101
|
Thierry AR, Salmon D. Inflammation-, immunothrombosis,- and autoimmune-feedback loops may lead to persistent neutrophil self-stimulation in long COVID. J Med Virol 2024; 96:e29887. [PMID: 39189651 DOI: 10.1002/jmv.29887] [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: 03/28/2024] [Revised: 07/10/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Understanding the pathophysiology of long COVID is one of the most intriguing challenges confronting contemporary medicine. Despite observations recently made in the relevant molecular, cellular, and physiological domains, it is still difficult to say whether the post-acute sequelae of COVID-19 directly correspond to the consequences of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This work hypothesizes that neutrophils and neutrophil extracellular traps (NETs) production are at the interconnection of three positive feedback loops which are initiated in the acute phase of SARS-CoV-2 infection, and which involve inflammation, immunothrombosis, and autoimmunity. This phenomenon could be favored by the fact that SARS-CoV-2 may directly bind and penetrate neutrophils. The ensuing strong neutrophil stimulation leads to a progressive amplification of an exacerbated and uncontrolled NETs production, potentially persisting for months beyond the acute phase of infection. This continuous self-stimulation of neutrophils leads, in turn, to systemic inflammation, micro-thromboses, and the production of autoantibodies, whose significant consequences include the persistence of endothelial and multiorgan damage, and vascular complications.
Collapse
Affiliation(s)
- Alain R Thierry
- IRCM, Institute of Research on Cancerology of Montpellier, INSERM U1194, University of Montpellier, Montpellier, France
- Montpellier Cancer Institute (ICM), Montpellier, France
| | | |
Collapse
|
102
|
Hatem BA, Jabir FA. The Role of ACE2 Receptor and Its Polymorphisms in COVID-19 Infection and Severity and Its Association with Lipid Profile, Thrombin, and D-Dimer Levels in Iraqi Patients: A Cross-Sectional Study. Biochem Genet 2024:10.1007/s10528-024-10890-7. [PMID: 39085685 DOI: 10.1007/s10528-024-10890-7] [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: 05/27/2023] [Accepted: 01/02/2024] [Indexed: 08/02/2024]
Abstract
COVID-19 patients experience a complex interplay involving ACE2, thrombin, D-dimer, and lipid profile, yet its full understanding remains elusive. ACE2, a pivotal regulator of the renin-angiotensin system and the primary receptor for SARS-CoV-2 undergoes downregulation upon viral binding, potentially leading to severe cases with acute respiratory distress syndrome (ARDS). A specific ACE2 gene polymorphism (rs2285666) may be associated with COVID-19 susceptibility, with the A allele potentially increasing infection risk. COVID-19 disease progression is linked to coagulation abnormalities, but the exact connection with thrombin and D-dimer remains uncertain. A study examining coagulation parameters in COVID-19 patients admitted to Al-Diwania Educational Hospital from February to May 2022 found that thrombin and D-dimer levels were directly related to disease severity. Severe cases exhibited significantly altered coagulation function compared to mild and recovered cases, with notably higher D-dimer levels and elevated thrombin serum concentrations. Moreover, dyslipidemia, particularly low HDL cholesterol, is a prevalent comorbidity in COVID-19 patients and may be linked to worse outcomes. In conclusion, COVID-19 is associated with a prothrombotic state and dysregulation of the renin-angiotensin system due to ACE2 downregulation following viral binding. The intricate interplay between ACE2, thrombin, D-dimer, and lipid profile necessitates further investigation. The multifaceted nature of the disease demands continued research to unravel its pathogenesis and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Ban Adnan Hatem
- Chemistry Department, College of Science, Al-Qadisiyah University, Al Diwaniyah, Iraq.
| | - Ferdous A Jabir
- Biochemistry Department, College of Medicine, Al-Qadisiyah University, Al Diwaniyah, Iraq
| |
Collapse
|
103
|
Rettew A, Garrahy I, Rahimian S, Brown R, Sangha N. COVID-19 Coagulopathy. Life (Basel) 2024; 14:953. [PMID: 39202695 PMCID: PMC11355811 DOI: 10.3390/life14080953] [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: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
Coronavirus disease of 2019 (COVID-19) is the respiratory viral infection caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite being a primary respiratory illness, it is commonly complicated by systemic involvement of the vasculature leading to arterial and venous thrombosis. In this review, we will focus on the association between COVID-19 and thrombosis. We will highlight the pathophysiology of COVID-19 coagulopathy. The clinical manifestations of COVID-19 vasculopathy will be discussed with a focus on venous and arterial thromboembolic events. COVID-19 vasculopathy and disseminated intravascular coagulation (DIC) are distinguished within, as well as areas of controversy, such as "long COVID". Finally, the current professional guidelines on prevention and treatment of thrombosis associated with SARS-CoV-2 infection will be discussed.
Collapse
Affiliation(s)
| | - Ian Garrahy
- Tower Health System, Reading Hospital, West Reading, PA 19611, USA; (A.R.); (S.R.); (R.B.); (N.S.)
| | | | | | | |
Collapse
|
104
|
Satoh K, Wada T, Tampo A, Takahashi G, Hoshino K, Matsumoto H, Taira T, Kazuma S, Masuda T, Tagami T, Ishikura H. Practical approach to thrombocytopenia in patients with sepsis: a narrative review. Thromb J 2024; 22:67. [PMID: 39039520 PMCID: PMC11265094 DOI: 10.1186/s12959-024-00637-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
Thrombocytopenia frequently occurs in patients with sepsis. Disseminated intravascular coagulation (DIC) may be a possible cause of thrombocytopenia owing to its high prevalence and association with poor outcomes; however, it is important to keep the presence of other diseases in mind in sepsis practice. Thrombotic microangiopathy (TMA), which is characterized by thrombotic thrombocytopenic purpura, Shiga toxin-producing Escherichia coli hemolytic uremic syndrome (HUS), and complement-mediated HUS, is characterized by thrombocytopenia, microangiopathic hemolytic anemia, and organ damage. TMA has become widely recognized in recent years because of the development of specific treatments. Previous studies have reported a remarkably lower prevalence of TMA than DIC; however, its epidemiology is not well defined, and there may be cases in which TMA is not correctly diagnosed, resulting in poor outcomes. Therefore, it is important to differentiate DIC from TMA. Nevertheless, differentiating between DIC and TMA remains a challenge as indicated by previous reports that most patients with TMA can be diagnosed as DIC using the universal coagulation scoring system. Several algorithms to differentiate sepsis-related DIC from TMA have been suggested, contributing to improving the care of septic patients with thrombocytopenia; however, it may be difficult to apply these algorithms to patients with coexisting DIC and TMA, which has recently been reported. This review describes the disease characteristics, including epidemiology, pathophysiology, and treatment, of DIC, TMA, and other diseases with thrombocytopenia and proposes a novel practical approach flow, which is characterized by the initiation of the diagnosis of TMA in parallel with the diagnosis of DIC. This practical flow also refers to the longitudinal diagnosis and treatment flow with TMA in mind and real clinical timeframes. In conclusion, we aim to widely disseminate the results of this review that emphasize the importance of incorporating consideration of TMA in the management of septic DIC. We anticipate that this practical new approach for the diagnostic and treatment flow will lead to the appropriate diagnosis and treatment of complex cases, improve patient outcomes, and generate new epidemiological evidence regarding TMA.
Collapse
Affiliation(s)
- Kasumi Satoh
- Advanced Emergency and Critical Care Center, Akita University Hospital, Akita, Japan
| | - Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Kita 15, Nishi 7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Akihito Tampo
- Department of Emergency Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Gaku Takahashi
- Department of Critical Care, Disaster and General Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Kota Hoshino
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Hironori Matsumoto
- Department of Emergency and Critical Care Medicine, Ehime University Graduate School of Medicine, Toon, Japan
| | - Takayuki Taira
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Satoshi Kazuma
- Department of Intensive Care Medicine, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Takamitsu Masuda
- Department of Emergency Medicine, Emergency and Critical Care Center, Fujieda Municipal General Hospital, Fujieda, Japan
| | - Takashi Tagami
- Department of Emergency and Critical Care Medicine, Nippon Medical School Musashikosugi Hospital, Tokyo, Japan
| | - Hiroyasu Ishikura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
105
|
Keita M, Seck M, Diallo AB, Touré SA, Bousso ES, Gueye SM, Dieng N, Dieng F, Faye BF, Diop S. Morbidity and Mortality Associated with COVID-19 and Acute Chest Syndrome in Sickle Cell Disease Patients. Hemoglobin 2024; 48:285-291. [PMID: 39034815 DOI: 10.1080/03630269.2024.2378069] [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: 02/02/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 07/23/2024]
Abstract
SUMMARYCOVID-19 infection has emerged as a comorbidity that can significantly increase morbidity and mortality in sickle cell patients with ACS (acute thoracic/chest syndrome). The aim of our study was to assess COVID-19-related morbidity and mortality in sickle cell patients with ACS. This was a retrospective, descriptive study of patient records followed over a 36-month period from January 2020 to December 2022. The study was conducted at the national blood transfusion center in Dakar. The sex ratio (M/F) was 0.82. The median age was 26 (17-39) years. The most represented age group was between 21 and 30 years. Factors associated with death were: at baseline, SS genotype, presence of comorbidities (asthma, chronic obstructive pulmonary disease, viral hepatitis B, ischemic heart disease), osteonecrosis of the femoral head, and use of NSAIDs (non-steroidal anti-inflammatory drugs) at diagnosis of COVID-19; at the diagnosis of ACS associated with COVID-19, respiratory distress, hypoxia (Sa02 < 92%), creatininemia >18.5 mg/l, CRP >192 mg/l, lymphopenia; the therapeutic modalities associated with death were: transfusion of RBCs (packed red blood cells) and curative anticoagulation. This study shows that patients with comorbidities and/or chronic complications of sickle cell disease can develop severe forms of ACS associated with COVID 19, leading to death. Other factors linked to death, notably diagnostic and therapeutic, were also identified in the course of this study.
