 opposes the effect of S1PR1 with vascular inflammation, endothelial permeability, and dysfunction as the concomitant outcome. This binding also promotes nod-like receptor pyrin 3 (NLRP3) inflammasome activation, causing liver inflammation and fibrogenesis. Thus, higher expression of macrophage S1PR2 contributes to the activation of the NLRP3 inflammasome and the release of pro-inflammatory cytokines. In conclusion, S1PR1 agonists and S1PR2 antagonists might effectively manage COVID-19 and its severe effects. Further studies are recommended to elucidate the potential conflict in the effects of S1P in COVID-19.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Nasser A Hadi Al-Harcan
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Rasheed University College, Baghdad, Iraq
| | - Nermeen N Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, 62511, Egypt.
| |
Collapse
|
23
|
Moguem Soubgui AF, Kojom Foko LP, Embolo Enyegue EL, Ndeme Mboussi WS, Mbebi Enone JP, Ntatou Lemouchele I, Koanga Mogtomo ML. Confounding role of comorbidities and COVID-19 vaccination uptake in clinical utility of hematological biomarkers in Cameroonian patients infected with SARS-CoV-2. IJID REGIONS 2023; 8:129-136. [PMID: 37663007 PMCID: PMC10472227 DOI: 10.1016/j.ijregi.2023.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 09/05/2023]
Abstract
Objectives To analyze variations in blood profile, the extent of hematological disorders, and the impact of comorbidities and COVID-19 vaccination on blood profile parameters and their clinical value for prognostic of SARS-CoV-2 infection. Methods This cross-sectional study took place in Douala, Cameroon. A complete blood count and molecular detection of SARS-CoV-2 were performed on patients. Clinical value was appraised using area under the curve (AUC) analysis. Results In total, 420 participants were included. A significant reduction of hematological parameters such as lymphocytes (p < 0.0001), red blood cells (P = 0.0025), mean corpuscular hemoglobin count (P < 0.0001), and platelets (P = 0.02) was seen in SARS-CoV-2 (+) patients. Anisocytosis was the main hematological disorder (95.5%). Normocytic normochromic anemia was predominant in SARS-CoV-2 (+) (35.3%) while microcytic normochromic anemia was more frequently seen in SARS-CoV-2 (-) (16.5%). Neutrophil-to-lymphocyte (NLR) consistently showed statistically significant AUC ∼0.75 upon stratification for age, gender, and comorbidities, with the exception of COVID-19 vaccination uptake. Conclusion In a context where molecular detection methods are difficultly affordable in health facilities in developing countries such as Cameroon, NLR could be clinically interesting for identifying SARS-CoV-2 infected individuals, especially those with comorbidities.
Collapse
Affiliation(s)
| | - Loick Pradel Kojom Foko
- Department of Animal Organisms, Faculty of Science, The University of Douala, Cameroon
- Centre de Recherche et d'Expertise en Biologie, Douala, Cameroon
| | | | - Wilfried Steve Ndeme Mboussi
- Department of Biochemistry, Faculty of Sciences, The University of Douala, Cameroon
- Centre de Recherche et d'Expertise en Biologie, Douala, Cameroon
| | | | - Idriss Ntatou Lemouchele
- Department of Biochemistry, Faculty of Sciences, The University of Douala, Cameroon
- Centre de Recherche et d'Expertise en Biologie, Douala, Cameroon
| | - Martin Luther Koanga Mogtomo
- Department of Biochemistry, Faculty of Sciences, The University of Douala, Cameroon
- Centre de Recherche et d'Expertise en Biologie, Douala, Cameroon
| |
Collapse
|
24
|
Al-kuraishy HM, Al-Gareeb AI, Albezrah NKA, Bahaa HA, El-Bouseary MM, Alexiou A, Al-Ziyadi SH, Batiha GES. Pregnancy and COVID-19: high or low risk of vertical transmission. Clin Exp Med 2023; 23:957-967. [PMID: 36251144 PMCID: PMC9574177 DOI: 10.1007/s10238-022-00907-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/25/2022] [Indexed: 11/03/2022]
Abstract
