51
|
Khan MW, Ahmad M, Qudrat S, Afridi F, Khan NA, Afridi Z, Fahad, Azeem T, Ikram J. Vagal nerve stimulation for the management of long COVID symptoms. INFECTIOUS MEDICINE 2024; 3:100149. [PMID: 39678231 PMCID: PMC11638592 DOI: 10.1016/j.imj.2024.100149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024]
Abstract
This review investigates the therapeutic potential of vagal nerve stimulation (VNS) in managing long COVID, a condition marked by persistent symptoms following acute SARS-CoV-2 infection. Long COVID manifests as ongoing fatigue, cognitive impairment, and autonomic dysfunction, hypothesized to arise from sustained inflammatory and neurological dysregulation. The vagus nerve, central to modulating systemic inflammation and autonomic homeostasis, represents a promising therapeutic target for symptom alleviation through VNS. A comprehensive literature search was conducted across PubMed, Scopus, and Web of Science to identify studies evaluating VNS in the context of long COVID. Preliminary evidence from small-scale pilot studies suggests VNS may attenuate systemic inflammation through activation of the cholinergic anti-inflammatory pathway (CAP), thus restoring autonomic balance and ameliorating symptoms such as fatigue, cognitive dysfunction, and anxiety. In targeting the inflammatory cascade that underlies both acute COVID-19 pathophysiology and its prolonged sequelae, VNS holds potential as an innovative intervention for persistent post-viral symptoms. While these initial findings indicate promise, current data remain limited in scope and robustness, underscoring the need for larger, controlled trials to validate the efficacy and mechanisms of VNS in long COVID management. Establishing a clearer understanding of VNS's impact on inflammation and autonomic regulation in this context is crucial to inform clinical guidelines and therapeutic strategies for long COVID, potentially offering a targeted approach for mitigating this disabling condition.
Collapse
Affiliation(s)
- Malik W.Z. Khan
- Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Muhammad Ahmad
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Salma Qudrat
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Fatma Afridi
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Najia Ali Khan
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Zain Afridi
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Fahad
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Touba Azeem
- Khyber Medical College, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Jibran Ikram
- Department of Cardiology, Cleveland Clinic Foundation, Ohio 44195, USA
| |
Collapse
|
52
|
Horii T, Kishino M, Morishita K, Kanda E, Takahashi M, Kimura K, Adachi T, Oyama J, Okawa S, Tateishi U. Factors Associated With Rebleeding and Early Mortality Following Transcatheter Arterial Embolization for Spontaneous Muscle Hematoma: A Single-Center Experience Including the Period of the Coronavirus Disease Pandemic. Cureus 2024; 16:e76347. [PMID: 39867017 PMCID: PMC11757941 DOI: 10.7759/cureus.76347] [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: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
Objectives We aim to investigate factors associated with rebleeding and mortality within one month of transcatheter arterial embolization (TAE) for spontaneous muscle hematoma (SMH) and the impact of the novel coronavirus disease 2019 (COVID-19). Methods This retrospective analysis included 33 patients who underwent TAE for SMH at a single center between 2012 and 2022. After 2020, eight of these patients had the COVID-19 infection. Patient characteristics, laboratory findings, embolic materials, and imaging findings were compared between the rebleeding and non-rebleeding groups, as well as between the early mortality and survival groups. Results Among all patients, 72.7% were on anticoagulant therapy before the onset of SMH. Of these, 27.2% required retreatment due to rebleeding. Patients who experienced rebleeding were more likely to have a platelet count below 50,000/µL, fibrinogen levels below 150 mg/dL, and an activated partial thromboplastin time (APTT) ratio above 2.5. Patients with SMH unrelated to anticoagulants had a higher rebleeding rate (56%), which may serve as a predictor of rebleeding. No significant difference in rebleeding rates was observed between patients with and without COVID-19 infection. Early mortality within one month of onset occurred in 24.2% of patients, with a higher prevalence among those with a history of malignancy. However, there was no increase in early mortality among patients who required retreatment for rebleeding. Conclusions Patients with a low platelet count, fibrinogen level, prolonged APTT, and non-anticoagulant-related SMH are at a high risk of rebleeding and require close monitoring. Severe comorbidities, including malignancies and COVID-19, can affect mortality rates. TAE remained effective even in cases of rebleeding. Advances in knowledge This study indicated non-anticoagulant-related SMH and hematological parameters as factors associated with rebleeding.
Collapse
Affiliation(s)
- Toshihiro Horii
- Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, JPN
- Diagnostic Radiology, National Cancer Center Hospital East, Chiba, JPN
| | - Mitsuhiro Kishino
- Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, JPN
| | - Koji Morishita
- Trauma and Acute Care Surgery, Tokyo Medical and Dental University Hospital, Tokyo, JPN
| | | | - Marie Takahashi
- Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, JPN
| | - Koichiro Kimura
- Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, JPN
| | - Takuya Adachi
- Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, JPN
| | - Jun Oyama
- Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, JPN
| | - Sayuri Okawa
- Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, JPN
| | - Ukihide Tateishi
- Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, JPN
| |
Collapse
|
53
|
Kuruca N, Atilla A, Kaya MT, Gokmen S, Nursal AF, Kilic O, Kuruoglu T, Temocin F, Guvenc T, Yigit S, Guvenc D. Effect of VDR and TLR2 gene variants on the clinical course of patients with COVID-19 disease. J Investig Med 2024; 72:876-882. [PMID: 39075671 DOI: 10.1177/10815589241270418] [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] [Indexed: 07/31/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, which has caused a major global health crisis, primarily targets the upper and lower respiratory tract. But infected individuals may experience different clinical symptoms, ranging from asymptomatic to critical. The vitamin D receptor (VDR) and Toll-like receptor 2 (TLR2) polymorphisms play a role in the immune response. This study aimed to evaluate the effect of VDR Bsml (rs1544410) and TLR2 23bp indel variants on the clinical status of Turkish patients with COVID-19 disease. A total of 312 people, including 106 intensive care unit (ICU) patients, 103 symptomatic hospitalized patients, and 103 healthy controls, were included in the study. The VDR BsmI and TLR2 23bp indel were genotyped using polymerase chain reaction and/or restriction fragment length fraction methods. The VDR BsmI b/b genotype and b allele were higher in symptomatic patients compared to the healthy control group (p = 0.035). The VDR BsmI B/B and B/b genotype distribution did not differ between ICU patients and both symptomatic patients and controls (p > 0.05). We found that B/B:B/b+b/b and B/B+B/b:b/b were significantly different in symptomatic patients compared to controls (p = 0.033 and p = 0.041, respectively). The VDR BsmI b/b genotype distribution was found to be lower in deceased patients than in living patients (p = 0.023). There was no significant difference between the groups in terms of TLR2 23bp indel genotype and allele distribution (p > 0.05). Our study results suggest that the VDR BsmI b allele may have a role in COVID-19 patients with symptomatic findings. These data need to be repeated in different ethnic and larger sample groups.
Collapse
Affiliation(s)
- Nilufer Kuruca
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Aynur Atilla
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Muhammed Taha Kaya
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Sedat Gokmen
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Kastamonu University, Kastamonu, Turkey
| | - Ayse Feyda Nursal
- Department of Medical Genetics, Faculty of Medicine, Hitit University, Corum, Turkey
| | - Ozgur Kilic
- Department of Internal Medicine, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Tuba Kuruoglu
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Fatih Temocin
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Tolga Guvenc
- Department of Pathology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Serbulent Yigit
- Department of Genetics, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Dilek Guvenc
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
54
|
Ibrahim R. The effect of pre-hospital use of RAS inhibitors on COVID-19 mortality. J Investig Med 2024; 72:863-875. [PMID: 39075674 DOI: 10.1177/10815589241270417] [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] [Indexed: 07/31/2024]
Abstract
The effect of pre-hospital use of renin-angiotensin system (RAS) inhibitors (angiotensin-converting enzyme inhibitors (ACEis)/angiotensin receptor blockers (ARBs)) on clinical outcomes of hypertensive patients with COVID-19 has been questioned due to conflicting reports on this issue. After applying exclusion criteria, 175 COVID-19 hospitalized patients admitted to the Tishreen Hospital from January 1 to July 31, 2021 were retrospectively enrolled in this study. Baseline characteristics and in-hospital mortality rate were assessed between hypertensive (N = 91, 52%) and non-hypertensive (N = 84, 48%) patients, as well as between patients taking ACEis/ARBs and non-ACEis/ARBs within the hypertensive group. A lower mortality rate (51.2 versus 31.9%, p = 0.009) was observed in the hypertensive group (mean age 64.6 years, 64.8% males) compared to the non-hypertensive (mean age 62.6 years, 66.7% males). Patients' mortality in the non-hypertensive group was associated with lower blood oxygen saturation (SPO2 = 75 versus 86%, p = 0.002), increased levels of inflammatory (CRP, white blood cell and neutrophils count), and tissue/renal injury markers (LDH, urea, and creatinine). In the hypertensive group, a lower mortality rate was noted in the ACEis/ARBs group compared to the non-ACEis/ARBs (24.1 versus 45.5%, p = 0.036), and this was associated with a decrease in D-DIMER levels, although not significant (1723 versus 2683 ng/mL, p > 0.05). Death in the non-ACEis/ARBs group was associated with decreased SPO2 and tissue/renal injury markers (LDH, CK, AST, urea, and creatinine). We concluded that hypertension is not a direct cause of poor prognosis in COVID-19 patients and that multi-organ damage is a significant indicator of death from COVID-19. RAS inhibitors could improve the survival of hypertensive COVID-19 patients.
Collapse
Affiliation(s)
- Rama Ibrahim
- Department of Biochemistry and Microbiology, Faculty of Pharmacy,Al-Sham Private University (ASPU), Lattakia, Syria
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Tishreen University, Lattakia, Syria
| |
Collapse
|
55
|
Rivera-Caravaca JM, Frost F, Marín F, Lip GYH. Impact of prior oral anticoagulation therapies on post-discharge outcomes after COVID-19: Results from a global federated health network analysis. Eur J Clin Invest 2024; 54:e14299. [PMID: 39105372 DOI: 10.1111/eci.14299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND The impact of chronic oral anticoagulant (OACs) use on long-term post-discharge outcomes after coronavirus disease 2019 (COVID-19) hospitalisation remains unclear. Herein, we compared clinical outcomes up to 2-years after COVID-19 hospitalisation between patients on vitamin K antagonists (VKAs), direct-acting OACs (DOACs) and no OAC therapy. METHODS Data from TriNetX, a global federated health research network, were used. Adult patients on VKAs, DOACs or no OAC therapy at diagnosis of COVID-19 between 20 January 2020 and 31 December 2021, who were hospitalised for COVID-19, were included. The primary outcomes were all-cause mortality, ischaemic stroke/transient ischaemic attack (TIA)/systemic embolism (SE) and the composite of intracranial haemorrhage (ICH)/gastrointestinal bleeding, at 2 years after COVID-19 hospitalisation. RESULTS We included 110,834 patients with COVID-19. Following propensity score matching (PSM), we identified a decreased mortality risk in DOAC-treated patients compared to the no OAC cohort (RR .808, 95% CI .751-.870). A higher risk of ischaemic stroke/TIA/SE was observed in VKA users compared to DOAC users (RR 1.100, 95% CI 1.020-1.220) and in VKA users compared to patients not taking OAC (RR 1.400, 95% CI 1.140-1.720). VKA use was associated with a greater risk of ICH/gastrointestinal bleeding than DOAC users (RR 1.198, 95% CI 1.066-1.347), while DOAC users had a lower risk compared to no OAC-treated patients (RR .840, 95% CI .754-.936). CONCLUSION COVID-19 patients taking prior DOACs were associated with lower long-term mortality risk and ICH/gastrointestinal bleeding than patients not taking OAC. Compared to patients on DOACs, VKA users were associated with higher risks of mortality, ischaemic stroke/TIA/SE and ICH/gastrointestinal bleeding.
Collapse
Affiliation(s)
- José Miguel Rivera-Caravaca
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, UK
- Faculty of Nursing, University of Murcia, Murcia, Spain
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), CIBERCV, University of Murcia, Murcia, Spain
| | - Freddy Frost
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Francisco Marín
- Department of Cardiology, Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), CIBERCV, University of Murcia, Murcia, Spain
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, UK
- Department of Clinical Medicine, Danish Center for Health Services Research, Aalborg University, Aalborg, Denmark
| |
Collapse
|
56
|
Osati EFO, Shayo GA, Sangeda RZ, Nagu TJ, Moshiro C, Adams N, Ramadhani A, Wajanga B, Muniko A, Seni J, Nicholaus MA, Nyaisonga G, Mbije C, Meda JR, Rainer D, Nkya ME, Mhame P, Samwel L, Vumilia L, Shekalaghe S, Kilonzo KG, Makubi A. Clinical manifestations and treatment outcomes among hospitalised COVID-19 patients in tertiary hospitals in Tanzania, 2021-2022: a retrospective cohort study. BMJ PUBLIC HEALTH 2024; 2:e000881. [PMID: 40018602 PMCID: PMC11816690 DOI: 10.1136/bmjph-2023-000881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/25/2024] [Indexed: 03/01/2025]
Abstract
Background There have been differential mortality rates from COVID-19 in different parts of the world. It is not clear whether the clinical presentation does also differ, thus the need for this study in a sub-Saharan African setting. The aim of this study was to describe the clinical manifestations and outcomes of patients diagnosed with COVID-19 in selected tertiary hospitals in Tanzania. Methods This was a retrospective analysis of hospitalised adults confirmed SAR-COV-2 infection in five tertiary-level hospitals in Tanzania. Data collected and analysed included sociodemographic, radiological and clinical characteristics of the patients as well as the outcome of the admission (discharge vs death). Results Out of 1387 COVID-19 patients, 52% were males. The median age was 60 years ((IQR)=(19-102)). The most common symptoms were dyspnoea (943,68%), cough (889, 64%), fever (597,43%) and fatigue (570, 41%). In-hospital mortality was (476, 34%). Mortality significantly increased with increasing age, being the most in age >90 years (aHR (95% CI)=4.4 (2.52 to 28.82), p=0.02). Other predictors of mortality were not possessing a health insurance, (aHR (95% CI)=3.7 (1.09 to 14.25), p=0.04); chest pain, (aHR (95% CI)=2.27 (1.36 to 4.13), p=0.03); HIV positivity, (aHR (95% CI)=3.9 (1.46 to 8.15), p=0.03); neutrophilia, (aHR (95% CI)=1.12 (1.01 to 2.65), p=0.03); no use of ivermectin, (aHR (95% CI)=1.21 (1.04 to 1.57), p=0.04) and non-use of steroids, (aHR (95% CI)=1.36 (1.18 to 2.78), p=0.04). The retrospective nature of this study which based on documented patients' records, with a large number of patients left out of the analysis due to missed data, this might in a way affect the results of the present study. Conclusion In-hospital mortality was 34%. The independent predictors of mortality were advanced age, HIV infection, no possession of a health insurance, chest pain, neutrophilia and no use of steroids or ivermectin.