Collapse
Affiliation(s)
- Mohamed Keita
- Clinical Hematology Department of National Blood Transfusion Center, Dakar, Senegal
| | - Moussa Seck
- Clinical Hematology Department of National Blood Transfusion Center, Dakar, Senegal
| | | | | | - Elimane Seydi Bousso
- Clinical Hematology Department of National Blood Transfusion Center, Dakar, Senegal
| | | | - Nata Dieng
- Department of Hematology, Cheikh Anta Diop University, Dakar, Senegal
| | - Fatma Dieng
- Clinical Hematology Department of National Blood Transfusion Center, Dakar, Senegal
| | - Blaise Felix Faye
- Clinical Hematology Department of National Blood Transfusion Center, Dakar, Senegal
| | - Saliou Diop
- Clinical Hematology Department of National Blood Transfusion Center, Dakar, Senegal
| |
Collapse
|
106
|
Dumitrescu TV, Meşină C, Ciorbagiu MC, Lascu LC, Honţaru SO, Ionovici N, Mogoantă L, Mogoantă SŞ. Postoperative multiple perforations of the small bowel in a patient with COVID-19 - case report. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:531-535. [PMID: 39529347 PMCID: PMC11657326 DOI: 10.47162/rjme.65.3.16] [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: 06/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, the infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presented quite a diverse symptomatology, in addition to respiratory symptoms, while other clinical signs such as thrombosis, postoperative hemorrhages, acute kidney or liver failure, digestive disorders (vomiting and diarrheal stools) were also reported. We present the case of a patient diagnosed with transverse colon neoplasm and asymptomatic SARS-CoV-2 infection, who presented to the Emergency Room (ER) of the Emergency County Clinical Hospital, Craiova, Romania, with a clinical picture of low intestinal occlusion. Surgery was decided and a right hemicolectomy extended to the left, with terminal ileostomy performed. The postoperative evolution was favorable, with the resumption of intestinal transit and discharge on the third postoperative day. The patient returned to the ER Department on the fifth day after surgery, with diffuse abdominal pain, absence of intestinal transit and flatulence. Clinical examination of the abdomen revealed the presence of bloating sounds on palpation. Emergency laparotomy was again performed with the suspicion of postoperative occlusion and five perforations were found in the small bowel, associated with fecaloid peritonitis and mechanic-inflammatory occlusion. The perforations were without any obvious lesion substrate, four of them being located on the jejunum and one on the terminal ileum. The histopathological examination revealed hemorrhage and recent transmural thrombosis on the intestinal wall, most likely caused by COVID-19. Without any respiratory symptoms, the COVID-19 infection caused multiple intestinal lesions, leading to peritonitis and septic shock, followed by the patient's death.
Collapse
Affiliation(s)
- Theodor Viorel Dumitrescu
- Department of Histology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Romania;
| | | | | | | | | | | | | | | |
Collapse
|
107
|
Zhao R, Li M, Xiao P, Song D, Li H. Advances in D-dimer testing: progress in harmonization of clinical assays and innovative detection methods. Anal Bioanal Chem 2024; 416:3737-3750. [PMID: 38503987 DOI: 10.1007/s00216-024-05207-x] [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: 12/08/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 03/21/2024]
Abstract
The D-dimer is a sensitive indicator of coagulation and fibrinolysis activation, especially valuable as a biomarker of intravascular thrombosis. Measurement of plasma D-dimer levels plays a crucial role in the diagnosis and monitoring of conditions such as deep vein thrombosis, pulmonary embolism, and disseminated intravascular coagulation. A variety of immunoassays, including enzyme-linked immunosorbent assays, latex-enhanced immunoturbidimetric assays, whole-blood aggregation analysis, and immunochromatography assays, are widely used in clinical settings to determine D-dimer levels. However, the results obtained from different D-dimer assays vary significantly. These assays exhibit intra-method coefficients of variation ranging from 6.4% to 17.7%, and the measurement discrepancies among different assays can be as high as 20-fold. The accuracy and reliability of D-dimer testing cannot be guaranteed due to the lack of an internationally endorsed reference measurement system (including reference materials and reference measurement procedures), which may lead to misdiagnosis and underdiagnosis, limiting its full clinical application. In this review, we present an in-depth analysis of clinical D-dimer testing, summarizing the existing challenges, the current state of metrology, and progress towards harmonization. We also review the latest advancements in D-dimer detection techniques, which include mass spectrometry and electrochemical and optical immunoassays. By comparing the basic principles, the definition of the measurand, and analytical performance of these methods, we provide an outlook on the potential improvements in D-dimer clinical testing.
Collapse
Affiliation(s)
- Rong Zhao
- National Institute of Metrology, Beijing, 100029, China
- Key Laboratory of Chemical Metrology and Applications on Nutrition and Health for State Market Regulation, Beijing, 100029, China
| | - Mengran Li
- Beijing University of Chemical Technology, Beijing, 100029, China
| | - Peng Xiao
- National Institute of Metrology, Beijing, 100029, China
- Key Laboratory of Chemical Metrology and Applications on Nutrition and Health for State Market Regulation, Beijing, 100029, China
| | - Dewei Song
- National Institute of Metrology, Beijing, 100029, China
| | - Hongmei Li
- National Institute of Metrology, Beijing, 100029, China.
- Key Laboratory of Chemical Metrology and Applications on Nutrition and Health for State Market Regulation, Beijing, 100029, China.
| |
Collapse
|
108
|
Nauriyal D, Dubey R, Agrawal P, Kumar D, Punj A, Nasser K. A cross-sectional study on clinical characteristics and severity of children with COVID-19 admitted to a teaching institute in North India. J Family Med Prim Care 2024; 13:2653-2662. [PMID: 39071009 PMCID: PMC11272026 DOI: 10.4103/jfmpc.jfmpc_1734_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: 10/25/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 07/30/2024] Open
Abstract
Background SARS-CoV-2 infection presentation in children is usually milder than in adults but can be severe and fatal as well. Data on the pediatric population regarding severity and clinical presentation are still limited, and there is a need to have a better understanding of clinical features, severity, and laboratory parameters. Aims and Objective To document clinical and laboratory characteristics and outcomes of children with SARS-CoV-2 in a low-middle-income country and to evaluate clinicodemographic factors and biochemical markers associated with severity and mortality. Materials and Methods A hospital-based cross-sectional study was conducted among 112 COVID-19-positive children at a designated Level-3 center in North India. Clinical characteristics, laboratory parameters, and severity of COVID-19 cases as well as factors associated with the severity of the disease, were analyzed by descriptive statistics and a Chi-square test. Results The adolescent age group (age 12-18 years) was affected most (64.3%). Male patients accounted for 56.3% of total cases. Fever was the most common symptom (41.1%) followed by cough. Presenting complaints were highest from the respiratory system (32.1%) followed by the gastrointestinal (8.9%) and the neurological system (7.1%). Majority of patients had mild disease (87%) while 13% had the moderate-severe disease. Spo2 < 95% (P = 0.00001), neutrophilia (P < 0.000001), lymphopenia (P < 0.000001), elevated values of C-reactive protein (P < 0.00001), Interleukin-6 (P = 0.002), D- dimer (P = 0.00014) and respiratory symptoms as presenting complaints (P < 0.000001) were found to be significantly associated with severity of disease. Conclusion The male and adolescent age group was affected most. Presenting complaints were highest from the respiratory system. Unusual presentation may have gastrointestinal or neurological presentation. Most children with COVID-19 had mild disease. Moderate to severe disease was not uncommon. Factors including neutrophilia, lymphopenia, elevated lab values of C-reactive protein, D-dimer, and interleukin-6 had a significant association with the severity of the disease. These biomarkers can help predict the severity of the disease.
Collapse
Affiliation(s)
- Deepty Nauriyal
- Department of Pediatrics, Subharti Medical, College, Meerut, Uttar Pradesh, India
| | - Rishabh Dubey
- Department of Pediatrics, Subharti Medical, College, Meerut, Uttar Pradesh, India
| | - Pulak Agrawal
- Department of Pediatrics, Subharti Medical, College, Meerut, Uttar Pradesh, India
| | - Deepak Kumar
- Department of Community Medicine, Maharishi Markandeshwar University and Hospital, Solan, Himachal Pradesh, India
| | - Ajay Punj
- Department of Pediatrics, Subharti Medical, College, Meerut, Uttar Pradesh, India
| | - Kaynat Nasser
- Department of Community Medicine, Subharti Medical College, Meerut, Uttar Pradesh, India
| |
Collapse
|
109
|
Lu L, Cao L, Zhang J, Lin B. Cutaneous manifestations associated with COVID-19 infection at a university hospital in eastern China. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2024; 13:117-132. [PMID: 39022792 PMCID: PMC11249857 DOI: 10.62347/bduc7952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/12/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) affects different organ systems, including the skin. A retrospective analysis of skin manifestations in Chinese outpatient and inpatient settings is lacking. The study aims to analyze cutaneous manifestations in COVID-19 patients and the recurrence or aggravation of previous skin diseases. MATERIALS AND METHODS A retrospective cross-sectional study was conducted from November 2022 to July 2023 in a university hospital in eastern China. It involved reverse transcriptase polymerase chain reaction (RT-PCR)-positive COVID-19 patients, documenting various skin manifestations and the recurrence or aggravation of pre-existing skin conditions. The pattern of skin lesions and other variables were assessed. RESULTS The study included 303 patients, with 127 males and 176 females. Maculopapular rash was the predominant new cutaneous manifestation (54.92%), mainly in middle-aged individuals. Other findings included urticaria (16.39%), herpes zoster (11.89%), and herpes simplex (4.10%), vesicular rashes (2.46%), purpura (2.05%), erythema multiforme (1.64%), livedo reticularis (0.41%) and so on. Severe disease was associated with herpes zoster and livedo reticularis. Critical COVID-19 cases were linked to vesicular rashes, purpura, and erythema multiforme. The mean time for skin lesion emergence post-infection varied from 3 days for seborrheic dermatitis to 17.48 days for herpes zoster. Vasculitic manifestations correlated with elevated D-dimer levels. A total of 59 cases (19.47%) of recurrent or aggravated skin diseases were reported following infection with COVID-19, with dermatitis being the most common, followed by acne and folliculitis, psoriasis, urticaria, bullous pemphigoid, pemphigus, tinea corporis and androgenetic alopecia. CONCLUSION The cutaneous phenotypes delineated in this study expand the dermatologic spectrum associated with COVID-19. Cutaneous manifestations may result from overactive immune responses, complement activation, and microvascular damage. Herpes zoster typically occurs in elderly COVID-19 patients with weaker immune systems or more severe diseases. Purpura and livedo reticularis, although rare, may indicate disease severity. It is possible to predict the course of COVID-19 with different severity through cutaneous manifestations. Recognizing these skin manifestations could aid in predicting COVID-19 severity and guide dermatologists in managing the pandemic response.