Coronavirus disease 19 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome 2 (SARS-CoV-2). Throughout the pandemic, evidence on the effects of COVID-19 during pregnancy has been inadequate due to the limited number of studies published. Therefore, the objective of this systematic review was to evaluate current literature regarding the effects of COVID-19 during pregnancy and establish pregnancy outcomes and vertical and perinatal transmission during pregnancy. Multiple databases were searched, including Embase, Medline, Web of Science, Scopus, and Cochrane Central Register of Control Clinical Trials, using the following keywords: [Pregnancy] AND [COVID-19 OR SARS-CoV-2 OR nCoV-19] OR [Perinatal transmission, Vertical transmission (VT), Pregnancy complications], [Pregnancy] AND [Hyperinflammation OR Cytokine storm]. We excluded in vitro and experimental studies, but also ex-vivo and animal study methods. To exclude the risk of bias during data collection and interpretation, all included studies were peer-reviewed publications. This review is estimated to tabulate the study intervention characteristics and compare them against the planned groups for each synthesis. Our findings showed that pregnant women are commonly susceptible to respiratory viral infections and severe pneumonia due to physiological immune suppression and pregnancy-induced changes. VT of SARS-CoV-2 infection during pregnancy is associated with a great deal of controversy and conflict. However, there is still no robust clinical evidence of VT. Furthermore, the clinical presentation and management of COVID-19 during pregnancy are nearly identical to those of non-pregnant women. Finally, chloroquine and remdesivir are the only two drugs evaluated as adequate for the management of COVID-19 during pregnancy.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyah University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyah University, Baghdad, Iraq
| | | | - Haitham Ahmed Bahaa
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
- AFNP Med Austria, Vienna, Austria
| | - Shatha Hallal Al-Ziyadi
- Saudi Board Certified in Obstetrics & Gynecology, Assistant Professor at Taif University, Taif, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 AlBeheira Egypt
| |
Collapse
|
25
|
Al-kuraishy HM, Al-Gareeb AI, Alkazmi L, El-Bouseary MM, Hamad RS, Abdelhamid M, Batiha GES. The Potential Nexus between Helminths and SARS-CoV-2 Infection: A Literature Review. J Immunol Res 2023; 2023:5544819. [PMID: 37383608 PMCID: PMC10299886 DOI: 10.1155/2023/5544819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/05/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
Chronic helminth infections (CHIs) can induce immunological tolerance through the upregulation of regulatory T cells. In coronavirus disease 2019 (COVID-19), abnormal adaptive immune response and exaggerated immune response may cause immune-mediated tissue damage. Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) and CHIs establish complicated immune interactions due to SARS-CoV-2-induced immunological stimulation and CHIs-induced immunological tolerance. However, COVID-19 severity in patients with CHIs is mild, as immune-suppressive anti-inflammatory cytokines counterbalance the risk of cytokine storm. Since CHIs have immunomodulatory effects, therefore, this narrative review aimed to clarify how CHIs modulate the immunoinflammatory response in SARS-CoV-2 infection. CHIs, through helminth-derived molecules, may suppress SARS-CoV-2 entry and associated hyperinflammation through attenuation of the inflammatory signaling pathway. In addition, CHIs may reduce the COVID-19 severity by reducing the SARS-CoV-2 entry points in the initial phase and immunomodulation in the late phase of the disease by suppressing the release of pro-inflammatory cytokines. In conclusion, CHIs may reduce the severity of SARS-CoV-2 infection by reducing hyperinflammation and exaggerated immune response. Thus, retrospective and prospective studies are recommended in this regard.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Luay Alkazmi
- Biology Department, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Rabab S. Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza 12411, Egypt
| | - Mahmoud Abdelhamid
- Department of Parasitology, Faculty of Veterinary Medicine, Aswan University, Aswan 81528, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| |
Collapse
|
26
|
Al Qahtani SY. Impact of hyperchloremia on inflammatory markers, serum creatinine, hemoglobin, and outcome in critically ill patients with COVID-19 infection. J Med Life 2023; 16:699-706. [PMID: 37520482 PMCID: PMC10375338 DOI: 10.25122/jml-2023-0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/05/2023] [Indexed: 08/01/2023] Open
Abstract