Collapse
Affiliation(s)
- Elisha Fred Otieno Osati
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Grace Ambrose Shayo
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Raphael Z Sangeda
- Department of Pharmaceutical Microbiology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
| | - Tumaini Joseph Nagu
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Candida Moshiro
- Department of Internal Medicine, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania, United Republic of
| | - Naveeda Adams
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Athumani Ramadhani
- Department of Internal Medicine, Muhimbili National Hospital, Dar es Salaam, Tanzania, United Republic of
| | - Bahati Wajanga
- Department of Internal Medicine, Bugando Medical Centre, Mwanza, Tanzania, United Republic of
| | - Albert Muniko
- Department of Internal Medicine, Bugando Medical Centre, Mwanza, Tanzania, United Republic of
| | - Jeremiah Seni
- Department of Internal Medicine, Catholic University of Health and Allied Sciences Bugando, Mwanza, Tanzania, United Republic of
| | - Mary A Nicholaus
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre, Moshi, Kilimanjaro, Tanzania, United Republic of
| | - Gervas Nyaisonga
- Department of Internal Medicine, Mbeya Zonal Referral Hospital, Mbeya, Tanzania, United Republic of
| | - Christian Mbije
- Department of Internal Medicine, Mbeya Zonal Referral Hospital, Mbeya, Tanzania, United Republic of
| | - John Robson Meda
- Department of Internal Medicine, University of Dodoma, Dodoma, Tanzania, United Republic of
| | - Denis Rainer
- Department of Internal Medicine, Benjamin Mkapa Hospital, Dodoma, Tanzania, United Republic of
| | - Martha Elisande Nkya
- Community, Management and Development for Health, Dar es Salaam, Tanzania, United Republic of
| | - Paulo Mhame
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| | - Lucy Samwel
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| | - Liggyle Vumilia
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| | - Seif Shekalaghe
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| | - Kajiru G Kilonzo
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre, Moshi, Kilimanjaro, Tanzania, United Republic of
| | - Abel Makubi
- Ministry of Health, Dar as Salaam, Tanzania, United Republic of
| |
Collapse
|
57
|
Farias PCS, Cabral LP, Neves PAF, Januário CAB, Cordeiro BM, Silva Júnior WJDA, Baseggio C, Paiva Júnior SSL, Araújo PSRDE, Lorena VMBDE, Balbino VQ, Lima Neto RG. Genetic variant in the AGT gene (rs699-GG) is associated with severe COVID-19 in Brazilian patients. AN ACAD BRAS CIENC 2024; 96:e20240274. [PMID: 39630802 DOI: 10.1590/0001-3765202420240274] [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: 03/18/2024] [Accepted: 08/30/2024] [Indexed: 12/07/2024] Open
Abstract
The COVID-19 pandemic has been the largest pandemic of the past century, and various genetic factors have played a significant role in this context. This study aimed to analyze the frequency and association between specific SNPs rs3806268 (NLRP3), rs4925543 (NLRP3), rs12150220 (NLRP1), rs455060 (NLRC4), rs699 (AGT), rs1137101 (LEPR), and rs1801133 (MTHFR) and severe/critical outcomes in Brazilian patients with COVID-19. A total of 100 patients were included in the study, comprising 66 cases and 34 controls. DNA was extracted, sequenced, and genotyped via next-generation sequencing (NGS). For non-parametric data, the Mann-Whitney and Kruskal-Wallis tests were used. Fisher's test and multivariate logistic regression, considering AIC and BIC criteria, were employed for risk analysis. Odds Ratios (OR) were calculated, with significance set at p<0.05. Among the seven evaluated SNPs, only rs699-GG (AGT) (OR=8.07; p=0.04) was significantly associated with an increased risk of developing severe/critical COVID-19. Moreover, a borderline protective association was noted between rs1801133-GA (MTHFR) and the disease, although lacking statistical significance. In conclusion, the SNP rs699-GG (AGT) was associated with an increased risk of severe/critical COVID-19.
Collapse
Affiliation(s)
- Pablo C S Farias
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Leandro P Cabral
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Patrícia A F Neves
- Instituto Aggeu Magalhães (FIOCRUZ-PE), Departamento de Imunologia, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
- Universidade Federal de Pernambuco, Departamento de Medicina Tropical, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Caio A B Januário
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Beatriz M Cordeiro
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Wilson J DA Silva Júnior
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Carolina Baseggio
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Sérgio S L Paiva Júnior
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Paulo S R DE Araújo
- Universidade Federal de Pernambuco, Departamento de Medicina Tropical, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Virgínia Maria B DE Lorena
- Instituto Aggeu Magalhães (FIOCRUZ-PE), Departamento de Imunologia, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Valdir Q Balbino
- Universidade Federal de Pernambuco, Departamento de Genética, Av. da Engenharia, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Reginaldo G Lima Neto
- Universidade Federal de Pernambuco, Departamento de Medicina Tropical, Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| |
Collapse
|
58
|
Zhang J, Kim MH, Lee S, Park S. Integration of nanobiosensors into organ-on-chip systems for monitoring viral infections. NANO CONVERGENCE 2024; 11:47. [PMID: 39589620 PMCID: PMC11599699 DOI: 10.1186/s40580-024-00455-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
The integration of nanobiosensors into organ-on-chip (OoC) models offers a promising advancement in the study of viral infections and therapeutic development. Conventional research methods for studying viral infection, such as two-dimensional cell cultures and animal models, face challenges in replicating the complex and dynamic nature of human tissues. In contrast, OoC systems provide more accurate, physiologically relevant models for investigating viral infections, disease mechanisms, and host responses. Nanobiosensors, with their miniaturized designs and enhanced sensitivity, enable real-time, continuous, in situ monitoring of key biomarkers, such as cytokines and proteins within these systems. This review highlights the need for integrating nanobiosensors into OoC systems to advance virological research and improve therapeutic outcomes. Although there is extensive literature on biosensors for viral infection detection and OoC models for replicating infections, real integration of biosensors into OoCs for continuous monitoring remains unachieved. We discuss the advantages of nanobiosensor integration for real-time tracking of critical biomarkers within OoC models, key biosensor technologies, and current OoC systems relevant to viral infection studies. Additionally, we address the main technical challenges and propose solutions for successful integration. This review aims to guide the development of biosensor-integrated OoCs, paving the way for precise diagnostics and personalized treatments in virological research.
Collapse
Affiliation(s)
- Jiande Zhang
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Seulgi Lee
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Biophysics, Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
| |
Collapse
|
59
|
Shaker MM, Abdelghany AE, Elaraby NM. Expression Levels of PF4, ALOX12, ITGA2B, F131A in Pregnant COVID-19 Survivors. Biochem Genet 2024:10.1007/s10528-024-10958-4. [PMID: 39543003 DOI: 10.1007/s10528-024-10958-4] [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: 01/19/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024]
Abstract
COVID-19 is viral illness caused by SARS-CoV-2. The immediate complications of COVID-19 are well defined and associated with increased mortality. A global effort is required to determine its effects on implantation, fetal growth and labor. Post COVID-19 recovery period presents a further challenge regarding service provision, prevention, and management. To assess the expression of Platelet Factor 4 (PF4), Arachidonate 12-lipoxygenase (ALOX 12), Integrin alpha-IIb (ITGA2B) & Coagulation Factor XIII A Chain F13A1 in post-acute COVID-19 survivors pregnant women. Prospective case control study, conducted on 400 pregnant women. Case group consists of 200 singleton pregnancies who had recovered from COVID-19 since 4-6 weeks before conception. Control group consists of 200 singleton pregnancies with no history for COVID-19. Expression levels of ALOX12, PF4, ITGA2B, and F13A1genes were determined using quantitative reverse transcription polymerase chain reaction method (qRT-PCR). Expression levels of ALOX12, ITGA2B, and F13A1, were significantly higher in the patients group (3.82±9.6, 6.63±8.45, and 8.9±9.1, respectively) (p < 0.05) compared to those in the control group (1.0±6.0, 1.0±8.1, and 0.6±7.6, respectively). No significant difference in PF4 expression between patients and control group (p = 0.3). Results obtained from enrichment analysis have also supported the above findings. Relative expression levels of these candidate genes could be distinguished between post-acute COVID-19 survivors' pregnant women and control group, significant relative gene expression of ALOX12, ITGA2B, and F13A1 may be associated with an increased risk of placenta-mediated adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Mai M Shaker
- Prenatal Diagnosis and Fetal Medicine Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| | - Asia E Abdelghany
- Prenatal Diagnosis and Fetal Medicine Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Nesma M Elaraby
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
60
|
Aikwanich A, Eksombatchai D, Petnak T, Tassaneeyasin T, Boonsarngsuk V. Risk Factors for Secondary Organizing Pneumonia and Acute Fibrinous and Organizing Pneumonia in Patients with COVID-19 Pneumonia. Infect Drug Resist 2024; 17:5017-5026. [PMID: 39554470 PMCID: PMC11566205 DOI: 10.2147/idr.s481540] [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: 07/31/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose Secondary organizing pneumonia (OP) and acute fibrinous and organizing pneumonia (AFOP) are frequently observed in cases of COVID-19 pneumonia. Nevertheless, the identification of risk factors related to OP/AFOP and their impact on patient outcomes remain inadequately elucidated. Patients and Methods This retrospective study aimed to identify risk factors associated with OP/AFOP in patients with COVID-19 pneumonia and to compare clinical outcomes between patients with and without OP/AFOP. The study included hospitalized patients with COVID-19 pneumonia admitted between July 1 and September 30, 2021. Factors associated with OP/AFOP were identified using multivariable regression analysis. Additionally, a multivariable Cox proportional hazard model was used to evaluate the association of OP/AFOP with 90-day mortality. Results Among the 666 hospitalized patients with COVID-19 pneumonia, 53 (8%) developed OP/AFOP during their admission. When compared to patients younger than 50 years old, those aged 50-70 and over 70 years old exhibited an increased risk of developing OP/AFOP, with adjusted odds ratios (aOR) of 3.87 (95% CI, 1.24-12.11; P=0.02) and 5.74 (95% CI, 1.80-18.27; P=0.003), respectively. Other factors associated with OP/AFOP included a history of diabetes mellitus (aOR 2.37; 95% CI, 1.27-4.44; P=0.01) and patients with oxygen saturation at admission below 88% (aOR 4.52; 95% CI, 1.22-16.67; P=0.02). Furthermore, the presence of OP/AFOP was correlated with an increased risk of various complications, such as respiratory failure, acute kidney injury, secondary infections, pneumothorax, pneumomediastinum, and pulmonary embolism. Lastly, patients with OP/AFOP exhibited significantly higher 90-day mortality (adjusted hazard ratio 3.40; 95% CI, 1.68-6.92; P=0.001) compared to those without OP/AFOP. Conclusion We identified factors associated with an increased risk of OP/AFOP in patients with COVID-19 pneumonia, which included age ≥50 years, a history of DM, and hypoxemia on admission (SpO2 <88%). Furthermore, our study revealed that OP/AFOP was significantly linked to higher 90-day mortality.
Collapse
Affiliation(s)
- Alisa Aikwanich
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Dararat Eksombatchai
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Tananchai Petnak
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Tanapat Tassaneeyasin
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Viboon Boonsarngsuk
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
61
|
Song HW, Jo HY, Kim SC, Choi SS. Immunopathological markers and cell types linked to COVID-19 symptom manifestation. BMC Infect Dis 2024; 24:1237. [PMID: 39497098 PMCID: PMC11533414 DOI: 10.1186/s12879-024-10139-z] [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: 07/30/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Numerous studies have investigated the molecular properties that contribute to the symptoms of COVID-19, such as the virus's genetic makeup, its replication mechanisms, and how it interacts with host cells. However, identifying the immunopathological properties, such as the immune system's response, cytokine levels, and the presence of specific biomarkers, that are associated with the severity of the infection remains crucial for developing effective treatments and preventions. METHODS We analyzed blood protein factor profiles from 420 individuals to identify features differentiating between test-negative healthy, asymptomatic, and symptomatic individuals using statistical comparison and the least absolute shrinkage and selection operator (i.e., LASSO) algorithm. Additionally, we examined single-cell RNA sequencing data from 141 individuals to identify specific cell types associated with the COVID-19 symptoms. RESULTS Healthy individuals who tested negative had distinct blood protein factor levels compared to asymptomatic individuals. We identified two key protein factors, Serpin A10 and Complement C9, that differentiate between asymptomatic and symptomatic patients. Symptomatic patients showed lower levels of CD4+ T naïve, CD4+ T effector & memory, and CD8+ T naïve cells, along with higher levels of CD14+ classical monocytes compared to asymptomatic patients. Additionally, CD16+ non-classical monocytes, major producers of C1QA/B/C, appeared to contribute to the observed Complement C9 levels. CONCLUSIONS These findings advance our understanding of the immunopathological mechanisms underlying COVID-19 and may inform the development of targeted therapies and preventative measures. Future research should focus on further elucidating these mechanisms and exploring their potential clinical applications in managing COVID-19 severity.
Collapse
Affiliation(s)
- Ha Won Song
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, 24341, Korea
| | - Hye-Yeong Jo
- Division of Healthcare and Artificial Intelligence, Department of Precision Medicine, Korea National Institute of Health, Osong, 28159, Korea
| | - Sang Cheol Kim
- Division of Healthcare and Artificial Intelligence, Department of Precision Medicine, Korea National Institute of Health, Osong, 28159, Korea
| | - Sun Shim Choi
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, 24341, Korea.
| |
Collapse
|
62
|
Agrawal A, Bajaj S, Bhagat U, Chandna S, Arockiam AD, El Dahdah J, Haroun E, Gupta R, Shekhar S, Raj K, Nayar D, Bajaj D, Chaudhury P, Griffin BP, Wang TKM. Incidence, Predictors, and Outcomes of Venous and Arterial Thrombosis in COVID-19: A Nationwide Inpatient Analysis. Heart Lung Circ 2024; 33:1563-1573. [PMID: 38942623 DOI: 10.1016/j.hlc.2024.04.167] [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: 10/26/2023] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is known to increase the risk of venous thromboembolism (VTE) and arterial thromboembolism (ATE). However, the incidence, predictors, and outcomes of clinical thrombosis for inpatients with COVID-19 are not well known. This study aimed to enhance our understanding of clinical thrombosis in COVID-19, its associated factors, and mortality outcomes. METHOD Hospitalised adult (≥18 years of age) patients with COVID-19 in 2020 were retrospectively identified from the US National Inpatient Sample database. Clinical characteristics, incident VTE, ATE, and in-hospital mortality outcomes were recorded. Multivariable logistic regression was performed to identify clinical factors associated with thrombosis and in-hospital mortality in COVID-19 inpatients. RESULTS A total of 1,583,135 adult patients with COVID-19 in the year 2020 were identified from the National Inpatient Sample database; patients with thrombosis were 41% females with a mean age of 65.4 (65.1-65.6) years. The incidence of thrombosis was 6.1% (97,185), including VTE at 4.8% (76,125), ATE at 3.0% (47,790), and the in-hospital mortality rate was 13.4% (212,785). Patients with thrombosis were more likely to have respiratory symptoms of COVID-19 (76.7% vs 75%, p<0.001) compared with patients without thrombosis. The main factors associated with overall thrombosis, VTE, and ATE were paralysis, ventilation, solid tumours without metastasis, metastatic cancer, and acute liver failure. Although all thrombosis categories were associated with higher in-hospital mortality for COVID-19 inpatients in univariable analyses (p<0.001), they were not in multivariable analyses-thrombosis (odds ratio [OR] 1.24; 95% confidence interval [CI] 0.90-1.70; p=0.19), VTE (OR 0.70; 95% CI 0.52-1.00; p=0.05), and ATE (OR 1.07; 95% CI 0.92-1.25; p=0.36). CONCLUSIONS The association of COVID-19 with thrombosis and VTE increases with increasing severity of the COVID-19 disease. Risk stratification of thrombosis is crucial in COVID-19 patients to determine the necessity of thromboprophylaxis.