Collapse
Affiliation(s)
- Lingyi Lu
- Department of Dermatology, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Ningbo 315010, Zhejiang, P. R. China
| | - Lu Cao
- Department of Dermatology, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Ningbo 315010, Zhejiang, P. R. China
| | - Jing Zhang
- Department of Dermatology, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Ningbo 315010, Zhejiang, P. R. China
| | - Bingjiang Lin
- Department of Dermatology, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Ningbo 315010, Zhejiang, P. R. China
| |
Collapse
|
110
|
Xiao Z, Lin M, Song N, Wu X, Hou J, Wang L, Tian X, An C, Dela Cruz CS, Sharma L, Chang D. Clinical features and multiomics profiles indicate coagulation and platelet dysfunction in COVID-19 viral sepsis. iScience 2024; 27:110110. [PMID: 38974472 PMCID: PMC11225851 DOI: 10.1016/j.isci.2024.110110] [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: 03/01/2024] [Revised: 04/16/2024] [Accepted: 05/23/2024] [Indexed: 07/09/2024] Open
Abstract
Increased cases of sepsis during COVID-19 in the absence of known bacterial pathogens highlighted role of viruses as causative agents of sepsis. In this study, we investigated clinical, laboratory, proteomic, and metabolomic characteristics of viral sepsis patients (n = 45) and compared them to non-sepsis patients with COVID-19 (n = 186) to identify molecular mechanisms underlying the pathology of viral sepsis in COVID-19. We identified unique metabolomic and proteomic signatures that suggest a substantial perturbation in the coagulation, complement, and platelet activation pathways in viral sepsis. Our proteomic data indicated elevated coagulation pathway protein (fibrinogen), whereas a decrease in many of the complement proteins was observed. These alterations were associated with the functional consequences such as susceptibility to secondary bacterial infections and potentially contributing to both local and systemic disease phenotypes. Our data provide novel aspect of COVID-19 pathology that is centered around presence of sepsis phenotype in COVID-19.
Collapse
Affiliation(s)
- Zhiqing Xiao
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Hebei North University, Zhangjiakou 075000, Hebei, China
| | - Minggui Lin
- Beijing Tsinghua Changgung Hospital, Tsinghua University School of Medicine, Beijing 102218, China
| | - Ning Song
- Department of Infectious Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Xue Wu
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jingyu Hou
- Department of Infectious Diseases, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Lili Wang
- Beijing Tsinghua Changgung Hospital, Tsinghua University School of Medicine, Beijing 102218, China
| | - XinLun Tian
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chunge An
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Charles S. Dela Cruz
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Lokesh Sharma
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - De Chang
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
111
|
Li K, Feng KC, Simon M, Fu Y, Galanakis D, Mueller S, Rafailovich MH. Molecular Basis for Surface-Initiated Non-Thrombin-Generated Clot Formation Following Viral Infection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30703-30714. [PMID: 38848451 DOI: 10.1021/acsami.4c02918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
In this paper, we propose a model that connects two standard inflammatory responses to viral infection, namely, elevation of fibrinogen and the lipid drop shower, to the initiation of non-thrombin-generated clot formation. In order to understand the molecular basis for the formation of non-thrombin-generated clots following viral infection, human epithelial and Madin-Darby Canine Kidney (MDCK, epithelial) cells were infected with H1N1, OC43, and adenovirus, and conditioned media was collected, which was later used to treat human umbilical vein endothelial cells and human lung microvascular endothelial cells. After direct infection or after exposure to conditioned media from infected cells, tissue surfaces of both epithelial and endothelial cells, exposed to 8 mg/mL fibrinogen, were observed to initiate fibrillogenesis in the absence of thrombin. No fibers were observed after direct viral exposure of the endothelium or when the epithelium cells were exposed to SARS-CoV-2 isolated spike proteins. Heating the conditioned media to 60 °C had no effect on fibrillogenesis, indicating that the effect was not enzymatic but rather associated with relatively thermally stable inflammatory factors released soon after viral infection. Spontaneous fibrillogenesis had previously been reported and interpreted as being due to the release of the alpha C domains due to strong interactions of the interior of the fibrinogen molecule in contact with hydrophobic material surfaces rather than cleavage of the fibrinopeptides. Contact angle goniometry and immunohistochemistry were used to demonstrate that the lipids produced within the epithelium and released in the conditioned media, probably after the death of infected epithelial cells, formed a hydrophobic residue responsible for fibrillogenesis. Hence, the standard inflammatory response constitutes the ideal conditions for surface-initiated clot formation.
Collapse
Affiliation(s)
- Kao Li
- School of Biomedicine and Nursing, Shandong Institute of Petroleum and Chemical Technology, Dongying 257061, Shandong, China
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Kuan-Che Feng
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Marcia Simon
- Department of Oral Biology and Pathology, Stony Brook University Medical Center, Stony Brook, New York 11794, United States
| | - Yuyang Fu
- Dongying Stem Cell Bank Medical Technology Co., Ltd., Dongying 257000, Shandong, China
| | - Dennis Galanakis
- Department of Pathology, Stony Brook University School of Medicine, Stony Brook, New York 11720, United States
| | | | - Miriam H Rafailovich
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
112
|
Shao HH, Yin RX. Pathogenic mechanisms of cardiovascular damage in COVID-19. Mol Med 2024; 30:92. [PMID: 38898389 PMCID: PMC11186295 DOI: 10.1186/s10020-024-00855-2] [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: 03/09/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND COVID-19 is a new infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS CoV-2). Since the outbreak in December 2019, it has caused an unprecedented world pandemic, leading to a global human health crisis. Although SARS CoV-2 mainly affects the lungs, causing interstitial pneumonia and severe acute respiratory distress syndrome, a number of patients often have extensive clinical manifestations, such as gastrointestinal symptoms, cardiovascular damage and renal dysfunction. PURPOSE This review article discusses the pathogenic mechanisms of cardiovascular damage in COVID-19 patients and provides some useful suggestions for future clinical diagnosis, treatment and prevention. METHODS An English-language literature search was conducted in PubMed and Web of Science databases up to 12th April, 2024 for the terms "COVID-19", "SARS CoV-2", "cardiovascular damage", "myocardial injury", "myocarditis", "hypertension", "arrhythmia", "heart failure" and "coronary heart disease", especially update articles in 2023 and 2024. Salient medical literatures regarding the cardiovascular damage of COVID-19 were selected, extracted and synthesized. RESULTS The most common cardiovascular damage was myocarditis and pericarditis, hypertension, arrhythmia, myocardial injury and heart failure, coronary heart disease, stress cardiomyopathy, ischemic stroke, blood coagulation abnormalities, and dyslipidemia. Two important pathogenic mechanisms of the cardiovascular damage may be direct viral cytotoxicity as well as indirect hyperimmune responses of the body to SARS CoV-2 infection. CONCLUSIONS Cardiovascular damage in COVID-19 patients is common and portends a worse prognosis. Although the underlying pathophysiological mechanisms of cardiovascular damage related to COVID-19 are not completely clear, two important pathogenic mechanisms of cardiovascular damage may be the direct damage of the SARSCoV-2 infection and the indirect hyperimmune responses.
Collapse
Affiliation(s)
- Hong-Hua Shao
- Department of Infectious Diseases, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People's Hospital of Nanning, No. 1 Erli, Changgang Road, Nanning, Guangxi, 530023, People's Republic of China
| | - Rui-Xing Yin
- Department of Infectious Diseases, HIV/AIDS Clinical Treatment Center of Guangxi (Nanning), The Fourth People's Hospital of Nanning, No. 1 Erli, Changgang Road, Nanning, Guangxi, 530023, People's Republic of China.
- Department of Cardiology, Institute of Cardiovascular Diseases, The First Affiliated Hospital, Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China.
| |
Collapse
|
113
|
Pottecher J, Raffi F, Jandrot-Perrus M, Binay S, Comenducci A, Desort-Henin V, François D, Gharakhanian S, Labart M, Meilhoc A, Toledano E, Pletan Y, Avenard G, Sato VH, the GARDEN Investigators. Targeting GPVI with glenzocimab in COVID-19 patients: Results from a randomized clinical trial. PLoS One 2024; 19:e0302897. [PMID: 38885234 PMCID: PMC11182546 DOI: 10.1371/journal.pone.0302897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/11/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Glenzocimab is a novel antithrombotic agent which targets platelet glycoprotein VI (GPVI) and does not induce haemorrhage. SARS-CoV-2 triggers a prothrombotic state and lung injury whose mechanisms include coagulopathy, endothelial dysfunction, and inflammation with dysregulated platelets. METHODS AND PATIENTS GARDEN was a randomised double-blind, exploratory phase II study of glenzocimab in SARS-CoV-2 respiratory failure (NCT04659109). PCR+ adults in Brazil and France (7 centres) were randomized to standard-of-care (SOC) plus glenzocimab (1000 mg/dayx3 days) or placebo, followed for 40 days. Primary efficacy endpoint was clinical progression at Day 4. All analyses concerned the intention-to-treat population. RESULTS Between December 2020 and August 2021, 61 patients received at least one dose (30 glenzocimab vs 32 placebo) and 58 completed the study (29 vs 29). Clinical progression of COVID-19 ARDS was not statistically different between glenzocimab and placebo arms (43.3% and 29.0%, respectively; p = 0.245). Decrease in the NEWS-2 category at D4 was statistically significant (p = 0.0290) in the glenzocimab arm vs placebo. No Serious Adverse Event (SAE) was deemed related to study drug; bleeding related events were reported in 6 patients (7 events) and 4 patients (4 events) in glenzocimab and placebo arms, respectively. CONCLUSIONS Therapeutic GPVI inhibition assessment during COVID-19 was conducted in response to a Public Health emergency. Glenzocimab in coagulopathic patients under therapeutic heparin was neither associated with increased bleeding, nor SAE. Clinical impact of glenzocimab on COVID-19 ARDS was not demonstrated. A potential role for GPVI inhibition in other types of ARDS deserves further experimentation. Glenzocimab is currently studied in stroke (ACTISAVE: NCT05070260) and cardiovascular indications.