Hyperchloremia has negative consequences, such as increased proinflammatory mediators, renal dysfunction, and mortality in patients with septic shock. However, data on the effects of hyperchloremia on COVID-19 infections are scarce. The study aimed to investigate the effects of hyperchloremia on inflammatory markers, serum creatinine, hemoglobin levels, and outcomes in critically ill COVID-19 patients. A retrospective review of all adult patients admitted to the ICU at King Fahd University Hospital with a moderate to severe COVID-19 infection from January 2020 to August 2021 was performed. Serum chloride levels, ferritin, lactate dehydrogenase (LDH), C-reactive protein (CRP), creatinine, and hemoglobin levels were collected on the first and third days of ICU admission. Demographic data, oxygen support modality, ICU length of stay (ICU LOS), renal replacement therapy (RRT), and deaths were collected. Of 420 patients, 255 were included; 97 (38%) had hyperchloremia, while 158 (62%) did not. Hyperchloremic patients had a higher percentage of increases in ferritin (54.6%), CRP (6.2%), and LDH (15.5%) between the first and third days of admission, compared to non-hyperchloremic patients (43.7%, 6.3%, and 5.7%, respectively). The decrease in hemoglobin levels was similar in both groups (p=0.103). There was a significant association between hyperchloremia and an increase in serum creatinine (p<0.0001). Sixty-six (68%) patients required endotracheal intubation in the hyperchloremic group (p=0.003). The mortality rate was significant in the hyperchloremic cohort (p=<0.0001). Hyperchloremia was significantly associated with increased risks of kidney injury, endotracheal intubation, and death. However, hyperchloremia was not associated with increased ferritin, CRP, or hemoglobin decreases in critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Shaya Yaanallah Al Qahtani
- Department of Internal Medicine and Critical Care, College of Medicine, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
27
|
Silva JM, Gomes Cochicho J, Cruz Nodarse A, Lavadinho I. A Compelling Case of Autoimmune Hemolytic Anemia and Its Potential Association With SARS-CoV-2. Cureus 2023; 15:e39566. [PMID: 37378094 PMCID: PMC10292631 DOI: 10.7759/cureus.39566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Autoimmune hemolytic anemia can be caused by infections, lymphoproliferative disorders, autoimmune disorders, or triggered by drugs or toxins. We present the case of a 92-year-old man admitted with gastrointestinal symptoms. He presented with autoimmune hemolytic anemia. The etiologic study was negative for autoimmune conditions or solid masses. Viral serologies were negative, and RT-PCR for SARS-CoV-2 was positive. The patient began treatment with corticoid, with resulted in cessation of hemolysis and improvement of the anemia. A few cases of autoimmune hemolytic anemia have been reported in COVID-19 patients. In this case, the infection seems to coincide with the hemolysis period, and we found no other cause for this event. So, we highlight the need to search for SARS-CoV-2 as a possible infective cause of autoimmune hemolytic anemia.
Collapse
Affiliation(s)
- José Miguel Silva
- Internal Medicine, Hospital Doutor José Maria Grande, Portalegre, PRT
| | | | | | - Isabel Lavadinho
- Internal Medicine, Hospital Doutor José Maria Grande, Portalegre, PRT
| |
Collapse
|
28
|
Choi R, Park W, Chun G, Lee SG, Lee EH. Utilization of Glucose-6-Phosphate Dehydrogenase Test and the Prevalence of Enzyme Deficiency in Korea. J Clin Med 2023; 12:jcm12093179. [PMID: 37176619 PMCID: PMC10179720 DOI: 10.3390/jcm12093179] [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: 03/30/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Glucose-5-phosphate dehydrogenase (G6PD) deficiency is an X-linked genetic disorder that affects red blood cells' metabolism. This retrospective study aimed to investigate the prevalence and characteristics of G6PD testing in Korea. Data were collected from laboratory information systems between July 2021 and June 2022. A total of 5193 patients (1722 males and 3471 females) with a median age of 55.1 years (interquartile range, IQR 44.6 to 64.5) were tested for whole blood G6PD, with 1.6% of tests performed on patients of non-Korean ethnicity. The majority of tests were performed in hospitals (37.7%) or local clinics (34.5%). Interestingly, no female children were tested for whole blood G6PD during the study period. The prevalence of decreased G6PD activity (<7.9 U/g Hb) was 0.4% (19/5111 Koreans and 2/82 non-Koreans), and only seven male patients with G6PD deficiency (<30% of the male median) were identified, with ages ranging from 4.8 months to 50.2 years. No female patients with G6PD deficiency were found. Further research is necessary to determine the clinical significance of G6PD test results and monitor their use.