Collapse
Affiliation(s)
- Ankit Agrawal
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suryansh Bajaj
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Umesh Bhagat
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Sanya Chandna
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Aro Daniela Arockiam
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Joseph El Dahdah
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elio Haroun
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rahul Gupta
- Lehigh Valley Heart Institute, Lehigh Valley Health Network, Allentown, PA, USA
| | - Shashank Shekhar
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kavin Raj
- Division of Cardiology, Department of Medicine, University of California Riverside School of Medicine, Riverside, CA, USA
| | - Divya Nayar
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Divyansh Bajaj
- Department of Pulmonary, Critical Care, and Sleep Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Pulkit Chaudhury
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brian P Griffin
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tom Kai Ming Wang
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
63
|
Askey D, Smith A. Are Pre-Hospitalization ECG Abnormalities Associated With Increased Mortality in COVID-19 Patients? A Quantitative Systematic Literature Review. Ann Noninvasive Electrocardiol 2024; 29:e70016. [PMID: 39394768 PMCID: PMC11470194 DOI: 10.1111/anec.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/21/2024] [Accepted: 09/09/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND While COVID-19 is predominantly a respiratory disease, cardiovascular complications occur and are associated with worse outcomes. Electrocardiogram (ECG) abnormalities are frequently observed in hospitalized COVID-19 patients, some of which are associated with increased mortality. It is unclear whether ECG abnormalities occurring before hospitalization are associated with increased mortality. This quantitative systematic literature review aims to determine which ECG changes occurring before hospitalization are associated with mortality and discuss whether these findings can aid the assessment of patients and decision-making in the pre-hospital environment. METHODS A systematic search of the following digital databases was conducted: CINAL, PUBMED, MEDLINE, and Coronavirus Research Database. Eight cohort studies (primary papers) including COVID-19 patients with ECGs taken in the Emergency Department before hospitalization were selected for quantitative synthesis and results were obtained for the prevalence of ECG changes among survivors compared with non-survivors. Odds and hazard ratios for ECG abnormalities associated with mortality were also collected and compared. RESULTS Identification of ECG abnormalities on pre-hospitalization ECG is associated with increased mortality in COVID-19 patients. These ECG abnormalities include non-sinus rhythm, QTc prolongation, left bundle branch block, axis deviation, atrial fibrillation, atrial flutter, right ventricular strain patterns, ST segment changes, T wave abnormalities, and evidence of left ventricular hypertrophy. CONCLUSION Electrocardiogram assessment in the pre-hospital environment may be beneficial when assessing COVID-19 patients and could help identify patients at increased risk of mortality.
Collapse
Affiliation(s)
- Danielle Askey
- Hazardous Area Response Team Paramedic, South Western Ambulance Service NHS Foundation TrustNorth Bristol Operations CentreBristolUK
| | - Ann Smith
- Senior Lecturer in Health StatisticsUniversity of the West of EnglandBristolUK
| |
Collapse
|
64
|
Dunton J, Bierman-Macke K, Little T, Zuk N, Beyersdorfer N, Goade S, Johnson K, Stahl G, Arnce RD. A Retrospective Analysis of Thrombosis and COVID-19 Mortality in Rural Midwestern Population. Cureus 2024; 16:e74320. [PMID: 39717294 PMCID: PMC11666284 DOI: 10.7759/cureus.74320] [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: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
Background COVID-19 disease has caused a major global impact on health and mortality. This infection may predispose patients to thrombotic disease, caused by excessive inflammation, endothelial dysfunction, platelet activation, and stasis. In this study, we compared mortality rates in patients admitted to the hospital with the diagnosis of COVID-19, who also had the additional diagnosis of thrombosis with those who did not have thrombosis as an additional diagnosis. Methods This retrospective observational study compared mortality rates in patients admitted to the hospital with the diagnosis of COVID-19, with and without thrombosis, as well as those patients admitted to the hospital with the diagnosis of thrombosis who did not have COVID-19. The diagnoses were verified using International Classification of Diseases Tenth Revision (ICD-10) codes, a standard among electronic medical records (EMR). The data were taken from the EMR at Freeman Health System in Joplin and Neosho, Missouri, from April 2020 to December 2021. This patient population is representative of not only Southwest Missouri but also the four-state area, including Oklahoma, Arkansas, and Kansas. The ICD-10 codes were used to separate the patient population into three main groups as follows: patients diagnosed with COVID-19 without thrombosis, patients diagnosed with thrombosis without COVID-19, and patients diagnosed with both COVID-19 and thrombosis. These three categories were then subdivided by age and biological sex. Sample proportions were completed using Wald's method, and the two-sample proportion summary hypothesis test with confidence intervals was used for the proportion difference. Results A total of 3,094 patients were included in the study population. Excluded from the study were patients who were previously admitted to a hospital for COVID-19 and duplicate admissions. The mortality rate was highest (0.4714) in patients concurrently diagnosed with COVID-19 and thrombosis (Population 1 {P1}), followed by patients diagnosed with COVID-19 without thrombosis (Population 2 {P2}, 0.1187). Patients diagnosed with thrombosis without COVID-19 (Population 3 {P3}, 0.1216) had the lowest mortality. Two sample proportion hypothesis tests determined confidence intervals (CI) for mortality risk comparing P3 to P1 (95% CI: 0.2888-0.4108, p<0.0001) and P2 to P1 (95% CI: 0.2919-0.4135, p<0.0001). Discussion In this rural, Midwestern population, patients admitted to the hospital with the diagnosis of COVID-19 and thrombosis had significantly increased mortality rates compared to patients admitted with the diagnosis of COVID-19 or thrombosis alone. Conclusion The data from this study indicated that individuals diagnosed with both COVID-19 and thrombosis had a higher likelihood of mortality compared to those diagnosed with COVID-19 without thrombosis and those diagnosed with thrombosis without COVID-19. This information could assist physicians in determining treatment plans for patients diagnosed with COVID-19 and a secondary complication of thrombosis.
Collapse
Affiliation(s)
- John Dunton
- Emergency Medicine, Kansas City University, Kansas City, USA
| | | | - Taylor Little
- Emergency Medicine, Kansas City University, Kansas City, USA
| | - Nicholas Zuk
- Emergency Medicine, Kansas City University, Kansas City, USA
| | | | - Scott Goade
- Clinical Pharmacy, Freeman Health System, Joplin, USA
- College of Medicine, Kansas City University, Joplin, USA
| | - Kerry Johnson
- Mathematics, Missouri Southern State University, Joplin, USA
| | - Greg Stahl
- Statistics, Freeman Health System, Joplin, USA
| | - Robert D Arnce
- College of Osteopathic Medicine, Kansas City University of Medicine and Biosciences, Joplin, USA
| |
Collapse
|
65
|
Shyama S, Vardhan H, Ojha VS, Biswas R, Ahmad S, Kumar A. Factors Affecting Mortality in COVID-19 Patients with Pre-Existing Chronic Kidney Disease. Indian J Nephrol 2024; 34:643-645. [PMID: 39649310 PMCID: PMC11619041 DOI: 10.25259/ijn_67_2024] [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: 02/08/2024] [Accepted: 03/17/2024] [Indexed: 12/10/2024] Open
Abstract
The emergence of COVID-19 triggered a global health crisis, sparking concerns within the medical community about its interaction with chronic kidney disease (CKD) and the heightened vulnerability of individuals with compromised renal function to severe viral infection and mortality. This retrospective study encompassed all adult patients with laboratory-confirmed COVID-19 and pre-existing CKD admitted between May 2020 and May 2023. Their demographic data, relevant clinical parameters, and laboratory values were collected. Kaplan-Meier curve analysis and Log Rank test were employed to compare survival times between CKD patients and those developing acute kidney injury (AKI), while Cox regression analyses were conducted to pinpoint factors influencing the hazard of a fatal outcome. The study, involving 150 COVID-19 patient records with pre-existing CKD, revealed that male gender, advanced age, requirement for invasive ventilation, and elevated levels of inflammatory markers such as total leukocyte count, lactate dehydrogenase, C-reactive protein, D-dimer, and IL-6 significantly increased the risk of death. These findings underscore the necessity for tailored care and meticulous management in COVID-19 patients with coexisting CKD, emphasizing the importance of addressing factors such as gender, age, and inflammatory status to mitigate mortality risks effectively.
Collapse
Affiliation(s)
- Shyama Shyama
- Department of General Medicine, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Harsh Vardhan
- Department of Nephrology, Patna Medical College, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Vishnu Shankar Ojha
- Department of General Medicine, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Ratnadeep Biswas
- Department of General Medicine, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Shamshad Ahmad
- Department of Community and Family Medicine, All India Institute of Medical Sciences, Patna, Bihar, India
| | - Amit Kumar
- Department of Nephrology, All India Institute of Medical Sciences, Patna, Bihar, India
| |
Collapse
|
66
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
67
|
Guarienti FA, Gonçalves JIB, Gonçalves JB, Antônio Costa Xavier F, Marinowic D, Machado DC. COVID-19: a multi-organ perspective. Front Cell Infect Microbiol 2024; 14:1425547. [PMID: 39492990 PMCID: PMC11527788 DOI: 10.3389/fcimb.2024.1425547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/18/2024] [Indexed: 11/05/2024] Open
Abstract
In this mini review, we explore the complex network of inflammatory reactions incited by SARS-CoV-2 infection, which extends its reach well beyond the respiratory domain to influence various organ systems. Synthesizing existing literature, it elucidates how the hyperinflammation observed in COVID-19 patients affects multiple organ systems leading to physiological impairments that can persist over long after the resolution of infection. By exploring the systemic manifestations of this inflammatory cascade, from acute respiratory distress syndrome (ARDS) to renal impairment and neurological sequelae, the review highlights the profound interplay between inflammation and organ dysfunction. By synthesizing recent research and clinical observations, this mini review aims to provide an overview of the systemic interactions and complications associated with COVID-19, underscoring the need for an integrated approach to treatment and management. Understanding these systemic effects is crucial for improving patient outcomes and preparing for future public health challenges.
Collapse
Affiliation(s)
- Fabiana Amaral Guarienti
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Júlia Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Daniel Marinowic
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Denise Cantarelli Machado
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| |
Collapse
|
68
|
da Silva AMV, Machado TL, Nascimento RDS, Rodrigues MPMD, Coelho FS, Tubarão LN, da Rosa LC, Bayma C, Rocha VP, Frederico ABT, Silva J, Cunha DRDADBE, de Souza AF, de Souza RBG, Barros CA, Fiscina DDS, Ribeiro LCP, de Carvalho CAM, da Silva BJD, Muller R, Azamor T, Melgaço JG, Gonçalves RB, Ano Bom APD. Immunomodulatory effect of bovine lactoferrin during SARS-CoV-2 infection. Front Immunol 2024; 15:1456634. [PMID: 39483459 PMCID: PMC11524939 DOI: 10.3389/fimmu.2024.1456634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/09/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction Lactoferrin (Lf) is an important immunomodulator in infections caused by different agents. During SARS-CoV-2 infection, Lf can hinder or prevent virus access to the intracellular environment. Severe cases of COVID-19 are related to increased production of cytokines, accompanied by a weak type 1 interferon response. Methods We investigated the influence of bovine Lf (bLf) in the immune response during SARS-CoV-2 infection in vitro and in vivo assays. Results Our results show a strong binding between bLf and TLR4/NF-κB in silico, as well as an increase in mRNA expression of these genes in peripheral blood mononuclear cells (PBMCs) treated with bLf. Furthermore, the treatment increased TLR4/TLR9 mRNA expression in infected K18-hACE2 mouse blood, indicating an activation of innate response. Our results show that, when bLf was added, a reduction in the NK cell population was found, presenting a similar effect on PD-1 in TCD4+ and TCD8+ cells. In the culture supernatant of PBMCs from healthy participants, bLf decreased IL-6 levels and increased CCL5 in COVID-19 participants. In addition, K18-hACE2 mice infected and treated with bLf presented an increase of serum pro-inflammatory markers (GM-CSF/IL-1β/IL-2) and upregulated mRNA expression of IL1B and IL6 in the lung tissue. Furthermore, bLf treatment was able to restore FTH1 levels in brain tissue. Discussion The data indicate that bLf can be part of a therapeutic strategy to promote the immunomodulation effect, leading to homeostasis during COVID-19.