Collapse
Affiliation(s)
- Julien Pottecher
- Strasbourg University Hospital, UR3072, FHU OMICARE, FMTS, Strasbourg, France
| | - Francois Raffi
- Nantes Université, CHU Nantes, INSERM, Department of Infectious Diseases, CIC 1413, Nantes, France
| | | | | | | | | | | | - Shahin Gharakhanian
- Acticor-Biotech, Paris, France
- Shahin Gharakhanian MD Consulting LLC, Cambridge Innovation Center, Cambridge, MA, United States of America
| | | | | | | | | | | | - Victor H. Sato
- International Research Center, Hospital Alemão Oswaldo Cruz, Sao Paulo, Brazil
| | | |
Collapse
|
114
|
Nechita LC, Ignat MD, Balta AAS, Barbu RE, Baroiu L, Voinescu DC, Nechita A, Debita M, Busila C, Stefanopol IA. The Impact of Cardiovascular Antecedents on the Prognosis of COVID-19 Critically Ill Patients. J Clin Med 2024; 13:3518. [PMID: 38930047 PMCID: PMC11205074 DOI: 10.3390/jcm13123518] [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: 04/24/2024] [Revised: 05/17/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Background/Objectives: The objective of the study is to analyze the impact of cardiovascular history on mortality in COVID-19 patients, hospitalized in the intensive care unit with indications for continuous positive airway pressure (CPAP) and subsequently mechanical ventilation, without oncological disease. Methods: A retrospective observational study was carried out on a group of 108 critical COVID-19 patients. We compared demographic data, paraclinical and clinical parameters, days of hospitalization, and mortality rate between two groups of patients, one group with a history of cardiovascular disease (81 patients) and a group without a history of cardiovascular disease (27 patients). Results: Patients with cardiovascular antecedents had a higher mortality rate than those without cardiovascular antecedents, presenting severe forms with shorter survival time in the intensive care unit and increased inflammatory evidence. Compared to patients without a history of cardiovascular illness, those with cardiovascular disease had a lower average age, and developed a severe form of COVID-19. Conclusions: Cardiovascular antecedents can worsen the prognosis of patients with COVID-19, requiring a careful screening and multidisciplinary approach.
Collapse
Affiliation(s)
- Luiza Camelia Nechita
- Doctoral School of Biomedical Sciences, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.C.N.); (A.A.S.B.); (R.E.B.)
| | - Mariana Daniela Ignat
- Doctoral School of Biomedical Sciences, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.C.N.); (A.A.S.B.); (R.E.B.)
| | | | - Raisa Eloise Barbu
- Doctoral School of Biomedical Sciences, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.C.N.); (A.A.S.B.); (R.E.B.)
| | - Liliana Baroiu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (D.C.V.); (A.N.); (C.B.)
- ‘Sf. Cuv. Parascheva’ Clinical Hospital of Infectious Diseases, 800179 Galati, Romania;
| | - Doina Carina Voinescu
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (D.C.V.); (A.N.); (C.B.)
- ‘Sf. Apostol Andrei’ Clinical Emergency County Hospital, 800578 Galati, Romania
| | - Aurel Nechita
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (D.C.V.); (A.N.); (C.B.)
- ‘Sf. Ioan’ Clinical Hospital for Children, 800487 Galati, Romania;
| | - Mihaela Debita
- ‘Sf. Cuv. Parascheva’ Clinical Hospital of Infectious Diseases, 800179 Galati, Romania;
- Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania
| | - Camelia Busila
- Clinical Medical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania; (L.B.); (D.C.V.); (A.N.); (C.B.)
- ‘Sf. Ioan’ Clinical Hospital for Children, 800487 Galati, Romania;
| | - Ioana Anca Stefanopol
- ‘Sf. Ioan’ Clinical Hospital for Children, 800487 Galati, Romania;
- Clinical Surgical Department, Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University, 800008 Galati, Romania
| |
Collapse
|
115
|
Li X, Mi Z, Liu Z, Rong P. SARS-CoV-2: pathogenesis, therapeutics, variants, and vaccines. Front Microbiol 2024; 15:1334152. [PMID: 38939189 PMCID: PMC11208693 DOI: 10.3389/fmicb.2024.1334152] [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: 11/06/2023] [Accepted: 05/29/2024] [Indexed: 06/29/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in December 2019 with staggering economic fallout and human suffering. The unique structure of SARS-CoV-2 and its underlying pathogenic mechanism were responsible for the global pandemic. In addition to the direct damage caused by the virus, SARS-CoV-2 triggers an abnormal immune response leading to a cytokine storm, culminating in acute respiratory distress syndrome and other fatal diseases that pose a significant challenge to clinicians. Therefore, potential treatments should focus not only on eliminating the virus but also on alleviating or controlling acute immune/inflammatory responses. Current management strategies for COVID-19 include preventative measures and supportive care, while the role of the host immune/inflammatory response in disease progression has largely been overlooked. Understanding the interaction between SARS-CoV-2 and its receptors, as well as the underlying pathogenesis, has proven to be helpful for disease prevention, early recognition of disease progression, vaccine development, and interventions aimed at reducing immunopathology have been shown to reduce adverse clinical outcomes and improve prognosis. Moreover, several key mutations in the SARS-CoV-2 genome sequence result in an enhanced binding affinity to the host cell receptor, or produce immune escape, leading to either increased virus transmissibility or virulence of variants that carry these mutations. This review characterizes the structural features of SARS-CoV-2, its variants, and their interaction with the immune system, emphasizing the role of dysfunctional immune responses and cytokine storm in disease progression. Additionally, potential therapeutic options are reviewed, providing critical insights into disease management, exploring effective approaches to deal with the public health crises caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Xi Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
116
|
Milentijević M, Katanić N, Joksimović B, Pavlović A, Filimonović J, Anđelković M, Bojović K, Elek Z, Ristić S, Vasiljević M, Stevanović J, Radomirović D, Elez-Burnjaković N, Lalović N, Kulić M, Kulić J, Milić M. The Impact of Cytokines on Coagulation Profile in COVID-19 Patients: Controlled for Socio-Demographic, Clinical, and Laboratory Parameters. Biomedicines 2024; 12:1281. [PMID: 38927488 PMCID: PMC11201770 DOI: 10.3390/biomedicines12061281] [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: 03/21/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Background: Severe coagulation abnormalities are common in patients with COVID-19 infection. We aimed to investigate the relationship between pro-inflammatory cytokines and coagulation parameters concerning socio-demographic, clinical, and laboratory characteristics. Methods: Our study included patients hospitalized during the second wave of COVID-19 in the Republic of Serbia. We collected socio-demographic, clinical, and blood-sample data for all patients. Cytokine levels were measured using flow cytometry. Results: We analyzed data from 113 COVID-19 patients with an average age of 58.15 years, of whom 79 (69.9%) were male. Longer duration of COVID-19 symptoms before hospitalization (B = 69.672; p = 0.002) and use of meropenem (B = 1237.220; p = 0.014) were predictive of higher D-dimer values. Among cytokines, higher IL-5 values significantly predicted higher INR values (B = 0.152; p = 0.040) and longer prothrombin times (B = 0.412; p = 0.043), and higher IL-6 (B = 0.137; p = 0.003) predicted longer prothrombin times. Lower IL-17F concentrations at admission (B = 0.024; p = 0.050) were predictive of higher INR values, and lower IFN-γ values (B = -0.306; p = 0.017) were predictive of higher aPTT values. Conclusions: Our findings indicate a significant correlation between pro-inflammatory cytokines and coagulation-related parameters. Factors such as the patient's level of education, gender, oxygen-therapy use, symptom duration before hospitalization, meropenem use, and serum concentrations of IL-5, IL-6, IL-17F, and IFN-γ were associated with worse coagulation-related parameters.
Collapse
Affiliation(s)
- Milica Milentijević
- Department of Infective Diseases, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.M.); (N.K.)