Collapse
Affiliation(s)
- Rihwa Choi
- Department of Laboratory Medicine, Green Cross Laboratories, Yongin 16924, Republic of Korea
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Wonseo Park
- Infectious Disease Research Center, Green Cross Laboratories, Yongin 16924, Republic of Korea
| | - Gayoung Chun
- Infectious Disease Research Center, Green Cross Laboratories, Yongin 16924, Republic of Korea
| | - Sang Gon Lee
- Department of Laboratory Medicine, Green Cross Laboratories, Yongin 16924, Republic of Korea
| | - Eun Hee Lee
- Green Cross Laboratories, Yongin 16924, Republic of Korea
| |
Collapse
|
29
|
Alomair BM, Al‐Kuraishy HM, Al‐Gareeb AI, Al‐Buhadily AK, Alexiou A, Papadakis M, Alshammari MA, Saad HM, Batiha GE. Mixed storm in SARS-CoV-2 infection: A narrative review and new term in the Covid-19 era. Immun Inflamm Dis 2023; 11:e838. [PMID: 37102645 PMCID: PMC10132185 DOI: 10.1002/iid3.838] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/28/2023] Open
Abstract
Coronavirus disease 2019 (Covid-19) is caused by a novel severe acute respiratory syndrome coronavirus virus type 2 (SARS-CoV-2) leading to the global pandemic worldwide. Systemic complications in Covid-19 are mainly related to the direct SARS-CoV-2 cytopathic effects, associated hyperinflammation, hypercytokinemia, and the development of cytokine storm (CS). As well, Covid-19 complications are developed due to the propagation of oxidative and thrombotic events which may progress to a severe state called oxidative storm and thrombotic storm (TS), respectively. In addition, inflammatory and lipid storms are also developed in Covid-19 due to the activation of inflammatory cells and the release of bioactive lipids correspondingly. Therefore, the present narrative review aimed to elucidate the interrelated relationship between different storm types in Covid-19 and the development of the mixed storm (MS). In conclusion, SARS-CoV-2 infection induces various storm types including CS, inflammatory storm, lipid storm, TS and oxidative storm. These storms are not developing alone since there is a close relationship between them. Therefore, the MS seems to be more appropriate to be related to severe Covid-19 than CS, since it develops in Covid-19 due to the intricate interface between reactive oxygen species, proinflammatory cytokines, complement activation, coagulation disorders, and activated inflammatory signaling pathway.