Collapse
Affiliation(s)
- Andrea Marques Vieira da Silva
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Thiago Lazari Machado
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Ryann de Souza Nascimento
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Miguel Pires Medeiros Diniz Rodrigues
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Felipe Soares Coelho
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Luciana Neves Tubarão
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Lorenna Carvalho da Rosa
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Camilla Bayma
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Vanessa Pimenta Rocha
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Ana Beatriz Teixeira Frederico
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Jane Silva
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Danielle Regina de Almeida de Brito e Cunha
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Alessandro Fonseca de Souza
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | | | - Caroline Augusto Barros
- Departamento de Bioquímica, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle da Silva Fiscina
- Departamento de Bioquímica, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Claudio Pereira Ribeiro
- Laboratório de Pesquisa Multiusuário 04 (LPM-04) Hospital Universitário Graffée Guinle, HUGG/EBSERH, Rio de Janeiro, Brazil
| | | | - Bruno Jorge Duque da Silva
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Rodrigo Muller
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Tamiris Azamor
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Juliana Gil Melgaço
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Rafael Braga Gonçalves
- Departamento de Bioquímica, Instituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula Dinis Ano Bom
- Departamento de Desenvolvimento Experimental e Pré-Clinico (DEDEP), Instituto de Tecnologia em Imunobiológico, Bio-Manguinhos, Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
69
|
Carnevale R, Nocella C, Marocco R, Zuccalà P, Carraro A, Picchio V, Oliva A, Cangemi R, Miele MC, De Angelis M, Cancelli F, Casciaro GE, Cristiano L, Pignatelli P, Frati G, Venditti M, Pugliese F, Mastroianni CM, Violi F, Ridola L, Del Borgo C, Palmerio S, Valenzi E, Carnevale R, Alvaro D, Lichtner M, Cardinale V. Association Between NOX2-Mediated Oxidative Stress, Low-Grade Endotoxemia, Hypoalbuminemia, and Clotting Activation in COVID-19. Antioxidants (Basel) 2024; 13:1260. [PMID: 39456513 PMCID: PMC11505442 DOI: 10.3390/antiox13101260] [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: 09/25/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Low-grade endotoxemia by lipopolysaccharide (LPS) has been detected in COVID-19 and could favor thrombosis via eliciting a pro-inflammatory and pro-coagulant state. The aim of this study was to analyze the mechanism accounting for low-grade endotoxemia and its relationship with oxidative stress and clotting activation thrombosis in COVID-19. We measured serum levels of sNOX2-dp, zonulin, LPS, D-dimer, and albumin in 175 patients with COVID-19, classified as having or not acute respiratory distress syndrome (ARDS), and 50 healthy subjects. Baseline levels of sNOX2-dp, LPS, zonulin, D-dimer, albumin, and hs-CRP were significantly higher in COVID-19 compared to controls. In COVID-19 patients with ARDS, sNOX2-dp, LPS, zonulin, D-dimer, and hs-CRP were significantly higher compared to COVID-19 patients without ARDS. Conversely, concentration of albumin was lower in patients with ARDS compared with those without ARDS and inversely associated with LPS. In the COVID-19 cohort, the number of patients with ARDS progressively increased according to sNOX2-dp and LPS quartiles; a significant correlation between LPS and sNOX2-dp and LPS and D-dimer was detected in COVID-19. In a multivariable logistic regression model, LPS/albumin levels and D-dimer predicted thrombotic events. In COVID-19 patients, LPS is significantly associated with a hypercoagulation state and disease severity. In vitro, LPS can increase endothelial oxidative stress and coagulation biomarkers that were reduced by the treatment with albumin. In conclusion, impaired gut barrier permeability, increased NOX2 activation, and low serum albumin may account for low-grade endotoxemia and may be implicated in thrombotic events in COVID-19.
Collapse
Affiliation(s)
- Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.E.C.); (G.F.)
- IRCCS Neuromed, 86077 Pozzilli, Italy;
| | - Cristina Nocella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (P.P.); (F.V.)
| | - Raffaella Marocco
- Infectious Diseases Unit, Santa Maria (SM) Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy; (R.M.); (P.Z.); (C.D.B.)
| | - Paola Zuccalà
- Infectious Diseases Unit, Santa Maria (SM) Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy; (R.M.); (P.Z.); (C.D.B.)
| | - Anna Carraro
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (A.C.); (A.O.); (M.C.M.); (M.D.A.); (F.C.); (M.V.); (C.M.M.); (M.L.)
| | | | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (A.C.); (A.O.); (M.C.M.); (M.D.A.); (F.C.); (M.V.); (C.M.M.); (M.L.)
| | - Roberto Cangemi
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.C.); (L.R.); (D.A.); (V.C.)
| | - Maria Claudia Miele
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (A.C.); (A.O.); (M.C.M.); (M.D.A.); (F.C.); (M.V.); (C.M.M.); (M.L.)
| | - Massimiliano De Angelis
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (A.C.); (A.O.); (M.C.M.); (M.D.A.); (F.C.); (M.V.); (C.M.M.); (M.L.)
| | - Francesca Cancelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (A.C.); (A.O.); (M.C.M.); (M.D.A.); (F.C.); (M.V.); (C.M.M.); (M.L.)
| | - Giovanni Enrico Casciaro
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.E.C.); (G.F.)
| | | | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (P.P.); (F.V.)
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (G.E.C.); (G.F.)
- IRCCS Neuromed, 86077 Pozzilli, Italy;
| | - Mario Venditti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (A.C.); (A.O.); (M.C.M.); (M.D.A.); (F.C.); (M.V.); (C.M.M.); (M.L.)
| | - Francesco Pugliese
- Department of General Surgery and Surgical Specialty, Sapienza University of Rome, 00161 Rome, Italy;
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (A.C.); (A.O.); (M.C.M.); (M.D.A.); (F.C.); (M.V.); (C.M.M.); (M.L.)
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (P.P.); (F.V.)
| | - Lorenzo Ridola
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.C.); (L.R.); (D.A.); (V.C.)
| | - Cosmo Del Borgo
- Infectious Diseases Unit, Santa Maria (SM) Goretti Hospital, Sapienza University of Rome, 04100 Latina, Italy; (R.M.); (P.Z.); (C.D.B.)
| | - Silvia Palmerio
- Centro Ricerche Cliniche di Verona (CRC), 37134 Verona, Italy;
| | | | - Rita Carnevale
- Corso di Laurea di I Livello in Infermieristica, Università Sapienza di Roma–Polo Pontino–Sede di Terracina, 04019 Terracina, Italy;
| | - Domenico Alvaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.C.); (L.R.); (D.A.); (V.C.)
| | - Miriam Lichtner
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (A.C.); (A.O.); (M.C.M.); (M.D.A.); (F.C.); (M.V.); (C.M.M.); (M.L.)
| | - Vincenzo Cardinale
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.C.); (L.R.); (D.A.); (V.C.)
| |
Collapse
|
70
|
Lian H, Cai H, Zhang H, Zhang Y, Wang X. Inflammation, immunity and biomarkers in procoagulant responses of critically ill patients. Am J Transl Res 2024; 16:5797-5812. [PMID: 39544782 PMCID: PMC11558399 DOI: 10.62347/edar9565] [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: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024]
Abstract
Understanding the pathobiology of critical illness is essential for patients' prognosis. Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. As part of the host response, procoagulant responses, one of the most primitive reactions in biology, start at the very beginning of diseases and can be monitored throughout the process. Currently, we can achieve near-complete monitoring of the coagulation process, and procoagulant responses serve as indicators of the severity of host response in critically ill patients. However, the rapid interpretation of the complex results of various biomarkers remains a challenge for many clinicians. The indicators commonly used for coagulation assessment are complex, typically divided into three categories for clarity: process index, functional index, and outcome index. Monitoring and understanding these indicators can help manage procoagulant responses. The intervention of procoagulant response should be part of the bundle therapy, alongside the treatment for primary disease, management for hemodynamics, and controlling for host response. Early intervention for procoagulant response mainly includes anti-inflammation, antiplatelet and anticoagulant therapy, as well as management of primary disease. In this review, we systemically introduce the onset, assessment and intervention of procoagulant response.
Collapse
Affiliation(s)
- Hui Lian
- Department of Health Care, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| | - Huacong Cai
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| | - Hongmin Zhang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| | - Yan Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 100730, China
| |
Collapse
|
71
|
Sultana R, Kamihira M. Multifaceted Heparin: Diverse Applications beyond Anticoagulant Therapy. Pharmaceuticals (Basel) 2024; 17:1362. [PMID: 39459002 PMCID: PMC11510354 DOI: 10.3390/ph17101362] [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: 09/17/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Heparin, a naturally occurring polysaccharide, has fascinated researchers and clinicians for nearly a century due to its versatile biological properties and has been used for various therapeutic purposes. Discovered in the early 20th century, heparin has been a key therapeutic anticoagulant ever since, and its use is now implemented as a life-saving pharmacological intervention in the management of thrombotic disorders and beyond. In addition to its known anticoagulant properties, heparin has been found to exhibit anti-inflammatory, antiviral, and anti-tumorigenic activities, which may lead to its widespread use in the future as an essential drug against infectious diseases such as COVID-19 and in various medical treatments. Furthermore, recent advancements in nanotechnology, including nano-drug delivery systems and nanomaterials, have significantly enhanced the intrinsic biofunctionalities of heparin. These breakthroughs have paved the way for innovative applications in medicine and therapy, expanding the potential of heparin research. Therefore, this review aims to provide a creation profile of heparin, space for its utilities in therapeutic complications, and future characteristics such as bioengineering and nanotechnology. It also discusses the challenges and opportunities in realizing the full potential of heparin to improve patient outcomes and elevate therapeutic interventions.
Collapse
Affiliation(s)
- Razia Sultana
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
- Department of Biotechnology and Genetic Engineering, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan;
| |
Collapse
|
72
|
Babkina AS, Pisarev MV, Grechko AV, Golubev AM. Arterial Thrombosis in Acute Respiratory Infections: An Underestimated but Clinically Relevant Problem. J Clin Med 2024; 13:6007. [PMID: 39408067 PMCID: PMC11477565 DOI: 10.3390/jcm13196007] [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: 09/02/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
During the COVID-19 pandemic, there was increased interest in the issue of thrombotic complications of acute respiratory infections. Clinical reports and pathological studies have revealed that thrombus formation in COVID-19 may involve the venous and arterial vasculature. As thrombotic complications of infectious respiratory diseases are increasingly considered in the context of COVID-19, the fact that thrombosis in lung diseases of viral and bacterial etiology was described long before the pandemic is overlooked. Pre-pandemic studies show that bacterial and viral respiratory infections are associated with an increased risk of thrombotic complications such as myocardial infarction, ischemic stroke, pulmonary embolism, and other critical illnesses caused by arterial and venous thrombosis. This narrative review article aims to summarize the current evidence regarding thrombotic complications and their pathogenesis in acute lower respiratory tract infections.
Collapse
Affiliation(s)
- Anastasiya S. Babkina
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia; (M.V.P.); (A.V.G.); (A.M.G.)
| | | | | | | |
Collapse
|
73
|
Peluso MJ, Deeks SG. Mechanisms of long COVID and the path toward therapeutics. Cell 2024; 187:5500-5529. [PMID: 39326415 PMCID: PMC11455603 DOI: 10.1016/j.cell.2024.07.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 09/28/2024]
Abstract
Long COVID, a type of post-acute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (PASC) defined by medically unexplained symptoms following infection with SARS-CoV-2, is a newly recognized infection-associated chronic condition that causes disability in some people. Substantial progress has been made in defining its epidemiology, biology, and pathophysiology. However, there is no cure for the tens of millions of people believed to be experiencing long COVID, and industry engagement in developing therapeutics has been limited. Here, we review the current state of knowledge regarding the biology and pathophysiology of long COVID, focusing on how the proposed mechanisms explain the physiology of the syndrome and how they provide a rationale for the implementation of a broad experimental medicine and clinical trials agenda. Progress toward preventing and curing long COVID and other infection-associated chronic conditions will require deep and sustained investment by funders and industry.
Collapse
Affiliation(s)
- Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | - Steven G Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
74
|
Stephen IR, Suman FR, Balasubramanian J, Shanmugam SG, Mani R. D-dimer as a Predictor of ICU Admission and Mortality in COVID-19 Patients: Insights From a Two-Year Retrospective Study From a Tertiary Care Center in South India. Cureus 2024; 16:e70682. [PMID: 39493094 PMCID: PMC11529902 DOI: 10.7759/cureus.70682] [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: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced pneumonitis results in a prothrombotic and hypercoagulable state. Prognostic indicators are crucial for identifying patients at risk of complications. D-dimer, a degradation product of cross-linked fibrin, is a specific marker for thrombosis. Elevated D-dimer levels have been strongly correlated with poor prognosis and increased severity of illness in COVID-19 patients. Given D-dimer's eight-hour half-life, periodic measurement is necessary to track disease progression. This study aimed to analyze and derive threshold and peak D-dimer values to predict outcomes in COVID-19 patients, comparing those treated in isolation wards to those requiring intensive care. Methods This two-year retrospective observational study included patients above 18 years with confirmed COVID-19. Patients were categorized into those treated in isolation wards and those admitted to the intensive care unit (ICU). Based on the outcome, they were further divided into survivors and non-survivors. Demographic and outcome-related data were collected from the hospital's laboratory information system. Serial D-dimer measurements were taken at eight time points. Statistical analysis was performed using the Mann-Whitney test for laboratory values and the chi-square test for demographic data. Receiver operating characteristic (ROC) curve analysis was utilized to derive critical D-dimer values. The area under the curve (AUC) was calculated for initial and peak D-dimer values. Results Of 2.149 patients with confirmed COVID-19, 811 (38%) presented with elevated D-dimer levels. ICU admission was required for 239 patients, either due to direct admission or worsening conditions. An initial D-dimer value of ≥0.93 mg/L FEU indicated the need for ICU admission, while a peak D-dimer value of 5.65 mg/L FEU predicted mortality. The AUC for the initial D-dimer was 0.60 (95% CI: 0.55-0.64), indicating moderate discriminatory power. The AUC for the peak D-dimer was 0.58 (95% CI: 0.54-0.62), suggesting lower predictive accuracy for peak values. Sensitivity was high for both initial (0.925) and peak (0.960) D-dimer values, although specificity was lower, especially for the peak D-dimer (0.486), resulting in a higher rate of false positives. Among the ICU patients, the age range was 27-97 years, with a mean of 53.5 years. Males were more affected than females (71% vs. 29%), with a male-to-female ratio of 1.4:1. Of the ICU patients, 64.8% recovered, while 35.2% succumbed to the disease. Younger patients (mean age: 50.5 ± 12 years) recovered faster than older patients (mean age: 64 ± 16 years), with a significant difference in recovery time (p < 0.001). Gender did not significantly impact outcomes (p = 0.743). Survivors spent less time in the ICU (3-7 days) compared to non-survivors (4-14 days) (p = 0.041). Conclusion Serial D-dimer monitoring is essential for predicting outcomes and guiding treatment in COVID-19 patients. Initial and peak D-dimer values can help identify patients requiring intensive care and those at risk of mortality, allowing for timely interventions. D-dimer levels should be integrated into routine clinical assessments for managing COVID-19 patients.
Collapse
Affiliation(s)
| | - Febe Renjitha Suman
- Pathology, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | | | | | - Rajkumar Mani
- Internal Medicine, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| |
Collapse
|
75
|
Kole C, Stefanou Ε, Karvelas N, Schizas D, Toutouzas KP. Acute and Post-Acute COVID-19 Cardiovascular Complications: A Comprehensive Review. Cardiovasc Drugs Ther 2024; 38:1017-1032. [PMID: 37209261 PMCID: PMC10199303 DOI: 10.1007/s10557-023-07465-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 05/22/2023]
Abstract
PURPOSE OF REVIEW The risk of cardiovascular complications due to SARS-CoV-2 are significantly increased within the first 6 months of the infection. Patients with COVID-19 have an increased risk of death, and there is evidence that many may experience a wide range of post-acute cardiovascular complications. Our work aims to provide an update on current clinical aspects of diagnosis and treatment of cardiovascular manifestations during acute and long-term COVID-19. RECENT FINDINGS SARS-CoV-2 has been shown to be associated with increased incidence of cardiovascular complications such as myocardial injury, heart failure, and dysrhythmias, as well as coagulation abnormalities not only during the acute phase but also beyond the first 30 days of the infection, associated with high mortality and poor outcomes. Cardiovascular complications during long-COVID-19 were found regardless of comorbidities such as age, hypertension, and diabetes; nevertheless, these populations remain at high risk for the worst outcomes during post-acute COVID-19. Emphasis should be given to the management of these patients. Treatment with low-dose oral propranolol, a beta blocker, for heart rate management may be considered, since it was found to significantly attenuate tachycardia and improve symptoms in postural tachycardia syndrome, while for patients on ACE inhibitors or angiotensin-receptor blockers (ARBs), under no circumstances should these medications be withdrawn. In addition, in patients at high risk after hospitalization due to COVID-19, thromboprophylaxis with rivaroxaban 10 mg/day for 35 days improved clinical outcomes compared with no extended thromboprophylaxis. In this work we provide a comprehensive review on acute and post-acute COVID-19 cardiovascular complications, symptomatology, and pathophysiology mechanisms. We also discuss therapeutic strategies for these patients during acute and long-term care and highlight populations at risk. Our findings suggest that older patients with risk factors such as hypertension, diabetes, and medical history of vascular disease have worse outcomes during acute SARS-CoV-2 infection and are more likely to develop cardiovascular complications during long-COVID-19.