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Nataša Katanić
- Department of Infective Diseases, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.M.); (N.K.)
| | - Bojan Joksimović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Aleksandar Pavlović
- Department of Surgery, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia;
| | - Jelena Filimonović
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| | - Milena Anđelković
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Ksenija Bojović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Zlatan Elek
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
- Department of Surgery, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia;
| | - Siniša Ristić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Miloš Vasiljević
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Jasmina Stevanović
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| | - Danica Radomirović
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Nikolina Elez-Burnjaković
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Nenad Lalović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Milan Kulić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Jovan Kulić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Marija Milić
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| |
Collapse
|
117
|
Zhu J, Bouzid R, Travert B, Géri G, Cohen Y, Picod A, Heming N, Rottman M, Joly-Laffargue B, Veyradier A, Capron C, Coppo P. Combined coagulation and inflammation markers as predictors of venous thrombo-embolism and death in COVID-19. Front Med (Lausanne) 2024; 11:1399335. [PMID: 38915768 PMCID: PMC11194426 DOI: 10.3389/fmed.2024.1399335] [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: 03/11/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024] Open
Abstract
Background The COVID-19 pandemic related to SARS-CoV-2 virus was responsible for global pandemic. The severe form of the disease was linked to excessive activation of immune pathways together with a systemic cytokine storm response and thrombotic venous or arterial complications. Factors predicting severe outcomes including venous and/or pulmonary thrombosis (VT) and death were identified, but the prognostic role of their combination was not addressed extensively. Objectives We investigated the role of prognostic factors from the coagulation or inflammatory pathways to better understand the outcome of the disease. Methods For this, we prospectively studied 167 SARS-CoV-2-positive patients from admission in intensive care units (ICU) or emergency departments from four academic hospitals over a 14-month period. Besides standard biology, we assessed serum concentrations of inflammatory markers, coagulation factors and peripheral blood cells immunophenotyping. Results Thirty-nine patients (23.3%) developed VT and 30 patients (18%) died. By univariate analysis, C-reactive protein (CRP) level > 150 mg/L, interleukin-6 (IL-6) ≥ 20 pg/mL, D-dimers > 1,500 μg/L, ADAMTS13 activity ≤ 50%, Von. Conclusion A combination of coagulation and inflammatory markers can refine the prognostication of severe outcome in COVID-19, and could be useful for the initial evaluation of other types of viral infection.
Collapse
Affiliation(s)
- Jaja Zhu
- Service d’Hématologie-Immunologie-Transfusion, AP-HP Paris Saclay, CHU Ambroise Paré, Université de Versailles Saint Quentin-Université Paris Saclay, Montigny-le-Bretonneux, France
- Laboratoire Cellules Souches et Applications Thérapeutiques, UMR INSERM 1184, Commissariat à l’Energie Atomique et aux Energies Alternatives, Fontenay-aux-Roses, France
| | - Raïda Bouzid
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
| | - Benoît Travert
- Service de Médecine Interne, Hôpital Ambroise-Paré, AP-HP, Boulogne-Billancourt, France
| | - Guillaume Géri
- Service de Médecine Intensive et Réanimation, Hôpital Ambroise-Paré, AP-HP, Boulogne-Billancourt, France
| | - Yves Cohen
- Service de Médecine Intensive et Réanimation, Hôpital Avicenne, AP-HP, Paris, France
| | - Adrien Picod
- Service de Médecine Intensive et Réanimation, Hôpital Avicenne, AP-HP, Paris, France
| | - Nicholas Heming
- Department of Intensive Care, Raymond Poincaré Hospital, APHP University Versailles Saint Quentin-University Paris Saclay, Garches, France
- Institut Hospitalo Universitaire PROMETHEUS, Garches, France
- Innovative Biomarkers Plateform, Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France
- FHU SEPSIS, Garches, France
| | - Martin Rottman
- Innovative Biomarkers Plateform, Laboratory of Infection & Inflammation-U1173, School of Medicine, INSERM, University Versailles Saint Quentin-University Paris Saclay, Garches, France; FHU SEPSIS, Garches, France
- General Intensive Care Unit, Raymond Poincaré Hospital (AP-HP), FHU SEPSIS, Laboratory of Infection and Inflammation-U1173, School of Medicine Simone Veil, Université Versailles Saint Quentin, University Paris Saclay, INSERM, Garches, France
| | - Bérangère Joly-Laffargue
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- EA3518, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Service D’hématologie Biologique, Laboratoire ADAMTS13, Hôpital Lariboisière, AP-HP Nord, Université de Paris, Paris, France
| | - Agnès Veyradier
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- EA3518, Institut de Recherche Saint Louis, Université de Paris, Paris, France
- Service D’hématologie Biologique, Laboratoire ADAMTS13, Hôpital Lariboisière, AP-HP Nord, Université de Paris, Paris, France
| | - Claude Capron
- Service d’Hématologie-Immunologie-Transfusion, AP-HP Paris Saclay, CHU Ambroise Paré, Université de Versailles Saint Quentin-Université Paris Saclay, Montigny-le-Bretonneux, France
- Université Paris-Saclay, Université de Versailles Saint Quentin en Yvelines (UVSQ), Biomarqueurs en cancérologie et onco-hématologie (BECCOH), Boulogne-Billancourt, France
| | - Paul Coppo
- Centre de Référence des Microangiopathies Thrombotiques (CNR-MAT), AP-HP, Paris, France
- Service d’Hématologie, Hôpital Saint-Antoine, AP-HP-Sorbonne Université, Paris, France
- NSERM UMRS 1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
118
|
Zhou D, Zhao S, He K, Liu Q, Zhang F, Pu Z, Xiao L, Zhang L, Chen S, Qian X, Wu X, Shen Y, Yu L, Zhang H, Jin J, Xu M, Wang X, Zhu D, Xie Z, Xu X. Longitudinal dynamic single-cell mass cytometry analysis of peripheral blood mononuclear cells in COVID-19 patients within 6 months after viral RNA clearance. BMC Infect Dis 2024; 24:567. [PMID: 38844850 PMCID: PMC11157885 DOI: 10.1186/s12879-024-09464-0] [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: 09/09/2023] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
This study investigates the longitudinal dynamic changes in immune cells in COVID-19 patients over an extended period after recovery, as well as the interplay between immune cells and antibodies. Leveraging single-cell mass spectrometry, we selected six COVID-19 patients and four healthy controls, dissecting the evolving landscape within six months post-viral RNA clearance, alongside the levels of anti-spike protein antibodies. The T cell immunophenotype ascertained via single-cell mass spectrometry underwent validation through flow cytometry in 37 samples. Our findings illuminate that CD8 + T cells, gamma-delta (gd) T cells, and NK cells witnessed an increase, in contrast to the reduction observed in monocytes, B cells, and double-negative T (DNT) cells over time. The proportion of monocytes remained significantly elevated in COVID-19 patients compared to controls even after six-month. Subpopulation-wise, an upsurge manifested within various T effector memory subsets, CD45RA + T effector memory, gdT, and NK cells, whereas declines marked the populations of DNT, naive and memory B cells, and classical as well as non-classical monocytes. Noteworthy associations surfaced between DNT, gdT, CD4 + T, NK cells, and the anti-S antibody titer. This study reveals the changes in peripheral blood mononuclear cells of COVID-19 patients within 6 months after viral RNA clearance and sheds light on the interactions between immune cells and antibodies. The findings from this research contribute to a better understanding of immune transformations during the recovery from COVID-19 and offer guidance for protective measures against reinfection in the context of viral variants.
Collapse
Affiliation(s)
- Diwenxin Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Shuai Zhao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Keting He
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Qiuhong Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Fen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Zhangya Pu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Lanlan Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Lingjian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Shangci Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaohan Qian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaoxin Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Yangfan Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Ling Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Huafen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Jiandi Jin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Min Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Xiaoyan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China
| | - Zhongyang Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China.
| | - Xiaowei Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City, 310003, China.
| |
Collapse
|
119
|
Abbas Q, Ali H, Amjad F, Hussain MZH, Rahman AR, Khan MH, Padhani ZA, Abbas F, Imam D, Alikhan Z, Belgaumi SM, Mohsin S, Sattar F, Siddiqui A, Lassi ZS, Das JK. Clinical presentation, diagnosis and management of multisystem inflammatory syndrome in children (MIS-C): a systematic review. BMJ Paediatr Open 2024; 8:e002344. [PMID: 38844384 PMCID: PMC11163633 DOI: 10.1136/bmjpo-2023-002344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/16/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Knowledge about multisystem inflammatory syndrome in children (MIS-C) is evolving, and evidence-based standardised diagnostic and management protocols are lacking. Our review aims to summarise the clinical and diagnostic features, management strategies and outcomes of MIS-C and evaluate the variances in disease parameters and outcomes between high-income countries (HIC) and middle-income countries (MIC). METHODS We searched four databases from December 2019 to March 2023. Observational studies with a sample size of 10 or more patients were included. Mean and prevalence ratios for various variables were pooled by random effects model using R. A mixed generalised linear model was employed to account for the heterogeneity, and publication bias was assessed via funnel and Doi plots. The primary outcome was pooled mean mortality among patients with MIS-C. Subgroup analysis was conducted based on the income status of the country of study. RESULTS A total of 120 studies (20 881 cases) were included in the review. The most common clinical presentations were fever (99%; 95% CI 99.6% to 100%), gastrointestinal symptoms (76.7%; 95% CI 73.1% to 79.9%) and dermatological symptoms (63.3%; 95% CI 58.7% to 67.7%). Laboratory investigations suggested raised inflammatory, coagulation and cardiac markers. The most common management strategies were intravenous immunoglobulins (87.5%; 95% CI 82.9% to 91%) and steroids (74.7%; 95% CI 68.7% to 79.9%). Around 53.1% (95% CI 47.3% to 58.9%) required paediatric intensive care unit admissions, and overall mortality was 3.9% (95% CI 2.7% to 5.6%). Patients in MIC were younger, had a higher frequency of respiratory distress and evidence of cardiac dysfunction, with a longer hospital and intensive care unit stay and had a higher mortality rate than patients in HIC. CONCLUSION MIS-C is a severe multisystem disease with better mortality outcomes in HIC as compared with MIC. The findings emphasise the need for standardised protocols and further research to optimise patient care and address disparities between HIC and MIC. PROSPERO REGISTRATION NUMBER CRD42020195823.