Collapse
Affiliation(s)
- Basil Mohammed Alomair
- Department of Medicine, College of Medicine, Internal Medicine and EndocrinologyJouf UniversityAl‐JoufSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali K. Al‐Buhadily
- Department of Clinical Pharmacology, Medicine, and Therapeutic, Medical Faculty, College of MedicineAl‐Mustansiriyah UniversityBaghdadIraq
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten‐HerdeckeUniversity of Witten‐HerdeckeWuppertalGermany
| | - Majed Ayed Alshammari
- Department of MedicinePrince Mohammed Bin Abdulaziz Medical CitySakakaAl‐JoufSaudi Arabia
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMarsaMatruhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourEgypt
| |
Collapse
|
30
|
Al-kuraishy HM, Al-Buhadily AK, Al-Gareeb AI, Alorabi M, Hadi Al-Harcan NA, El-Bouseary MM, Batiha GES. Citicoline and COVID-19: vis-à-vis conjectured. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1463-1475. [PMID: 36063198 PMCID: PMC9442587 DOI: 10.1007/s00210-022-02284-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a current pandemic disease caused by a novel severe acute respiratory syndrome coronavirus virus respiratory type 2 (SARS-CoV-2). SARS-CoV-2 infection is linked with various neurological manifestations due to cytokine-induced disruption of the blood brain barrier (BBB), neuroinflammation, and peripheral neuronal injury, or due to direct SARS-CoV-2 neurotropism. Of note, many repurposed agents were included in different therapeutic protocols in the management of COVID-19. These agents did not produce an effective therapeutic eradication of SARS-CoV-2, and continuing searching for novel anti-SARS-CoV-2 agents is a type of challenge nowadays. Therefore, this study aimed to review the potential anti-inflammatory and antioxidant effects of citicoline in the management of COVID-19.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali K. Al-Buhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Mohammed Alorabi
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif, 21944 Saudi Arabia
| | - Nasser A. Hadi Al-Harcan
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Rasheed University College, Baghdad, Iraq
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, Damanhour, 22511 Egypt
| |
Collapse
|
31
|
Giordano G, Teresa Bochicchio M, Niro G, Lucchesi A, Napolitano M. Genetic regulation of iron homeostasis in sideropenic patients with mild COVID-19 disease under a new oral iron formulation: Lessons from a different perspective. Immunobiology 2022; 227:152297. [PMID: 36327544 PMCID: PMC9597571 DOI: 10.1016/j.imbio.2022.152297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 11/05/2022]
Abstract
Background Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) needs iron to replicate itself. Coronaviruses are able to upregulate Chop/Gadd153 and Arg1 genes, consequently leading to CD8 lymphocytes decrease, degradation of asparagine and decreased nitric oxide (NO), thus impairing immune response and antithrombotic functions. Little is known about regulation of genes involved in iron metabolism in paucisymptomatic patients with COVID-19 disease or in patients with iron deficiency treated with sucrosomial iron. Methods Whole blood was taken from the COVID-19 patients and from patients with sideropenic anemia, treated or not (control group) with iron supplementations. Enrolled patients were: affected by COVID19 under sucrosomal iron support (group A), affected by COVID-19 not under oral iron support (group B), iron deficiency not under treatment, not affected by COVID19 (control group). After RNA extraction and complementary DNA (cDNA) synthesis of Arg1, Hepcidin and Chop/Gadd153, gene expression from the 3 groups was measured by qRT-PCR. M2 macrophages were detected by cytofluorimetry using CD163 and CD14 markers. Results Forty patients with COVID-19 (group A), 20 patients with iron deficiency treated with sucrosomial iron (group B) and 20 patients with iron deficiency not under treatment (control group) were enrolled. In all the patients supported with oral sucrosomial iron, the gene expression of Chop, Arg1 and Hepcidin genes was lower than in sideropenic patients not supported with iron, M1 macrophages polarization and functional iron deficiency was also lower in group A and B, than observed in the control group. Conclusions New oral iron formulations, as sucrosomial iron, are able to influence the expression of genes like Chop and Arg1 and to influence M2 macrophage polarization mainly in the early phase of COVID-19 disease.