Collapse
Affiliation(s)
- Christo Kole
- Cardiology Department, Sismanoglio General Hospital of Attica, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Εleni Stefanou
- Artificial Kidney Unit, General Hospital of Messinia, Kalamata, Greece
| | - Nikolaos Karvelas
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | | |
Collapse
|
76
|
Furman S, Green K, Lane TE. COVID-19 and the impact on Alzheimer's disease pathology. J Neurochem 2024; 168:3415-3429. [PMID: 37850241 PMCID: PMC11024062 DOI: 10.1111/jnc.15985] [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: 08/18/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has rapidly escalated into a global pandemic that primarily affects older and immunocompromised individuals due to underlying clinical conditions and suppressed immune responses. Furthermore, COVID-19 patients exhibit a spectrum of neurological symptoms, indicating that COVID-19 can affect the brain in a variety of manners. Many studies, past and recent, suggest a connection between viral infections and an increased risk of neurodegeneration, raising concerns about the neurological effects of COVID-19 and the possibility that it may contribute to Alzheimer's disease (AD) onset or worsen already existing AD pathology through inflammatory processes given that both COVID-19 and AD share pathological features and risk factors. This leads us to question whether COVID-19 is a risk factor for AD and how these two conditions might influence each other. Considering the extensive reach of the COVID-19 pandemic and the devastating impact of the ongoing AD pandemic, their combined effects could have significant public health consequences worldwide.
Collapse
Affiliation(s)
- Susana Furman
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Kim Green
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
| | - Thomas E. Lane
- Department of Neurobiology & Behavior, School of Biological Sciences, University of California, Irvine 92697
- Department of Molecular Biology & Biochemistry, School of Biological Sciences, University of California, Irvine 92697, USA
- Center for Virus Research, University of California, Irvine 92697, USA
| |
Collapse
|
77
|
He Q, Wei Y, Qian Y, Zhong M. Pathophysiological dynamics in the contact, coagulation, and complement systems during sepsis: Potential targets for nafamostat mesilate. JOURNAL OF INTENSIVE MEDICINE 2024; 4:453-467. [PMID: 39310056 PMCID: PMC11411436 DOI: 10.1016/j.jointm.2024.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/17/2024] [Accepted: 02/07/2024] [Indexed: 09/25/2024]
Abstract
Sepsis is a life-threatening syndrome resulting from a dysregulated host response to infection. It is the primary cause of death in the intensive care unit, posing a substantial challenge to human health and medical resource allocation. The pathogenesis and pathophysiology of sepsis are complex. During its onset, pro-inflammatory and anti-inflammatory mechanisms engage in intricate interactions, possibly leading to hyperinflammation, immunosuppression, and long-term immune disease. Of all critical outcomes, hyperinflammation is the main cause of early death among patients with sepsis. Therefore, early suppression of hyperinflammation may improve the prognosis of these patients. Nafamostat mesilate is a serine protease inhibitor, which can inhibit the activation of the complement system, coagulation system, and contact system. In this review, we discuss the pathophysiological changes occurring in these systems during sepsis, and describe the possible targets of the serine protease inhibitor nafamostat mesilate in the treatment of this condition.
Collapse
Affiliation(s)
- Qiaolan He
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yilin Wei
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiqi Qian
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public Health, Fudan University, Shanghai, China
| |
Collapse
|
78
|
Gracia Aznar A, Moreno Egea F, Gracia Banzo R, Gutierrez R, Rizo JM, Rodriguez-Ledo P, Nerin I, Regidor PA. Pro-Resolving Inflammatory Effects of a Marine Oil Enriched in Specialized Pro-Resolving Mediators (SPMs) Supplement and Its Implication in Patients with Post-COVID Syndrome (PCS). Biomedicines 2024; 12:2221. [PMID: 39457534 PMCID: PMC11505212 DOI: 10.3390/biomedicines12102221] [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: 07/28/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVES This study aimed to evaluate the eicosanoid and pro-resolutive parameters in patients with Post-COVID Syndrome (PCS) during a 12-week supplementation with a marine oil enriched in specialized pro-resolving mediators (SPMs). PATIENT AND METHODS This study was conducted on 53 adult patients with PCS. The subjects included must have had a positive COVID-19 test (PCR, fast antigen test, or serologic test) and persistent symptoms related to COVID-19 at least 12 weeks before their enrolment in the study. The following parameters were evaluated: polyunsaturated fatty acids EPA, DHA, ARA, and DPA; specialized pro-resolving mediators (SPMs), 17-HDHA, 18-HEPE, 14-HDHA, resolvins, maresins, protectins, and lipoxins. The eicosanoids group included prostaglandins, thromboxanes, and leukotrienes. The development of the clinical symptoms of fatigue and dyspnea were evaluated using the Fatigue Severity Scale (FSS) and the Modified Medical Research Council (mMRC) Dyspnea Scale. Three groups with different intake amounts were evaluated (daily use of 500 mg, 1500 mg, and 3000 mg) and compared to a control group not using the product. RESULTS In the serum from patients with PCS, an increase in 17-HDHA, 18-HEPE, and 14-HDHA could be observed, and a decrease in the ratio between the pro-inflammatory and pro-resolutive lipid mediators was detected; both differences were significant (p < 0.05). There were no differences found between the three treatment groups. Fatigue and dyspnea showed a trend of improvement after supplementation in all groups. CONCLUSIONS A clear enrichment in the serum of the three monohydroxylated SPMs could be observed at a dosage of 500 mg per day. Similarly, a clear improvement in fatigue and dyspnea was observed with this dosage.
Collapse
Affiliation(s)
- Asun Gracia Aznar
- Sociedad Española de Médicos Generales y de Familia (SEMG), 28005 Madrid, Spain; (A.G.A.); (P.R.-L.)
| | | | - Rafael Gracia Banzo
- Solutex GC SL, Parque Empresarial Utebo, Avda. Miguel Servet nº 81, 50180 Utebo, Spain;
| | - Rocio Gutierrez
- OTC Chemo, Manuel Pombo Angulo 28-4th Floor, 28050 Madrid, Spain; (R.G.); (J.M.R.)
| | - Jose Miguel Rizo
- OTC Chemo, Manuel Pombo Angulo 28-4th Floor, 28050 Madrid, Spain; (R.G.); (J.M.R.)
| | - Pilar Rodriguez-Ledo
- Sociedad Española de Médicos Generales y de Familia (SEMG), 28005 Madrid, Spain; (A.G.A.); (P.R.-L.)
| | - Isabel Nerin
- Directora de la Cátedra SEMG-Estilos de Vida Unidad de Tabaquismo FMZ Profª Dpto. Medicina, Psiquiatría y Dermatología Facultad de Medicina, Universidad de Zaragoza, 50009 Zaragoza, Spain;
| | | |
Collapse
|
79
|
Delpino MV, Quarleri J. Aging mitochondria in the context of SARS-CoV-2: exploring interactions and implications. FRONTIERS IN AGING 2024; 5:1442323. [PMID: 39380657 PMCID: PMC11458564 DOI: 10.3389/fragi.2024.1442323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has presented global challenges with a diverse clinical spectrum, including severe respiratory complications and systemic effects. This review explores the intricate relationship between mitochondrial dysfunction, aging, and obesity in COVID-19. Mitochondria are vital for cellular energy provision and resilience against age-related macromolecule damage accumulation. They manage energy allocation in cells, activating adaptive responses and stress signals such as redox imbalance and innate immunity activation. As organisms age, mitochondrial function diminishes. Aging and obesity, linked to mitochondrial dysfunction, compromise the antiviral response, affecting the release of interferons, and worsening COVID-19 severity. Furthermore, the development of post-acute sequelae of SARS-CoV-2 infection (PASC), also known as long COVID has been associated with altered energy metabolism, and chronic immune dysregulation derived from mitochondrial dysfunction. Understanding the interplay between mitochondria, aging, obesity, and viral infections provides insights into COVID-19 pathogenesis. Targeting mitochondrial health may offer potential therapeutic strategies to mitigate severe outcomes and address long-term consequences in infected individuals.
Collapse
|
80
|
Ebrahim Babai M, Kabiri A, Movahedi M, Ghahiri A, Hajhashemi M, Dehghan M. Evaluation of the Relationship between Early Clinical Manifestations and Changes in Biochemical, Inflammatory, and Coagulation Parameters and the Prognosis of Pregnant Women with COVID-19 Admitted to the ICU. Adv Biomed Res 2024; 13:76. [PMID: 39512403 PMCID: PMC11542693 DOI: 10.4103/abr.abr_257_23] [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: 07/20/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 11/15/2024] Open
Abstract
Background In the SARSCov2 virus epidemic, pregnant women are more susceptible to infectious diseases due to changes in biochemical parameters and are at higher risk of severe respiratory disease and pneumonia. This study aimed to evaluate the biochemical, inflammatory and coagulation parameters in pregnant women with severe disease conditions (as one of the high-risk groups) as well as prognosis and outcome. Materials and Methods This cross-sectional study was performed on 135 pregnant women with COVID-19 admitted to ICU. Demographic and clinical information and laboratory parameters of the patients were evaluated and recorded at the time of admission and in the next follow-up until discharge or death in addition to the outcome and also the pregnancy outcome. Results The mortality rate of pregnant women with COVID-19 was 9.6%. The mortality rate decreases with increasing Hb (OR (95% CI): 0.68 (0.47-0.99); P value = 0.043) and lymphocytes (OR (95% CI): 0.92 (0.85-0.96); P value = 0.028) and will increase significantly with increasing PT (OR (95% CI): 1.24 (1.01-1.51); P value = 0.037), INR (OR (95% CI): 1.89 (1.26-2.25); P value = 0.004), D-dimer (OR (95% CI): 1.68 (1.10-2.08); P value = 0.027), and LDH (OR (95% CI): 1.20 (1.01-1.61); P value = 0.010). Conclusion According to the results of the present study, inflammatory factors such as leukocytes, neutrophils, NLR, CRP have an increasing and lymphocytes have a decreasing trend, so that lymphocytopenia is more common in non-survivors. In addition, increase of PT, INR, D-dimer and LDH and decrease of Hb were significantly associated with increased chance of mortality. But fibrinogen, ferritin, ALT and AST were not significantly associated with mortality in these women.
Collapse
Affiliation(s)
- Mahtab Ebrahim Babai
- Department of Obstetrics and Gynecology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azita Kabiri
- Department of Obstetrics and Gynecology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Minoo Movahedi
- Department of Obstetrics and Gynecology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ataollah Ghahiri
- Department of Obstetrics and Gynecology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Hajhashemi
- Department of Obstetrics and Gynecology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Dehghan
- Department of Obstetrics and Gynecology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
81
|
Oliván-Blázquez B, Bona-Otal M, Méndez-López F, Lerma-Irureta D, García-Izuel P, Ibáñez-Ruiz J, Montolío A, Ruiz-Herreros M, Godino J, Jimeno-Beltran B, Encabo-Berzosa MDM, Arenaz I, Medel-Martínez A, Casado-Vicente V, Coiras M, Tellería-Orriols C, Schoorlemmer J, Magallón-Botaya R. Characterization model of the post COVID-19 condition based on immunological, biochemical, and cytokine markers. iScience 2024; 27:110839. [PMID: 39318534 PMCID: PMC11420445 DOI: 10.1016/j.isci.2024.110839] [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: 01/27/2024] [Revised: 06/02/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Post-coronavirus disease condition (PCC) continues to affect many people globally, yet there remains a lack of diagnostic biomarkers to distinguish PCC from those recovered from acute COVID-19. This study compared biomarkers between two age- and gender-matched groups: PCC individuals and those recovered within three months of acute COVID-19 in 2020 (n = 85 each). Biomarkers were assessed 12-24 months after initial diagnosis, examining biochemical profiles, blood cell counts, coagulation status, antibody serology, lymphocyte populations, and cytokine levels. PCC individuals exhibited significant alterations in 49 of 167 markers, including K+ levels, αGAD antibodies, antithrombin III, insulin-like growth factor-binding protein 3 (IGFBP3), and interleukin-10 (IL-10). A panel of αGAD, IL-10, potassium levels, and CD16brightCD56- cell presence distinguished PCC individuals from recovered patients with >88% accuracy and <92% precision.
Collapse
Affiliation(s)
- Bárbara Oliván-Blázquez
- Department of Psychology and Sociology, University of Zaragoza, Zaragoza, Spain
- Aragonese Primary Care Research Group (GAIAP), Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Carlos III Health Institute, Madrid, Spain
| | | | - Fátima Méndez-López
- Aragonese Primary Care Research Group (GAIAP), Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Carlos III Health Institute, Madrid, Spain
| | - David Lerma-Irureta
- Aragonese Primary Care Research Group (GAIAP), Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
| | - Paula García-Izuel
- Department of Medicine, Psychiatry and Dermatology, University of Zaragoza, Zaragoza, Spain
| | - Jesús Ibáñez-Ruiz
- Biocomputing Unit, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Alberto Montolío
- Biocomputing Unit, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - María Ruiz-Herreros
- Tarazona Primary Health Care Center, Aragonese Healthcare Service (SALUD), Zaragoza, Spain
| | - Javier Godino
- Cell Separation and Flow Cytometry Core, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Beatriz Jimeno-Beltran
- Cell Separation and Flow Cytometry Core, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | | | - Izaskun Arenaz
- Biobank of the Aragon Health System, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Ana Medel-Martínez
- Placental Pathophysiology & Fetal Programming Research Group, Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
| | - Verónica Casado-Vicente
- Parquesol University Health Center, Castilla y León Health Service (SACYL), University Teaching Unit of Family and Community Medicine, University of Valladolid, Valladolid, Spain
| | - Mayte Coiras
- Immunopathology and Viral Reservoir Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jon Schoorlemmer
- Endogenous Retroviruses (ERVs) in Development and Disease Group, Instituto Aragonés deCiencias de la Salud (IACS), Zaragoza, Spain; and ARAID Foundation, Zaragoza, Spain
| | - Rosa Magallón-Botaya
- Aragonese Primary Care Research Group (GAIAP), Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Carlos III Health Institute, Madrid, Spain
- Department of Medicine, Psychiatry and Dermatology, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
82
|
González-Acedo A, Manzano-Moreno FJ, García-Recio E, Ruiz C, de Luna-Bertos E, Costela-Ruiz VJ. Assessment of Supplementation with Different Biomolecules in the Prevention and Treatment of COVID-19. Nutrients 2024; 16:3070. [PMID: 39339670 PMCID: PMC11434975 DOI: 10.3390/nu16183070] [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: 07/25/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/30/2024] Open
Abstract
Consequences of the disease produced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have led to an urgent search for preventive and therapeutic strategies. Besides drug treatments, proposals have been made for supplementation with biomolecules possessing immunomodulatory and antioxidant properties. The objective of this study was to review published evidence on the clinical usefulness of supplementation with vitamin D, antioxidant vitamins (vitamin A, vitamin E, and vitamin C), melatonin, lactoferrin and natural products found in food (curcumin, luteolin, ginger, allicin, magnesium and zinc) as supplements in SARS-CoV-2 infection. In general, supplementation of conventional treatments with these biomolecules has been found to improve the clinical symptoms and severity of the coronavirus disease (COVID-19), with some indications of a preventive effect. In conclusion, these compounds may assist in preventing and/or improving the symptoms of COVID-19. Nevertheless, only limited evidence is available, and findings have been inconsistent. Further investigations are needed to verify the therapeutic potential of these supplements.