Collapse
Affiliation(s)
- Qalab Abbas
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Haider Ali
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Fatima Amjad
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | | | - Abdu R Rahman
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Maryam Hameed Khan
- Institute for Global Health and Development, The Aga Khan University, Karachi, Sind, Pakistan
| | - Zahra A Padhani
- School of Public Health, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Fatima Abbas
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Danyal Imam
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Zuviya Alikhan
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Sameer M Belgaumi
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Shazia Mohsin
- Department of Pediatric cardiology, Division of cardiothoracic sciences, Sindh institute of Urology and Transplantation (SIUT), Karachi, Sind, Pakistan
| | - Faiza Sattar
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Arsalan Siddiqui
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
| | - Zohra S Lassi
- School of Public Health, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Jai K Das
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi, Sind, Pakistan
- Institute for Global Health and Development, The Aga Khan University, Karachi, Sind, Pakistan
| |
Collapse
|
120
|
Xue Y, Feng W, Shi L, Cui N, Zhang W, Dong J, Li C, Hu J, Wei J. Review of clinical characteristics and mortality outcomes in patients on maintenance hemodialysis during the Omicron surge: a single center experience. BMC Public Health 2024; 24:1481. [PMID: 38831260 PMCID: PMC11145803 DOI: 10.1186/s12889-024-18999-5] [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: 01/05/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND This hemodialysis center experienced the pandemic from December 2022 to January 2023. Therefore, we sought to describe the clinical characteristics and mortality outcomes in hemodialysis patients during this Omicron surge. METHODS According to whether they are infected, they are divided into two groups: SARS-CoV-2-positive and SARS-CoV-2-negative. The SARS-CoV-2-positive group was divided into a survival group and a non-survival group for comparison. RESULTS 366 of 457 hemodialysis patients were infected with SARS-CoV-2. The most common symptoms observed were fever (43.2%) and cough (29.8%), Followed by diarrhea (1.4%). Hemodialysis patients with hypertension were more susceptible to SARS-CoV-2 infection. The lymphocyte count, serum creatinine, serum potassium, and serum phosphorus in the SARS-CoV-2-positive group were significantly lower than those in the SARS-CoV-2-negative group. The all-cause mortality rate for infection with SARS-CoV-2 was 5.2%. Only 7 of 366 SARS-CoV-2-positive patients were admitted to the intensive care unit, but 6 of them died. Intensive care unit hospitalization rates were significantly higher in the non-survival group compared with the survival group. White blood cells count, neutrophil count, C-reactive protein, AST, and D-dimer in the non-survival group were higher than those in the survival group. The lymphocyte count, hemoglobin concentration, serum creatinine, serum albumin, serum phosphorus and parathyroid hormone in the non-survival group were lower than those in the survival group. Age > 65 years, elevated C-reactive protein and AST are independent risk factors for death. Finally, no significant difference in vaccination status was found between the SARS-CoV-2-positive group and the negative group. CONCLUSIONS Hemodialysis patients are at high risk for SARS-CoV-2 infection. Ensuring the adequacy of hemodialysis treatment and maintaining good physical condition of patients are the top priorities.
Collapse
Affiliation(s)
- Yiyang Xue
- Health Science Center, Ningbo University, 818 Fenghua Rd, 315211, Ningbo, Zhejiang, P. R. China
| | - Weiwei Feng
- Blood Purification Center, Ningbo Urol & Nephrol Hospital, 998 Qianhe Rd, 315100, Ningbo, Zhejiang, P. R. China
| | - Ling Shi
- Blood Purification Center, Ningbo Urol & Nephrol Hospital, 998 Qianhe Rd, 315100, Ningbo, Zhejiang, P. R. China
| | - Ning Cui
- Blood Purification Center, Ningbo Urol & Nephrol Hospital, 998 Qianhe Rd, 315100, Ningbo, Zhejiang, P. R. China
| | - Wei Zhang
- Blood Purification Center, Ningbo Urol & Nephrol Hospital, 998 Qianhe Rd, 315100, Ningbo, Zhejiang, P. R. China
| | - Junxiu Dong
- Blood Purification Center, Ningbo Urol & Nephrol Hospital, 998 Qianhe Rd, 315100, Ningbo, Zhejiang, P. R. China
| | - Chunying Li
- Blood Purification Center, Ningbo Urol & Nephrol Hospital, 998 Qianhe Rd, 315100, Ningbo, Zhejiang, P. R. China
| | - Jinjin Hu
- Blood Purification Center, Ningbo Urol & Nephrol Hospital, 998 Qianhe Rd, 315100, Ningbo, Zhejiang, P. R. China
| | - Junjun Wei
- Blood Purification Center, Ningbo Urol & Nephrol Hospital, 998 Qianhe Rd, 315100, Ningbo, Zhejiang, P. R. China.
| |
Collapse
|
121
|
Iba T, Levy JH, Maier CL, Connors JM, Levi M. Four years into the pandemic, managing COVID-19 patients with acute coagulopathy: what have we learned? J Thromb Haemost 2024; 22:1541-1549. [PMID: 38428590 DOI: 10.1016/j.jtha.2024.02.013] [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: 12/11/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024]
Abstract
Coagulopathy alongside micro- and macrovascular thrombotic events were frequent characteristics of patients presenting with acute COVID-19 during the initial stages of the pandemic. However, over the past 4 years, the incidence and manifestations of COVID-19-associated coagulopathy have changed due to immunity from natural infection and vaccination and the appearance of new SARS-CoV-2 variants. Diagnostic criteria and management strategies based on early experience and studies for COVID-19-associated coagulopathy thus require reevaluation. As many other infectious disease states are also associated with hemostatic dysfunction, the coagulopathy associated with COVID-19 may be compounded, especially throughout the winter months, in patients with diverse etiologies of COVID-19 and other infections. This commentary examines what we have learned about COVID-19-associated coagulopathy throughout the pandemic and how we might best prepare to mitigate the hemostatic consequences of emerging infection agents.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Cheryl L Maier
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jean M Connors
- Hematology Division Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marcel Levi
- Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands; Department of Medicine, University College London Hospitals NHS Foundation Trust, Cardio-metabolic Programme-National Institute for Health and Care Research University College London Hospitals/University College London Biomedical Research Center, London, United Kingdom
| |
Collapse
|
122
|
Safdari H, Bagheri S, Talkhi N, Saberi Teymourian E, Hosseini Bafghi M, Ahmadi MH. Cq values as an indicator for COVID-19 outcomes: A study of the correlation between laboratory parameters. Immun Inflamm Dis 2024; 12:e1326. [PMID: 38923849 PMCID: PMC11194972 DOI: 10.1002/iid3.1326] [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: 02/13/2024] [Revised: 05/26/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVE The ongoing outbreak of the respiratory disease coronavirus disease 2019 (COVID-19) is currently presenting a major global health threat. This pandemic is unprecedented in recent human history. The objective of this study was to examine the relationship between cycle quantitation (Cq) and laboratory parameters in COVID-19 patients, aiming to determine if Cq levels can provide valuable insights into the COVID-19 disease. METHODS This study involved 234 participants who were divided into case and control groups. Real-time PCR tests were used to diagnose COVID-19 cases in the study participants. Blood tests, including complete blood count, C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), lactate dehydrogenase (LDH), D-dimer, IgG, and IgM, were also conducted. Statistical analysis was performed using SPSS 22 software. RESULTS The findings showed that COVID-19-positive cases had significantly higher levels of the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), D-dimer, ESR, CRP, and LDH compared to normal cases. Additionally, the case group had significantly lower lymphocyte and platelet counts. There was a statistically significant positive correlation between Cq levels and lymphocyte count (r = .124, p = .014). Conversely, there was a statistically significant inverse correlation between Cq levels and NLR (r = -.208, p = .017). Furthermore, the evaluation of hematological, inflammatory, and biochemical indexes in COVID-19 patients using the receiver-operating characteristics curve demonstrated statistically appropriate sensitivity and specificity. CONCLUSION Our outcomes indicated a significant association between Cq levels and PLR, NLR, D-dimer, CRP, and ESR in COVID-19 patients. Consequently, including the report of laboratory parameters alongside Cq values offers a promising prognosis.
Collapse
Affiliation(s)
- Hadi Safdari
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| | - Saeede Bagheri
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| | - Nasrin Talkhi
- Department of Biostatistics, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Elahe Saberi Teymourian
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| | - Mahdi Hosseini Bafghi
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| | - Mohammad Hossein Ahmadi
- Department of Laboratory Sciences, School of Paramedical Sciences, Faculty of Paramedical and Rehabilitation SciencesMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
123
|
Ertan-Bolelli T, Bolelli K, Elçi SD, Belen-Apak FB. Promising Drug Fondaparinux for the Treatment of COVID-19: an In Silico Analysis of Low Molecular Weight Heparin, Direct Oral Anticoagulant, and Antiplatelet Drug Interactions with Host Protease Furin. Cardiovasc Drugs Ther 2024; 38:425-432. [PMID: 36401727 PMCID: PMC9676724 DOI: 10.1007/s10557-022-07406-z] [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] [Accepted: 11/07/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE As of July 2022, the COVID-19 pandemic has affected over 555 million worldwide confirmed cases and caused more than 6.3 million deaths. The studies showed that the D-dimer levels were increased in non-survivors compared to survivors and heparin treatment has begun to be administered to the patients in severe clinics. As we knew that the entrance of SARS-CoV-2 to the host cell needs to be facilitated by host proteases; we published our hypothesis that heparin as a serine protease inhibitor may block the interaction between spike protein receptor-binding domain and host proteases. In our study, we aimed to investigate the interactions between not only heparins but also other antiplatelet and anticoagulant drugs including fondaparinux. METHODS In this study, docking studies were carried out to evaluate the interactions between low molecular weight heparins (LMWHs) (enoxaparin, dalteparin, tinzaparin), direct oral anticoagulant, and antiplatelet drugs with host proteases. Molecular docking studies were performed by using Schrödinger molecular modeling software. 3D structures of the ligands were obtained from the 2D structures by assigning the OPLS-2005 force field using the Maestro 12.7. The 3D crystal structure of the furin complexed with an inhibitor, 2,5-dideoksistreptamin derivative, was extracted from the Protein Data Bank (PDB ID: 5MIM). Docking studies were carried out using the Grid-based Ligand Docking with Energetics module of the Schrödinger Software. RESULTS The docking studies revealed that fondaparinux was the most relevant molecule to interact with furin with a docking score of - 12.74. It showed better interaction than the natural ligand of furin with an increased score compared to the docking score of - 8.155 of the natural ligand. AnaGA*IsA structure representing LMWH structure has shown a docking score of - 11.562 which was also better than the score of the natural ligand of furin. CONCLUSION Our findings have shown that LMWHs and fondaparinux can be used for their possible antiviral effects in COVID-19 patients. Our results have shown that in accordance with heparin and LMWH, fondaparinux can also be a candidate for "drug repurposing" in COVID-19 therapy, not only because of their anticoagulant but also possible antiviral effects.