Collapse
Affiliation(s)
- Giulio Giordano
- Division of Internal Medicine, Hematology Service, Regional Hospital “A. Cardarelli”, 86100 Campobasso, Italy
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Giovanna Niro
- Division of Laboratory Medicine, Regional Hospital “A. Cardarelli”, 86100 Campobasso, Italy
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) “Dino Amadori”, Meldola (FC), Italy,Corresponding author
| | - Mariasanta Napolitano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), Haematology Unit, University Hospital “P. Giaccone”, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
32
|
Atnaf A, Shiferaw AA, Tamir W, Akelew Y, Toru M, Tarekegn D, Bewket B, Reta A. Hematological Profiles and Clinical Outcome of COVID-19 Among Patients Admitted at Debre Markos Isolation and Treatment Center, 2020: A Prospective Cohort Study. J Blood Med 2022; 13:631-641. [PMID: 36405428 PMCID: PMC9667503 DOI: 10.2147/jbm.s380539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/21/2022] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is coronavirus isolated from SARS patients. As far as the researchers' knowledge, there was paucity of studies conducted in Ethiopia, particularly in the study area. As immune protection is arisen from our blood cells, assessing their level will provide a clue for controlling the disease and monitoring the prognosis. This study will also provide additional information for clinical intervention and patient management. PURPOSE This study aimed to investigate the hematological profile and clinical outcome of coronavirus disease-19 (COVID-19) among patients admitted to the Debre Markos Isolation and Treatment Center (DMITC). MATERIAL AND METHODS A prospective cohort study was conducted among 136 COVID-19 adult patients at DMITC from January 1, 2020 to March 30, 2021. Data related to clinical, hematological profiles and socio-demographic factors were collected, entered into Epi data, and analyzed using STATA 14.2 software. Multivariable logistic regression was applied to determine the predictor variable and a p-value <0.05 was considered significant. RESULTS Of 136 COVID-19 patients, 28.68% had died. The mean age of patients was 47.21±1.29 years. The hematological profile of the patients revealed that 28% had abnormal leukocyte, 23% abnormal lymphocyte, 44.85% abnormal granulocyte, 22.06% abnormal monocyte, 30.15% abnormal RBC and 87% abnormal platelet counts. The prevalence of anemia was 13.24%. CONCLUSION Leukocytosis (mainly granulocytosis and monocytosis) and lymphopenia, were the predominant abnormal findings of complete blood cell count (CBC) analysis of the patient's blood. Most of the patients had abnormally low platelet counts. RBC count and hematocrit determination were the only significant predictors of death. The clinician could manage cases according to the hematological findings of the patients. Further experimental studies should be conducted to determine hematological parameter changes and the clinical outcome of the disease.
Collapse
Affiliation(s)
- Aytenew Atnaf
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Abtie Abebaw Shiferaw
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Workineh Tamir
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Yibeltal Akelew
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Milkiyas Toru
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Daniel Tarekegn
- Department of Public health, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bekalu Bewket
- Department of Nursing, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Alemayehu Reta
- Department of Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
33
|
Cosentino M, Marino F. Understanding the Pharmacology of COVID-19 mRNA Vaccines: Playing Dice with the Spike? Int J Mol Sci 2022; 23:10881. [PMID: 36142792 PMCID: PMC9502275 DOI: 10.3390/ijms231810881] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease-19 (COVID-19) mRNA vaccines are the mainstays of mass vaccination campaigns in most Western countries. However, the emergency conditions in which their development took place made it impossible to fully characterize their effects and mechanism of action. Here, we summarize and discuss available evidence indicating that COVID-19 mRNA vaccines better reflect pharmaceutical drugs than conventional vaccines, as they do not contain antigens but an active SARS-CoV-2 S protein mRNA, representing at the same time an active principle and a prodrug, which upon intracellular translation results in the endogenous production of the SARS-CoV-2 S protein. Both vaccine-derived SARS-CoV-2 S protein mRNA and the resulting S protein exhibit a complex pharmacology and undergo systemic disposition. Defining COVID-19 mRNA vaccines as pharmaceutical drugs has straightforward implications for their pharmacodynamic, pharmacokinetic, clinical and post-marketing safety assessment. Only an accurate characterization of COVID-19 mRNA vaccines as pharmaceutical drugs will guarantee a safe, rational and individualized use of these products.
Collapse
Affiliation(s)
- Marco Cosentino
- Center for Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
| | | |
Collapse
|
34
|
Autoimmune Diseases Induced or Exacerbated by COVID-19: A Single Center Experience. Autoimmune Dis 2022; 2022:9171284. [PMID: 36111059 PMCID: PMC9470368 DOI: 10.1155/2022/9171284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/17/2022] [Accepted: 08/12/2022] [Indexed: 12/03/2022] Open
Abstract
The association between infectious diseases and autoimmunity has long been reported. Specifically, during the coronavirus disease 2019 (COVID-19) pandemic, this relation was further emphasized. The interplay between the two disease processes remains interesting, yet incompletely defined. Herein, we report a case series of six patients presenting with autoimmune phenomena first developed or exacerbated following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We describe the disease course and discuss the possible mechanisms underlying the association between autoimmunity and COVID-19.
Collapse
|