Collapse
Affiliation(s)
- Anabel González-Acedo
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
| | - Francisco Javier Manzano-Moreno
- Biomedical Group (BIO277), Department of Stomatology, School of Dentistry, University of Granada, 18016 Granada, Spain;
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
| | - Enrique García-Recio
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
- Institute of Neuroscience, University of Granada, 18016 Granada, Spain
| | - Elvira de Luna-Bertos
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
| | - Víctor Javier Costela-Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
| |
Collapse
|
83
|
Santana-de Anda K, Torres-Ruiz J, Mejía-Domínguez NR, Alcalá-Carmona B, Maravillas-Montero JL, Páez-Franco JC, Vargas-Castro AS, Lira-Luna J, Camacho-Morán EA, Juarez-Vega G, Meza-Sánchez D, Núñez-Álvarez C, Rull-Gabayet M, Gómez-Martín D. Novel Clinical, Immunological, and Metabolic Features Associated with Persistent Post-Acute COVID-19 Syndrome. Int J Mol Sci 2024; 25:9661. [PMID: 39273608 PMCID: PMC11395921 DOI: 10.3390/ijms25179661] [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: 08/12/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) survivors are frequently observed to present persistent symptoms constituting what has been called "post-acute COVID-19 syndrome" (PACS) or "long COVID-19". Some clinical risk factors have been identified to be associated with PACS development; however, specific mechanisms responsible for PACS pathology remain unknown. This study investigates clinical, immunological, and metabolomic risk factors associated with post-acute COVID-19 syndrome (PACS) in 51 patients, assessed 7-19 months after acute infection. Among the participants, 62.7% were male and 37.2% were female, with an average age of 47.8 years. At the follow-up, 37.2% met the criteria for PACS, revealing significant differences in immunological and metabolomic profiles at the time of acute infection. Patients with PACS were characterized by elevated levels of mature low-density granulocytes (LDGs), interleukin-8 (IL-8), pyruvate, pseudouridine, and cystine. Baseline multivariate analysis showed increased pyruvate and decreased alpha tocopherol levels. At follow-up, there was a decrease in absolute B lymphocytes and an increase in non-classical monocytes and 3-hydroxyisovaleric acid levels. These findings suggest that specific immunological and metabolomic markers during acute infection can help identify patients at higher risk of developing persistent PACS.
Collapse
Affiliation(s)
- Karina Santana-de Anda
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Jiram Torres-Ruiz
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Beatriz Alcalá-Carmona
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - José L Maravillas-Montero
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - José Carlos Páez-Franco
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | | | - Jaquelin Lira-Luna
- Departamento de Cirugía General, Hospital Regional 1ro de Octubre, ISSSTE, Mexico City 07760, Mexico
| | - Emmanuel A Camacho-Morán
- Departamento de Medicina Crítica, Instituto Nacional de Perinatología, Isidro Espinosa de los Reyes, Mexico City 11000, Mexico
| | - Guillermo Juarez-Vega
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - David Meza-Sánchez
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Carlos Núñez-Álvarez
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Marina Rull-Gabayet
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| | - Diana Gómez-Martín
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico
| |
Collapse
|
84
|
Wu G, Wu Y, Gharaibeh NM, Li T, Cao X, Li X. Magnetic resonance (MR) evaluation of deep venous thrombosis of 338 discharged viral pneumonia patients. Quant Imaging Med Surg 2024; 14:6413-6424. [PMID: 39281170 PMCID: PMC11400653 DOI: 10.21037/qims-23-1607] [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: 11/12/2023] [Accepted: 08/05/2024] [Indexed: 09/18/2024]
Abstract
Background Viral pneumonia (VP) often leads to the development of deep vein thrombosis (DVT) in hospitalized patients. The aim of the study was to investigate the incidence of DVT in discharged patients with VP, and whether new and old DVT differ in transverse relaxation time. Methods In this prospective cohort study in Wuhan, China, 338 consecutive discharged VP patients from February 2021 to March 2023 who underwent T2 weighted Sampling Perfection with Application Optimized Contrast Evolution (SPACE) were recruited to detect DVT. T2 mapping and T2* mapping were performed for the patients with DVT detected by magnetic resonance imaging (MRI). The minimum, maximum, mean of T2 time and T2* time of DVT were recorded as T2min, T2max, T2mean, T2*min, T2*max, and T2*mean, respectively. Clinical data and laboratory findings were compared between new and old DVT cases, which were defined based on the examination results before and after discharge. A Mann-Whitney test was used to compare transverse relaxation time parameters between new and old DVT. Results Twelve percent of VP patients (40/338) developed new DVT after discharge. Thirty-three out of 104 DVTs did not resolve after discharge. Compared with patients with new DVT, patients with old DVT were older (67 vs. 59 years, P=0.003); and had a higher proportion of bedridden time >72 hours (72.7% vs. 37.0%, P<0.001). Patients with old DVT had a lower lymphocyte count (0.67×109/L vs. 0.97×109/L, P=0.01), higher C-reactive protein (59 vs. 35 mg/L, P=0.019), and higher levels of D-dimer (6.7 vs. 0.9 µg/mL, P<0.001) than patients with new DVT. Patients with old DVT received more invasive mechanical ventilation (30.3% vs. 7.4%, P<0.001) and had a higher proportion of acute respiratory distress syndrome (75.8% vs. 51.9%, P<0.001), and a higher proportion of cardiac injury (39.4% vs. 14.8%, P=0.033) than patients with new DVT. T2min, T2max, T2mean, and T2*max of new DVT were significantly greater than old DVT (17.6±10.4 vs. 13.2±5.9 ms, 94.9±44.9 vs. 42.3±23.6 ms, 46.8±24.0 vs. 25.0±12.6 ms, 22.5±12.4 vs. 10.7±3.5 ms, P<0.05 for all). There was no significant difference in T2*min or T2*mean between new and old DVT (3.2±0.4 vs. 3.1±0.4 ms, 8.2±4.9 vs. 5.5±1.5 ms, P>0.05 for both). Conclusions T2 weighted SPACE magnetic resonance (MR) is valuable in the follow-up of thrombosis of discharged VP patients. T2 mapping distinguishes between new and old DVT.
Collapse
Affiliation(s)
- Gang Wu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yin Wu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Nadeer M Gharaibeh
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Li
- Department of Radiology, The Central Hospital of Wuhan, Tongji Medical College affiliated to Huazhong University of Science and Technology, Wuhan, China
| | - Xueqin Cao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoming Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
85
|
Mahroum N, Habra M, Alrifaai MA, Shoenfeld Y. Antiphospholipid syndrome in the era of COVID-19 - Two sides of a coin. Autoimmun Rev 2024; 23:103543. [PMID: 38604461 DOI: 10.1016/j.autrev.2024.103543] [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: 01/31/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
In addition to the respiratory symptoms associated with COVID-19, the disease has consistently been linked to many autoimmune diseases such as systemic lupus erythematous and antiphospholipid syndrome (APS). APS in particular was of paramount significance due to its devastating clinical sequela. In fact, the hypercoagulable state seen in patients with acute COVID-19 and the critical role of anticoagulant treatment in affected individuals shed light on the possible relatedness between APS and COVID-19. Moreover, the role of autoimmunity in the assumed association is not less important especially with the accumulated data available regarding the autoimmunity-triggering effect of SARS-CoV-2 infection. This is furtherly strengthened at the time patients with COVID-19 manifested antiphospholipid antibodies of different types following infection. Additionally, the severe form of the APS spectrum, catastrophic APS (CAPS), was shown to have overlapping characteristics with severe COVID-19 such as cytokine storm and multi-organ failure. Interestingly, COVID vaccine-induced autoimmune phenomena described in the medical literature have pointed to an association with APS. Whether the antiphospholipid antibodies were present or de novo, COVID vaccine-induced vascular thrombosis in certain individuals necessitates further investigations regarding the possible mechanisms involved. In our current paper, we aimed to focus on the associations mentioned, their implications, importance, and consequences.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Mona Habra
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | | | - Yehuda Shoenfeld
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel; Reichman University, Herzliya, Israel
| |
Collapse
|
86
|
Eustes AS, Ahmed A, Swamy J, Patil G, Jensen M, Wilson KM, Kudchadkar S, Wahab A, Perepu U, Miller FJ, Lentz SR, Dayal S. Extracellular histones: a unifying mechanism driving platelet-dependent extracellular vesicle release and thrombus formation in COVID-19. J Thromb Haemost 2024; 22:2514-2530. [PMID: 38815756 PMCID: PMC11343660 DOI: 10.1016/j.jtha.2024.05.019] [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: 11/21/2023] [Revised: 05/12/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND COVID-19 can cause profound inflammation and coagulopathy, and while many mechanisms have been proposed, there is no known common pathway leading to a prothrombotic state. OBJECTIVES From the beginning of the COVID-19 pandemic, elevated levels of extracellular histones have been found in plasma of patients infected with SARS-CoV-2. We hypothesized that platelet activation triggered by extracellular histones might represent a unifying mechanism leading to increased thrombin generation and thrombosis. METHODS We utilized blood samples collected from an early clinical trial of hospitalized COVID-19 patients (NCT04360824) and recruited healthy subjects as controls. Using plasma samples, we measured the procoagulant and prothrombotic potential of circulating extracellular histones and extracellular vesicles (EVs). Platelet prothrombotic activity was assessed via thrombin generation potential and platelet thrombus growth. Circulating EVs were assessed for thrombin generation potential in vitro in plasma and enhancement of thrombotic susceptibility in vivo in mice. RESULTS Compared with controls, COVID-19 patients had elevated plasma levels of citrullinated histone H3, cell-free DNA, nucleosomes, and EVs. Plasma from COVID-19 patients promoted platelet activation, platelet-dependent thrombin generation, thrombus growth under venous shear stress, and release of platelet-derived EVs. These prothrombotic effects of COVID-19 plasma were inhibited by an RNA aptamer that neutralizes both free and DNA-bound histones. EVs isolated from COVID-19 plasma enhanced thrombin generation in vitro and potentiated venous thrombosis in mice in vivo. CONCLUSION We conclude that extracellular histones and procoagulant EVs drive the prothrombotic state in COVID-19 and that histone-targeted therapy may prove beneficial.
Collapse
Affiliation(s)
- Alicia S Eustes
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Azaj Ahmed
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jagadish Swamy
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Gokul Patil
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Melissa Jensen
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Katina M Wilson
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Shibani Kudchadkar
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Abdul Wahab
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Usha Perepu
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Francis J Miller
- Department of Internal Medicine, Vanderbilt University Medical Center and VA Medical Center, Nashville, Tennessee, USA
| | - Steven R Lentz
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA; Iowa City VA Healthcare System, Iowa City, Iowa, USA.
| |
Collapse
|
87
|
Steinbeis F, Kedor C, Meyer HJ, Thibeault C, Mittermaier M, Knape P, Ahrens K, Rotter G, Temmesfeld-Wollbrück B, Sander LE, Kurth F, Witzenrath M, Scheibenbogen C, Zoller T. A new phenotype of patients with post-COVID-19 condition is characterised by a pattern of complex ventilatory dysfunction, neuromuscular disturbance and fatigue symptoms. ERJ Open Res 2024; 10:01027-2023. [PMID: 39377086 PMCID: PMC11456967 DOI: 10.1183/23120541.01027-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/10/2024] [Indexed: 10/09/2024] Open
Abstract
Background Patients with post-COVID-19 condition frequently suffer from chronic dyspnoea. The causes and mechanism for dyspnoea in these patients without evidence of structural lung disease are unclear. Methods Patients treated for COVID-19 at Charité University Hospital in Berlin received pulmonary function testing including respiratory muscle strength tests and completed health-related quality-of-life questionnaires during follow-up. Patients with post-COVID-19 condition during outpatient follow-up with fatigue and exertional intolerance (PCF) were compared to patients with post-COVID-19 condition with evidence of chronic pulmonary sequelae (post-COVID-19 restriction (PCR)) as well as to patients without post-COVID-19 condition (NCF). Results A total of 170 patients presented for follow-up. 36 participants met criteria for PCF, 28 for PCR and 24 for NCF. PCF patients reported dyspnoea in 63.8%. % predicted value of respiratory muscle strength (median (IQR)) was reduced in PCF (55.8 (41.5-75.9)) compared to NCF and PCR (70.6 (66.3-88.9) and 76.8 (63.6-102.2), respectively; p=0.011). A pattern of reduced forced vital capacity (FVC), but normal total lung capacity (TLC), termed complex ventilatory dysfunction defined as TLC - FVC >10% predicted was observed and occurred more frequently in PCF (88.9%) compared to NCF and PCR (29.1% and 25.0%, respectively; p<0.001). Conclusion Dyspnoea in PCF is characterised by reduced respiratory muscle strength and complex ventilatory dysfunction indicating neuromuscular disturbance as a distinct phenotype among patients with post-COVID-19 condition. These observations could be a starting point for developing personalised rehabilitation concepts.