Collapse
Affiliation(s)
- Tugba Ertan-Bolelli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey
- Bolelli Lab LLC, Stone Mountain, GA 30083 USA
| | - Kayhan Bolelli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, 06560 Ankara, Turkey
- Bolelli Lab LLC, Stone Mountain, GA 30083 USA
- LumiLabs, Ulus, Ankara, 06610 Turkey
| | | | - F. Burcu Belen-Apak
- Department of Pediatric Hematology and Oncology, Medical Faculty, Baskent University, Sehit Temel Kuguluoglu Street No 24, 06490 Bahcelievler/Ankara, Turkey
| |
Collapse
|
124
|
Gadelha LR, Costa MJB, Abreu JPAD, Venancio LPR, Fabres-Klein MH, Klein RC, Lima JB, Araújo-Santos T. Prostaglandin E 2/Leukotriene B 4 balance and viral load in distinct clinical stages of COVID-19: A cross-sectional study. Prostaglandins Other Lipid Mediat 2024; 172:106820. [PMID: 38346573 DOI: 10.1016/j.prostaglandins.2024.106820] [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: 08/17/2023] [Revised: 12/16/2023] [Accepted: 02/09/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Prostaglandin E2 (PGE2) and leukotriene B4 (LTB4) are eicosanoids involved in modulation of the antiviral immune response. Recent studies have identified increased levels of several eicosanoids in the plasma and bronchoalveolar lavage of patients with coronavirus disease (COVID-19). This study investigated correlations between plasma levels of PGE2 and LTB4 and clinical severity of COVID-19. METHODS This cross-sectional study involved non-infected (n = 10) individuals and COVID-19 patients classified as cured (n = 13), oligosymptomatic (n = 29), severe (n = 15) or deceased (n = 11). Levels of D-dimer a, known COVID-19 severity marker, PGE2 and LTB4 were measured by ELISAs and data were analysed with respect to viral load. RESULTS PGE2 plasma levels were decreased in COVID-19 patients compared to the non-infected group. Changes in PGE2 and LTB4 levels did not correlate with any particular clinical presentations of COVID-19. However, LTB4 was related to decreased SARS-CoV-2 burden in patients, suggesting that only LTB4 is associated with control of viral load. CONCLUSIONS Our data indicate that PGE2/LTB4 plasma levels are not associated with COVID-19 clinical severity. Hospitalized patients with COVID-19 are treated with corticosteroids, which may influence the observed eicosanoid imbalance. Additional analyses are required to fully understand the participation of PGE2 receptors in the pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Larisse Ricardo Gadelha
- Universidade Federal do Oeste da Bahia (UFOB), Núcleo de Estudos de Agentes Infecciosos e Vetores (NAIVE), Centro das Ciências Biológicas e da Saúde, Barreiras, BA, Brazil
| | - Maria Juliana Bezerra Costa
- Universidade Federal do Oeste da Bahia (UFOB), Núcleo de Estudos de Agentes Infecciosos e Vetores (NAIVE), Centro das Ciências Biológicas e da Saúde, Barreiras, BA, Brazil
| | - João Paulo Alecrim de Abreu
- Universidade Federal do Oeste da Bahia (UFOB), Núcleo de Estudos de Agentes Infecciosos e Vetores (NAIVE), Centro das Ciências Biológicas e da Saúde, Barreiras, BA, Brazil
| | - Larissa Paola Rodrigues Venancio
- Universidade Federal do Oeste da Bahia (UFOB), Núcleo de Estudos de Agentes Infecciosos e Vetores (NAIVE), Centro das Ciências Biológicas e da Saúde, Barreiras, BA, Brazil
| | - Mary Hellen Fabres-Klein
- Universidade Federal do Oeste da Bahia (UFOB), Núcleo de Estudos de Agentes Infecciosos e Vetores (NAIVE), Centro das Ciências Biológicas e da Saúde, Barreiras, BA, Brazil
| | - Raphael Contelli Klein
- Universidade Federal do Oeste da Bahia (UFOB), Núcleo de Estudos de Agentes Infecciosos e Vetores (NAIVE), Centro das Ciências Biológicas e da Saúde, Barreiras, BA, Brazil
| | - Jonilson Berlink Lima
- Universidade Federal do Oeste da Bahia (UFOB), Núcleo de Estudos de Agentes Infecciosos e Vetores (NAIVE), Centro das Ciências Biológicas e da Saúde, Barreiras, BA, Brazil
| | - Théo Araújo-Santos
- Universidade Federal do Oeste da Bahia (UFOB), Núcleo de Estudos de Agentes Infecciosos e Vetores (NAIVE), Centro das Ciências Biológicas e da Saúde, Barreiras, BA, Brazil.
| |
Collapse
|
125
|
El Malki H, Morjane Y, Belarbi A, Berkane MT, Moutaouekkil EM. [Une combinaison très rare de thrombose veineuse et artérielle chez un patient atteint du COVID 19]. Ann Cardiol Angeiol (Paris) 2024; 73:101744. [PMID: 38636246 DOI: 10.1016/j.ancard.2024.101744] [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: 02/21/2024] [Accepted: 03/10/2024] [Indexed: 04/20/2024]
Abstract
COVID-19 is an infectious disease caused by the SARS-CoV-2 virus. This epidemic has caused serious socio-economic problems worldwide, with a very high mortality rate. Several articles have been published in the literature showing the consequences of this disease on the human body. Among the most serious complications are venous and arterial thrombosis, which are rarely observed together in the same patient. In this article, we report an exceptional case of a patient with COVID-19 with the combination of intracardiac thrombus (left atrium) and venous thrombosis (splenic vein and portal trunk).
Collapse
Affiliation(s)
- Hicham El Malki
- Department of Cardiovascular surgery, Mohammed VI University Hospital Center, Oujda, Morocco; Faculty of Medicine and Pharmacy, Mohammed the First University, Oujda, Morocco
| | - Yassine Morjane
- Department of Cardiovascular surgery, Mohammed VI University Hospital Center, Oujda, Morocco.
| | - Adam Belarbi
- Department of Cardiovascular surgery, Mohammed VI University Hospital Center, Oujda, Morocco
| | - Mohammed Taha Berkane
- Department of Cardiovascular surgery, Mohammed VI University Hospital Center, Oujda, Morocco
| | - El Mehdi Moutaouekkil
- Department of Cardiovascular surgery, Mohammed VI University Hospital Center, Oujda, Morocco; Faculty of Medicine and Pharmacy, Mohammed the First University, Oujda, Morocco
| |
Collapse
|
126
|
Chen JY, Huang TR, Hsu SY, Huang CC, Wang HS, Chang JS. Effect and mechanism of quercetin or quercetin-containing formulas against COVID-19: From bench to bedside. Phytother Res 2024; 38:2597-2618. [PMID: 38479376 DOI: 10.1002/ptr.8175] [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: 08/13/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 06/13/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global coronavirus disease 2019 (COVID-19) pandemic since 2019. Immunopathogenesis and thromboembolic events are central to its pathogenesis. Quercetin exhibits several beneficial activities against COVID-19, including antiviral, anti-inflammatory, immunomodulatory, antioxidative, and antithrombotic effects. Although several reviews have been published, these reviews are incomplete from the viewpoint of translational medicine. The authors comprehensively evaluated the evidence of quercetin against COVID-19, both basically and clinically, to apply quercetin and/or its derivatives in the future. The authors searched the PubMed, Embase, and the Cochrane Library databases without any restrictions. The search terms included COVID-19, SARS-CoV-2, quercetin, antiviral, anti-inflammatory, immunomodulatory, thrombosis, embolism, oxidative, and microbiota. The references of relevant articles were also reviewed. All authors independently screened and reviewed the quality of each included manuscript. The Cochrane Risk of Bias Tool, version 2 (RoB 2) was used to assess the quality of the included randomized controlled trials (RCTs). All selected studies were discussed monthly. The effectiveness of quercetin against COVID-19 is not solid due to methodological flaws in the clinical trials. High-quality studies are also required for quercetin-containing traditional Chinese medicines. The low bioavailability and highly variable pharmacokinetics of quercetin hinder its clinical applications. Its positive impact on immunomodulation through reverting dysbiosis of gut microbiota still lacks robust evidence. Quercetin against COVID-19 does not have tough clinical evidence. Strategies to improve its bioavailability and/or to develop its effective derivatives are needed. Well-designed RCTs are also crucial to confirm their effectiveness in the future.