Collapse
Affiliation(s)
- Fridolin Steinbeis
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Claudia Kedor
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Medical Immunology, Berlin, Germany
| | - Hans-Jakob Meyer
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Charlotte Thibeault
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Mirja Mittermaier
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Knape
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Katharina Ahrens
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Gabriele Rotter
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Social Medicine, Epidemiology and Health Economics, Berlin, Germany
| | - Bettina Temmesfeld-Wollbrück
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Leif Erik Sander
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Florian Kurth
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Medicine I, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Witzenrath
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Carmen Scheibenbogen
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Medical Immunology, Berlin, Germany
| | - Thomas Zoller
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| |
Collapse
|
88
|
Palazzuoli A, Mingiano C, Manetti N, Leolini C, Fossi A. Pacing lead thrombus in patient with recent COVID-19 infection and subsequent vaccination: a case report. Eur Heart J Case Rep 2024; 8:ytae447. [PMID: 39258023 PMCID: PMC11384885 DOI: 10.1093/ehjcr/ytae447] [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: 03/01/2024] [Revised: 04/19/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024]
Abstract
Background The association between acute coronavirus disease-19 (COVID-19) infection and a hypercoagulable state has been exhaustively described throughout the pandemic. The presence of external devices, such as intracardiac leads, could predispose to higher thrombotic risk in this setting. We present a clinical case of intracardiac thrombosis on right ventricle device that occurred after COVID-19 infection and subsequent vaccination. Case summary A 56-year-old man, suffering from usual interstitial pneumonia-pattern fibrosis, was admitted to our hospital because of worsening of his clinical status. About 10 days earlier, he had got vaccinated for COVID-19. Three months earlier, the patient had been reported to have severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection. A chest computed tomography scan showed thrombus partially occluding the left pulmonary artery. A transthoracic echocardiography and later a transoesophageal echocardiogram showed a mass adhered to the lead in the right ventricle, compatible with thrombosis, confirmed on a cardiac computed tomography scan. Blood tests showed no major changes except for a slight increase in D-dimer and fibrinogen. Therefore, the subject was treated with anticoagulants. Discussion COVID-19 infection results in a hypercoagulable state with risk of developing thrombus diffusely, including intracardiac thrombosis. The presence of external devices, such as the intracardiac leads, may increase thrombotic risk since the presence of an external device in the bloodstream could trigger coagulation cascade. This case report highlights the need for special care in this patient setting, using specific imaging techniques for early and rapid diagnosis to optimize therapy.
Collapse
Affiliation(s)
- Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department, Le Scotte Hospital, University of Siena, 53100 Siena, Italy
| | - Christian Mingiano
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Niccolò Manetti
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department, Le Scotte Hospital, University of Siena, 53100 Siena, Italy
| | - Chiara Leolini
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department, Le Scotte Hospital, University of Siena, 53100 Siena, Italy
| | - Antonella Fossi
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Siena, Italy
| |
Collapse
|
89
|
Deora N, Venkatraman K. Potential use of plant-based therapeutics for the management of SARS-COV2 infection in diabetes mellitus – a review. J Herb Med 2024; 47:100923. [DOI: 10.1016/j.hermed.2024.100923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
90
|
Dumea E, Lazar M, Chitu-Tisu CE, Barbu EC, Ion DA. COVID-19 associated pulmonary embolism: clinical, biochemical and CT imaging findings. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:307-322. [PMID: 38641909 DOI: 10.2478/rjim-2024-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Indexed: 04/21/2024]
Abstract
INTRODUCTION The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection represented a disruptive pathology that emerged in late 2019 with profound implications ranging from individual health to health systems and world economy. Our study aimed to evaluate clinical, biochemical and computerized tomography (CT) parameters values in determining the severity of pulmonary embolism (PE) associated with COVID-19. METHODS We performed an observational cohort study evaluating demographic, clinical, biochemical, coagulation markers, as well as CT imaging parameters. RESULTS In our study on 186 patients with COVID-19, we found that 31 patients (16,66%) had pulmonary embolism. Significant correlations for the patients with PE were detected in C-reactive protein, lactate dehydrogenase, serum ferritin, IL-6, serum myoglobin, NT-proBNP, D-dimers, serum proteins, transaminases as well as white cell blood counts. Patients with pulmonary embolism had a more severe lung involvement, with thrombi distribution mainly involving the lower lobes. CONCLUSION Early identification of PE is an important step for timely and efficient treatment in the intensive care management of COVID-19 patients. Our study showed that high plasmatic values of lactate dehydrogenase, ferritin, IL-6, white blood cells and D-dimers and low proteins serum levels are strongly linked with COVID-19-associated pulmonary embolism.
Collapse
Affiliation(s)
- Eduard Dumea
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Mihai Lazar
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
- 2"Prof. Dr. Matei Bals" National Institute for Infectious Diseases, No. 1, Calistrat Grozovici Street, Sector 2, 021105 Bucharest, Romania
| | - Cristina Emilia Chitu-Tisu
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Ecaterina Constanta Barbu
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| | - Daniela Adriana Ion
- 1Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, No. 37, Dionisie Lupu Street, Sector 2, 020021 Bucharest, Romania
| |
Collapse
|
91
|
Wu K, Van Name J, Xi L. Cardiovascular abnormalities of long-COVID syndrome: Pathogenic basis and potential strategy for treatment and rehabilitation. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:221-231. [PMID: 39234483 PMCID: PMC11369840 DOI: 10.1016/j.smhs.2024.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 09/06/2024] Open
Abstract
Cardiac injury and sustained cardiovascular abnormalities in long-COVID syndrome, i.e. post-acute sequelae of coronavirus disease 2019 (COVID-19) have emerged as a debilitating health burden that has posed challenges for management of pre-existing cardiovascular conditions and other associated chronic comorbidities in the most vulnerable group of patients recovered from acute COVID-19. A clear and evidence-based guideline for treating cardiac issues of long-COVID syndrome is still lacking. In this review, we have summarized the common cardiac symptoms reported in the months after acute COVID-19 illness and further evaluated the possible pathogenic factors underlying the pathophysiology process of long-COVID. The mechanistic understanding of how Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) damages the heart and vasculatures is critical in developing targeted therapy and preventive measures for limiting the viral attacks. Despite the currently available therapeutic interventions, a considerable portion of patients recovered from severe COVID-19 have reported a reduced functional reserve due to deconditioning. Therefore, a rigorous and comprehensive cardiac rehabilitation program with individualized exercise protocols would be instrumental for the patients with long-COVID to regain the physical fitness levels comparable to their pre-illness baseline.
Collapse
Affiliation(s)
- Kainuo Wu
- Virginia Commonwealth University School of Medicine (M.D. Class 2024), Richmond, VA, 23298, USA
| | - Jonathan Van Name
- Virginia Commonwealth University School of Medicine (M.D. Class 2024), Richmond, VA, 23298, USA
| | - Lei Xi
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA
| |
Collapse
|
92
|
Jerah AA, Farasani A, Abu-Tawil H, Kuriri H, Taha MME, Abdelwahab SI, Albasheer O. Unveiling coagulation dysfunction in patients with COVID-19: a retrospective analysis. J Med Life 2024; 17:886-891. [PMID: 39628975 PMCID: PMC11611055 DOI: 10.25122/jml-2024-0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/01/2024] [Indexed: 12/06/2024] Open
Abstract
Coagulation dysfunction has emerged as a significant aspect of COVID-19 pathophysiology, with abnormal coagulation parameters observed in severe cases. This study aimed to investigate the predictive value of coagulation parameters, including prothrombin time (PT), activated partial thromboplastin time (PTT), and international normalized ratio (INR) for mortality in patients with COVID-19. A retrospective analysis was conducted on a cohort of patients diagnosed with COVID-19. Coagulation parameters, including PT, PTT, and INR, were measured upon admission. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive performance of these parameters. Sensitivity and specificity were calculated, and the area under the curve (AUC) values were determined. The analysis included 156 patients diagnosed with COVID-19. The t-test revealed a significant difference (P < 0.05) in PTT, PT, and INR. PTT demonstrated the highest predictive performance, with an AUC value of 0.68, indicating superior discrimination compared with PT and INR. PTT exhibited a sensitivity of 83% and a specificity of 46% for identifying deceased patients. These findings suggest that PTT may serve as a valuable prognostic marker of mortality risk in patients with COVID-19. Coagulation indicators, particularly PTT, predicted COVID-19 mortality. Monitoring coagulation markers may help stratify the risk and guide treatment. Further research and validation studies are needed to corroborate these findings and to establish the clinical importance of coagulation markers in COVID-19 therapy. COVID-19 coagulation dysfunction mechanisms must be understood in order to design targeted therapies to reduce thrombotic consequences.
Collapse
Affiliation(s)
- Ahmed Ali Jerah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Abdullah Farasani
- Biomedical Research Unit, Medical Research Center, Jazan University, Jazan, Saudi Arabia
| | - Hisham Abu-Tawil
- Department of Clinical Laboratory and Blood Bank, King Faisal Medical City for Southern Regions, Abha, Saudi Arabia
| | - Hadi Kuriri
- Department of Clinical Laboratory and Blood Bank, Samtah General Hospital, Samtah, Saudi Arabia
| | | | | | - Osama Albasheer
- Family and Community Medicine Department, College of Medicine, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
93
|
Aragona M, Mhalhel K, Pansera L, Montalbano G, Guerrera MC, Levanti M, Laurà R, Abbate F, Vega JA, Germanà A. Localization of Piezo 1 and Piezo 2 in Lateral Line System and Inner Ear of Zebrafish ( Danio rerio). Int J Mol Sci 2024; 25:9204. [PMID: 39273152 PMCID: PMC11395407 DOI: 10.3390/ijms25179204] [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: 07/31/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Piezo proteins have been identified as mechanosensitive ion channels involved in mechanotransduction. Several ion channel dysfunctions may be associated with diseases (including deafness and pain); thus, studying them is critical to understand their role in mechanosensitive disorders and to establish new therapeutic strategies. The current study investigated for the first time the expression patterns of Piezo proteins in zebrafish octavolateralis mechanosensory organs. Piezo 1 and 2 were immunoreactive in the sensory epithelia of the lateral line system and the inner ear. Piezo 1 (28.7 ± 1.55 cells) and Piezo 2 (28.8 ± 3.31 cells) immunopositive neuromast cells were identified based on their ultrastructural features, and their overlapping immunoreactivity to the s100p specific marker (28.6 ± 1.62 cells), as sensory cells. These findings are in favor of Piezo proteins' potential role in sensory cell activation, while their expression on mantle cells reflects their implication in the maintenance and regeneration of the neuromast during cell turnover. In the inner ear, Piezo proteins' colocalization with BDNF introduces their potential implication in neuronal plasticity and regenerative events, typical of zebrafish mechanosensory epithelia. Assessing these proteins in zebrafish could open up new scenarios for the roles of these important ionic membrane channels, for example in treating impairments of sensory systems.
Collapse
Affiliation(s)
- Marialuisa Aragona
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Kamel Mhalhel
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Lidia Pansera
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Maria Cristina Guerrera
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Maria Levanti
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Rosaria Laurà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Francesco Abbate
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - José A Vega
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, 33006 Oviedo, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 7500912, Chile
| | - Antonino Germanà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
94
|
de Souza Goncalves B, Sangani D, Nayyar A, Puri R, Irtiza M, Nayyar A, Khalyfa A, Sodhi K, Pillai SS. COVID-19-Associated Sepsis: Potential Role of Phytochemicals as Functional Foods and Nutraceuticals. Int J Mol Sci 2024; 25:8481. [PMID: 39126050 PMCID: PMC11312872 DOI: 10.3390/ijms25158481] [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: 07/08/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
The acute manifestations of coronavirus disease 2019 (COVID-19) exhibit the hallmarks of sepsis-associated complications that reflect multiple organ failure. The inflammatory cytokine storm accompanied by an imbalance in the pro-inflammatory and anti-inflammatory host response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to severe and critical septic shock. The sepsis signature in severely afflicted COVID-19 patients includes cellular reprogramming and organ dysfunction that leads to high mortality rates, emphasizing the importance of improved clinical care and advanced therapeutic interventions for sepsis associated with COVID-19. Phytochemicals of functional foods and nutraceutical importance have an incredible impact on the healthcare system, which includes the prevention and/or treatment of chronic diseases. Hence, in the present review, we aim to explore the pathogenesis of sepsis associated with COVID-19 that disrupts the physiological homeostasis of the body, resulting in severe organ damage. Furthermore, we have summarized the diverse pharmacological properties of some potent phytochemicals, which can be used as functional foods as well as nutraceuticals against sepsis-associated complications of SARS-CoV-2 infection. The phytochemicals explored in this article include quercetin, curcumin, luteolin, apigenin, resveratrol, and naringenin, which are the major phytoconstituents of our daily food intake. We have compiled the findings from various studies, including clinical trials in humans, to explore more into the therapeutic potential of each phytochemical against sepsis and COVID-19, which highlights their possible importance in sepsis-associated COVID-19 pathogenesis. We conclude that our review will open a new research avenue for exploring phytochemical-derived therapeutic agents for preventing or treating the life-threatening complications of sepsis associated with COVID-19.
Collapse
Affiliation(s)
- Bruno de Souza Goncalves
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Darshan Sangani
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Aleen Nayyar
- Department of Medicine, Sharif Medical and Dental College, Lahore 55150, Pakistan;
| | - Raghav Puri
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Mahir Irtiza
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Asma Nayyar
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Abdelnaby Khalyfa
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Komal Sodhi
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| | - Sneha S. Pillai
- Department of Surgery, Internal Medicine and Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA; (B.d.S.G.); (D.S.); (R.P.); (M.I.); (A.N.); (A.K.); (K.S.)
| |
Collapse
|
95
|
Yada N, Zhang Q, Bignotti A, Ye Z, Zheng XL. ADAMTS13 or Caplacizumab Reduces the Accumulation of Neutrophil Extracellular Traps and Thrombus in Whole Blood of COVID-19 Patients under Flow. Thromb Haemost 2024; 124:725-738. [PMID: 38272066 PMCID: PMC11260255 DOI: 10.1055/a-2253-9359] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
BACKGROUND Neutrophil NETosis and neutrophil extracellular traps (NETs) play a critical role in pathogenesis of coronavirus disease 2019 (COVID-19)-associated thrombosis. However, the extents and reserve of NETosis, and potential of thrombus formation under shear in whole blood of patients with COVID-19 are not fully elucidated. Neither has the role of recombinant ADAMTS13 or caplacizumab on the accumulation of NETs and thrombus in COVID-19 patients' whole blood under shear been investigated. METHODS Flow cytometry and microfluidic assay, as well as immunoassays, were employed for the study. RESULTS We demonstrated that the percentage of H3Cit + MPO+ neutrophils, indicative of NETosis, was dramatically increased in patients with severe but not critical COVID-19 compared with that in asymptomatic or mild disease controls. Upon stimulation with poly [I:C], a double strain DNA mimicking viral infection, or bacterial shigatoxin-2, the percentage of H3Cit + MPO+ neutrophils was not significantly increased in the whole blood of severe and critical COVID-19 patients compared with that of asymptomatic controls, suggesting the reduction in NETosis reserve in these patients. Microfluidic assay demonstrated that the accumulation of NETs and thrombus was significantly enhanced in the whole blood of severe/critical COVID-19 patients compared with that of asymptomatic controls. Like DNase I, recombinant ADAMTS13 or caplacizumab dramatically reduced the NETs accumulation and thrombus formation under arterial shear. CONCLUSION Significantly increased neutrophil NETosis, reduced NETosis reserve, and enhanced thrombus formation under arterial shear may play a crucial role in the pathogenesis of COVID-19-associated coagulopathy. Recombinant ADAMTS13 or caplacizumab may be explored for the treatment of COVID-19-associated thrombosis.
Collapse
Affiliation(s)
- Noritaka Yada
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kanas City, Kansas, United States
| | - Quan Zhang
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kanas City, Kansas, United States
| | - Antonia Bignotti
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kanas City, Kansas, United States
| | - Zhan Ye
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kanas City, Kansas, United States
| | - X. Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kanas City, Kansas, United States
- Institute of Reproductive Medicine and Developmental Sciences, The University of Kansas Medical Center, Kanas City, Kansas, United States
| |
Collapse
|
96
|
Lemon NM, Taylor LK, Rech MA, Nguyen Q, Matthews GJ, Lew G, Lovett S. Utility of D-dimer in predicting pulmonary embolism in patients with COVID-19 presenting to the emergency department. J Am Coll Emerg Physicians Open 2024; 5:e13237. [PMID: 39027350 PMCID: PMC11255020 DOI: 10.1002/emp2.13237] [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: 09/22/2023] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Objectives While our understanding of coronavirus disease 2019 (COVID-19) has evolved, uncertainty remains regarding utility of previously established pulmonary embolism (PE) screening guidelines in patients with COVID-19. Many studies have investigated the efficacy of D-dimer (DD) screenings for patients with COVID-19 admitted to inpatient services, but few have evaluated patients in the emergency department (ED). The purpose of this study was to investigate utility of DD threshold for PE screening in patients with COVID-19 presenting to the ED. Methods This was a retrospective, multicenter cohort including patients presenting to three EDs between March 1, 2020 and February 1, 2021 who tested positive for COVID-19 during ED visit or in 60 days prior to presentation and had DD ordered in ED. Patients were grouped by those who underwent computed tomography pulmonary angiogram (CTPA) to evaluate for PE and those who did not, and descriptive statistics were performed. Those who underwent CTPA were further divided into PE-positive and PE-negative groups. The discriminative ability of DD in predicting PE in patients with COVID-19 was analyzed using the receiver operating characteristic (ROC) curve. Results A total of 570 patients with COVID-19 were included in the study, of which 107 underwent CTPA to evaluate for PE. History of diabetes, elevated glucose, elevated lactate dehydrogenase, elevated white blood cell count, elevated platelets, elevated respiratory rate, and lower temperature were associated with increased risk for PE. Compared to those without PE, patients with PE were significantly more likely to be hospitalized (100% vs. 82%, p = 0.020) and admitted to the ICU (64% vs. 24%, p = 0.002). Those with PE had a significantly higher median DD value (21,177 ng/mL) compared to PE-negative group (952 ng/mL, p < 0.001). The ROC curve for DD in predicting PE had an area under the curve of 0.91 (95% confidence interval [0.84, 0.98]). In our study population, the optimal DD threshold for predicting PE was 1815 ng/mL (sensitivity 93% and specificity 80%). A conservative threshold of 1089 ng/mL could be used with sensitivity 100% and specificity 58%. Conclusion DD is often elevated in patients with COVID-19, regardless of PE. While the classically used DD cutoff is 500 ng/mL, our study demonstrated a threshold of 1089 ng/mL safely predicted PE in patients with COVID-19 .
Collapse
Affiliation(s)
- Natalie M. Lemon
- Department of Emergency MedicineUniversity of Chicago Medical CenterChicagoIllinoisUSA
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Luke K. Taylor
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Megan A. Rech
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
- Loyola University Medical CenterChicagoIllinoisUSA
- Center of Innovation for Complex Chronic HealthcareEdward Hines VA HospitalHinesIllinoisUSA
| | - Quang Nguyen
- Department of Statistics and Data ScienceCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Center for Data Science and ConsultingLoyola University ChicagoChicagoIllinoisUSA
| | - Gregory J. Matthews
- Center for Data Science and ConsultingLoyola University ChicagoChicagoIllinoisUSA
- Department of Mathematics and StatisticsLoyola University ChicagoChicagoIllinoisUSA
| | - George Lew
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
- Loyola University Medical CenterChicagoIllinoisUSA
| | - Shannon Lovett
- Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
- Loyola University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
97
|
Zuin M, Ferrari R, Guardigli G, Malagù M, Vitali F, Zucchetti O, D'Aniello E, Di Ienno L, Gibiino F, Cimaglia P, Grosseto D, Corzani A, Galvani M, Ortolani P, Rubboli A, Tortorici G, Casella G, Sassone B, Navazio A, Rossi L, Aschieri D, Mezzanotte R, Manfrini M, Bertini M. A COVID-19 specific multiparametric and ECG-based score for the prediction of in-hospital mortality: ELCOVID score. Intern Emerg Med 2024; 19:1279-1290. [PMID: 38652232 DOI: 10.1007/s11739-024-03599-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
We aimed to develop and validate a COVID-19 specific scoring system, also including some ECG features, to predict all-cause in-hospital mortality at admission. Patients were retrieved from the ELCOVID study (ClinicalTrials.gov identifier: NCT04367129), a prospective, multicenter Italian study enrolling COVID-19 patients between May to September 2020. For the model validation, we randomly selected two-thirds of participants to create a derivation dataset and we used the remaining one-third of participants as the validation set. Over the study period, 1014 hospitalized COVID-19 patients (mean age 74 years, 61% males) met the inclusion criteria and were included in this analysis. During a median follow-up of 12 (IQR 7-22) days, 359 (35%) patients died. Age (HR 2.25 [95%CI 1.72-2.94], p < 0.001), delirium (HR 2.03 [2.14-3.61], p = 0.012), platelets (HR 0.91 [0.83-0.98], p = 0.018), D-dimer level (HR 1.18 [1.01-1.31], p = 0.002), signs of right ventricular strain (RVS) (HR 1.47 [1.02-2.13], p = 0.039) and ECG signs of previous myocardial necrosis (HR 2.28 [1.23-4.21], p = 0.009) were independently associated to in-hospital all-cause mortality. The derived risk-scoring system, namely EL COVID score, showed a moderate discriminatory capacity and good calibration. A cut-off score of ≥ 4 had a sensitivity of 78.4% and 65.2% specificity in predicting all-cause in-hospital mortality. ELCOVID score represents a valid, reliable, sensitive, and inexpensive scoring system that can be used for the prognostication of COVID-19 patients at admission and may allow the earlier identification of patients having a higher mortality risk who may be benefit from more aggressive treatments and closer monitoring.
Collapse
Affiliation(s)
- Marco Zuin
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy
| | - Roberto Ferrari
- Unit of Cardiology, Maria Cecilia Hospital, Cotignola, Ravenna, Italy
| | - Gabriele Guardigli
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy
| | - Michele Malagù
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy
| | - Francesco Vitali
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy
| | - Ottavio Zucchetti
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy
| | - Emanuele D'Aniello
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy
| | - Luca Di Ienno
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy
| | - Federico Gibiino
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy
| | - Paolo Cimaglia
- Unit of Cardiology, Maria Cecilia Hospital, Cotignola, Ravenna, Italy
| | | | | | | | - Paolo Ortolani
- Unit of Cardiology, Ospedale S. Maria della Scaletta, Imola, Italy
| | - Andrea Rubboli
- Unit of Cardiology, Ospedale S. Maria delle Croci, Ravenna, Italy
| | | | - Gianni Casella
- Unit of Cardiology, Ospedale Maggiore C.A. Pizzardi, Bologna, Italy
| | - Biagio Sassone
- Unit of Cardiology, Ospedale del Delta, Lagosanto, Ferrara, Italy
| | | | - Luca Rossi
- Unit of Cardiology, Ospedale Guglielmo da Saliceto, Piacenza, Italy
| | - Daniela Aschieri
- Unit of Cardiology, Ospedale Civile di Castel San Giovanni, Piacenza, Italy
| | | | - Marco Manfrini
- Unit of Cardiology, Maria Cecilia Hospital, Cotignola, Ravenna, Italy
| | - Matteo Bertini
- Unit of Cardiology, Department of Translational Medicine, Centro Cardiologico, Universita' degli studi di Ferrara, Via Aldo Moro, 8, 44124, Cona, Ferrara, Italy.
| |
Collapse
|
98
|
Sanders AP, Vosburg RW. Early postoperative COVID infection is associated with significantly increased risk of venous thromboembolism after metabolic and bariatric surgery. Surg Obes Relat Dis 2024; 20:730-736. [PMID: 38556419 DOI: 10.1016/j.soard.2024.01.021] [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: 10/01/2023] [Revised: 12/11/2023] [Accepted: 01/28/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Patients who undergo metabolic and bariatric surgery (MBS) are at risk for thromboembolism. Patients are susceptible to coronavirus throughout the perioperative period, which also has a well-known association with thrombotic complications. OBJECTIVES To identify and define the association between venous thromboembolism (VTE) and postoperative coronavirus diagnosis in bariatric surgery patients. SETTING United States. METHODS We conducted a retrospective cohort study using the MBS Accreditation and Quality Improvement Program (MBSAQIP) 2021 database to analyze the incidence of VTE within 30 days of surgery. VTE was a composite variable defined as either postoperative pulmonary embolism or postoperative venous thrombus requiring treatment. Cohorts were stratified by whether the patient was diagnosed with postoperative coronavirus. We created a multivariable logistic regression model to determine the adjusted odds of postoperative VTE based on various factors. Additionally, we conducted subset analyses of sleeve gastrectomy and Roux-en-Y bypass cases, the 2 most frequent bariatric operations in the United States. RESULTS Patients diagnosed with postoperative coronavirus were significantly more likely to develop postoperative VTE (1.1% versus .3%, P < .001). In our logistic regression model, the adjusted odds of postoperative VTE for patients with postoperative coronavirus was 3.55 (95% CI: 2.15-5.87, P < .001). For patients who underwent Roux-en-Y bypass, the adjusted odds was even greater at 5.69 (95% CI: 2.76-11.70, P < .001). CONCLUSIONS Early postoperative coronavirus infection after MBS is associated with higher odds of postoperative VTE. This persisted on subset analyses of the 2 most common procedures and appeared particularly important for Roux-en-Y bypass. COVID infection after MBS may warrant prolonged VTE prophylaxis.
Collapse
Affiliation(s)
- Andrew P Sanders
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - R Wesley Vosburg
- Department of Surgery, Mount Auburn Hospital, Harvard Medical School, Cambridge, Massachusetts.
| |
Collapse
|
99
|
Mariappan V, Adla D, Jangili S, Ranganadin P, Green SR, Mohammed S, Mutheneni SR, Pillai AB. Understanding COVID-19 outcome: Exploring the prognostic value of soluble biomarkers indicative of endothelial impairment. Cytokine 2024; 180:156673. [PMID: 38857562 DOI: 10.1016/j.cyto.2024.156673] [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: 02/03/2024] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
Host proteins released by the activated endothelial cells during SARS-CoV-2 infection are implicated to be involved in coagulation and endothelial dysfunction. However, the underlying mechanism that governs the vascular dysfunction and disease severity in COVID-19 remains obscure. The study evaluated the serum levels of Bradykinin, Kallikrein, SERPIN A, and IL-18 in COVID-19 (N-42 with 20 moderate and 22 severe) patients compared to healthy controls (HC: N-10) using ELISA at the day of admission (DOA) and day 7 post-admission. The efficacy of the protein levels in predicting disease severity was further determined using machine learning models. The levels of bradykinins and SERPIN A were higher (P ≤ 0.001) in both severe and moderate cases on day 7 post-admission compared to DOA. All the soluble proteins studied were found to elevated (P ≤ 0.01) in severe compared to moderate in day 7 and were positively correlated (P ≤ 0.001) with D-dimer, a marker for coagulation. ROC analysis identified that SERPIN A, IL-18, and bradykinin could predict the clinical condition of COVID-19 with AUC values of 1, 0.979, and 1, respectively. Among the models trained using univariate model analysis, SERPIN A emerged as a strong prognostic biomarker for COVID-19 disease severity. The serum levels of SERPIN A in conjunction with the coagulation marker D-dimer, serve as a predictive indicator for COVID-19 clinical outcomes. However, studies are required to ascertain the role of these markers in disease virulence.
Collapse
Affiliation(s)
- Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Deepthi Adla
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Shraddha Jangili
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Pajanivel Ranganadin
- Department of Pulmonary Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Siva Ranaganthan Green
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Salma Mohammed
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Srinivasa Rao Mutheneni
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Tarnaka, Hyderabad 500 007, Telangana, India.
| | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| |
Collapse
|
100
|
Akcilar R, Kocak FE, Kar F, Isiklar OO, Atlanoglu S, Genc O, Yaman F. Evaluation of the relationship between ACE2 G8790A and AT2R A1675G gene polymorphisms in COVID-19 patients with and without lung involvement. ASIAN BIOMED 2024; 18:157-170. [PMID: 39309472 PMCID: PMC11414776 DOI: 10.2478/abm-2024-0022] [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] [Indexed: 09/25/2024]
Abstract
Background The SARS-CoV-2 virus produces severe acute respiratory syndrome. The severity of coronavirus disease 2019 (COVID-19) infection is determined by a number of factors, including inherited ones. Objectives Our goal is to investigate the link between ACE2 G8790A (rs2285666) and AT2R A1675G (rs14035430) gene polymorphisms in COVID-19 patients with and without lung involvement. Methods A total of 160 COVID-19 patients were divided into 2 groups based on their clinical symptoms: those without lung involvement (control group) and those with lung involvement (infected group). The ACE2 G8790A and AT2R A1675G gene polymorphisms were analyzed using the PCR-RFLP methods. Results The GG genotype, G allele of ACE2 G8790A, and GG genotype of AT2R A1675G were significantly higher in the control group and had a protective effect against COVID-19 as well as decreased the development of lung involvement (OR = 0.29, 95% CI = 0.10-0.84; OR = 0.40, 95% CI = 0.22-0.72; and OR = 0.33, 95% CI = 0.14-0.78, respectively). Moreover, we found that the AA genotype, A allele of ACE2 G8790A, and AG genotype of AT2R A1675G increased the risk of COVID-19 in the infected group (OR = 3.50, 95% CI = 1.18-10.3; OR = 2.49, 95% CI = 1.39-4.48; and OR = 3.08, 95% CI = 1.28-7.38, respectively). Conclusions These results revealed that a greater frequency of COVID-19 lung involvement in the Turkish population was connected with the AA genotype, the A allele of ACE2 G8790A, and the AG genotype of AT2R A1675G.
Collapse
Affiliation(s)
- Raziye Akcilar
- Department of Physiology, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Fatma Emel Kocak
- Department of Medical Biochemistry, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Fatih Kar
- Department of Basic Sciences, Kütahya Health Sciences University, Faculty of Natural and Engineering Sciences, Kutahya43100, Turkey
| | - Ozben Ozden Isiklar
- Department of Medical Biochemistry, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Sahinde Atlanoglu
- Department of Radiology, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Ozlem Genc
- Department of Medical Microbiology, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| | - Fatima Yaman
- Department of Physical Medicine and Rehabilitation, Kütahya Health Sciences University, Faculty of Medicine, Kutahya43100, Turkey
| |
Collapse
|