Collapse
Affiliation(s)
- Jhong Yuan Chen
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung Rung Huang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih Yun Hsu
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching Chun Huang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Huei Syun Wang
- Department of Traditional Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jung San Chang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- PhD Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
127
|
Driouich Z, Palmieri F, Hasan Gad Ali A, Younis S. Early presentation of Covid-19 related paracentral acute middle maculopathy in a healthy young patient. J Fr Ophtalmol 2024; 47:104121. [PMID: 38485629 DOI: 10.1016/j.jfo.2024.104121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 06/12/2024]
Affiliation(s)
- Z Driouich
- Western Eye Hospital, Imperial College NHS Healthcare Trust, London, UK.
| | - F Palmieri
- Western Eye Hospital, Imperial College NHS Healthcare Trust, London, UK
| | - A Hasan Gad Ali
- Western Eye Hospital, Imperial College NHS Healthcare Trust, London, UK
| | - S Younis
- Western Eye Hospital, Imperial College NHS Healthcare Trust, London, UK
| |
Collapse
|
128
|
Ebrahimi R, Nasri F, Kalantari T. Coagulation and Inflammation in COVID-19: Reciprocal Relationship between Inflammatory and Coagulation Markers. Ann Hematol 2024; 103:1819-1831. [PMID: 38349409 DOI: 10.1007/s00277-024-05630-1] [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: 09/17/2023] [Accepted: 01/16/2024] [Indexed: 05/14/2024]
Abstract
The coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), formerly known as 2019-nCoV. Numerous cellular and biochemical issues arise after COVID-19 infection. The severe inflammation that is caused by a number of cytokines appears to be one of the key hallmarks of COVID-19. Additionally, people with severe COVID-19 have coagulopathy and fulminant thrombotic events. We briefly reviewed the COVID-19 disease at the beginning of this paper. The inflammation and coagulation markers and their alterations in COVID-19 illness are briefly discussed in the parts that follow. Next, we talked about NETosis, which is a crucial relationship between coagulation and inflammation. In the end, we mentioned the two-way relationship between inflammation and coagulation, as well as the factors involved in it. We suggest that inflammation and coagulation are integrated systems in COVID-19 that act on each other in such a way that not only inflammation can activate coagulation but also coagulation can activate inflammation.
Collapse
Affiliation(s)
- Rasoul Ebrahimi
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Nasri
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Kalantari
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
129
|
Zaborska M, Chruszcz M, Sadowski J, Klaudel T, Pelczarski M, Sztangreciak-Lehun A, Bułdak RJ. The most common skin symptoms in young adults and adults related to SARS-CoV-2 virus infection. Arch Dermatol Res 2024; 316:292. [PMID: 38819524 PMCID: PMC11142985 DOI: 10.1007/s00403-024-02991-5] [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: 04/11/2024] [Revised: 04/12/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024]
Abstract
Scientists from various areas of the world indicate in their studies that skin lesions occur in the course of infection with the SARS-CoV-2 virus. This article is a review of the most frequently described cutaneous manifestations of SARS-CoV-2 virus infection and the potential pathophysiology of their development, as well as information on abnormalities in histopathological tests. The article describes the impact of some factors related to the COVID-19 pandemic on the exacerbation of chronic dermatological diseases. This work was constructed on the basis of 142 research studies, reviews, and meta-analyses, focusing on the methods and materials used in individual works as well as the results and conclusions resulting from them. Some skin lesions may be a potential prognostic marker of the course of the disease and may also be a prodromal symptom or the only symptom of SARS-CoV-2 virus infection. Stress related to the COVID-19 pandemic may exacerbate some chronic dermatological diseases. A correlation was observed between the type of skin lesions and the patient's age. The occurrence of skin diseases may also be influenced by drugs used to treat infections caused by SARS-CoV-2. A relationship was observed between the patient's ethnic origin and skin lesions occurring in the course of COVID-19. There is a need to further diagnose the cutaneous manifestations of SARS-CoV-2 infection and to learn the detailed pathomechanism of their occurrence in order to better understand the essence of the disease and find an appropriate treatment method.
Collapse
Affiliation(s)
- Monika Zaborska
- Student Scientific Society of Clinical Biochemistry and Regenerative Medicine, Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland.
| | - Maksymilan Chruszcz
- Student Scientific Society of Clinical Biochemistry and Regenerative Medicine, Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland
| | - Jakub Sadowski
- Student Scientific Society of Clinical Biochemistry and Regenerative Medicine, Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland
| | - Tomasz Klaudel
- Student Scientific Society of Clinical Biochemistry and Regenerative Medicine, Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland
| | - Michał Pelczarski
- Student Scientific Society of Clinical Biochemistry and Regenerative Medicine, Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland
| | - Anna Sztangreciak-Lehun
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland
| | - Rafał Jakub Bułdak
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052, Opole, Poland
| |
Collapse
|
130
|
Yang Y, Zheng Q, Yang L, Wu L. Comparison of inflammatory markers, coagulation indicators and outcomes between influenza and COVID-19 infection amongst children: A systematic review and meta-analysis. Heliyon 2024; 10:e30391. [PMID: 38765052 PMCID: PMC11096948 DOI: 10.1016/j.heliyon.2024.e30391] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/21/2024] Open
Abstract
Background Influenza and COVID-19 patients share similar features and outcomes amongst adults. However, the difference between these diseases is not explored in paediatric age group especially in terms of inflammatory markers, coagulation profile and outcomes. Hence, we did this review to compare the inflammatory, coagulation features and outcomes between influenza and COVID-19 infected children. Methods Literature search was done in PubMed Central, Scopus, EMBASE, CINAHL, Cochrane library, Google Scholar & ScienceDirect from November 2019 to May 2022. Risk of bias assessment was done through Newcastle Ottawa scale. Meta-analysis was done using random-effects model and the final pooled estimate was reported as pooled odds ratio (OR) or standardized mean difference (SMD) along with 95 % confidence interval (CI) depending on the type of outcome. Results About 16 studies were included with most studies having higher risk of bias. Influenza paediatric patients had significantly higher erythrocyte sedimentation rate (ESR) (pooled SMD = 0.60; 95%CI: 0.30-0.91; I2 = 0 %), lactate dehydrogenase (LDH) (pooled SMD = 2.01; 95%CI: 0.37-3.66; I2 = 98.4 %) and prothrombin time (PT) (pooled SMD = 2.12; 95%CI: 0.44-3.80; I2 = 98.3 %) when compared to paediatric COVID-19 patients. There was no significant difference in terms of features like CRP, procalcitonin, serum albumin, aPTT, mortality and need for mechanical ventilation. Conclusion Inflammatory markers like ESR, LDH and PT was significantly higher in influenza patients when compared to COVID-19 in children, while rest of the markers and adverse clinical outcomes were similar between both the groups. Identification of these biomarkers has helped in understanding the distinctness of COVID-19 and influenza virus and develop better management strategies.
Collapse
Affiliation(s)
- Yutang Yang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250021, China
| | - Qi Zheng
- Department of Gynecology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250013, China
| | - Linlin Yang
- Department of Hematology and Rheumatology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250013, China
| | - Lei Wu
- Department of Pediatrics, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, 250013, China
| |
Collapse
|
131
|
Gando S, Akiyama T. Disseminated intravascular coagulation is associated with poor prognosis in patients with COVID-19. Sci Rep 2024; 14:12443. [PMID: 38816405 PMCID: PMC11139854 DOI: 10.1038/s41598-024-63078-9] [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: 02/04/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024] Open
Abstract
This study aimed to investigate the incidence and significance of disseminated intravascular coagulation (DIC) in coronavirus disease 2019 (COVID-19). A multicenter cohort study was conducted using large-scale COVID-19 registry data. The patients were classified into DIC and non-DIC groups based on the diagnosis on admission (day 1) and on any of the days 1, 4, 8, and 15. In total, 23,054 patients were divided into DIC (n = 264) and non-DIC (n = 22,790) groups on admission. Thereafter, 1654 patients were divided into 181 patients with DIC and 1473 non-DIC patients based on the DIC diagnosis on any of the days from 1 to 15. DIC incidence was 1.1% on admission, increasing to 10.9% by day 15. DIC diagnosis on admission had moderate predictive performance for developing multiple organ dysfunction syndrome (MODS) on day 4 and in-hospital death and was independently associated with MODS and in-hospital death. DIC diagnosis on any of the days from 1 to 15, especially days 8 and 15, was associated with lower survival probability than those without DIC and showed significant association with in-hospital death. In conclusion, despite its low incidence, DIC, particularly late-onset DIC, plays a significant role in the pathogenesis of poor prognosis in patients with COVID-19.
Collapse
Affiliation(s)
- Satoshi Gando
- Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, N34, E14, Higashi-ku, Sapporo, 065-0033, Japan.
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan.
| | - Takayuki Akiyama
- Large-Scale Data Archiving and Processing Section, Institute of Economic Research, Hitotsubashi University, Tokyo, Japan
- AMR Clinical Reference Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
132
|
Del Carmen Camacho-Rea M, Martínez-Gómez LE, Martinez-Armenta C, Martínez-Nava GA, Ortega-Peña S, Olea-Torres J, Herrera-López B, Suarez-Ahedo C, Vázquez-Cárdenas P, Vidal-Vázquez RP, Ramírez-Hinojosa JP, Vargas-Alarcón G, Posadas-Sánchez R, Fragoso JM, De Jesús Martínez-Ruiz F, Zayago-Angeles DM, Mata-Miranda MM, Vazquez-Zapien GJ, Martínez-Cuazitl A, Garcia-Galicia A, Granados J, Ramos L, Rodríguez-Pérez JM, Pineda C, López-Reyes A. Association of TLR8 Variants in Sex-Based Clinical Differences in Patients with COVID-19. Biochem Genet 2024:10.1007/s10528-024-10839-w. [PMID: 38814383 DOI: 10.1007/s10528-024-10839-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 05/13/2024] [Indexed: 05/31/2024]
Abstract
The host immune response might confer differential vulnerability to SARS-CoV-2 infection. The Toll-like receptor 8 (TLR8), could participated for severe COVID-19 outcomes. To investigated the relationship of TLR8 rs3764879-C/G, rs3764880-A/G, and rs3761624-A/G with COVID-19 outcomes and with biochemical parameters. A cross-sectional study of 830 laboratory-confirmed COVID-19 patients was performed, and classified into mild, severe, critical, and deceased outcomes. The TLR8 rs3764879-C/G, rs3764880-A/G, and rs3761624-A/G polymorphisms were genotyped. A logistic regression analysis was performed to determinate the association with COVID-19. A stratified analysis was by alleles was done with clinical and metabolic markets. In all outcomes, men presented the highest ferritin levels compared to women (P < 0.001). LDH levels were significantly different between sex in mild (P = 0.003), severe (P < 0.001) and deceased (P = 0.01) COVID-19 outcomes. The GGG haplotype showed an Odds Ratio of 1.55 (Interval Confidence 95% 1.05-2.32; P = 0.03) in men. Among patients with severe outcome, we observed that the carriers of the GGG haplotype had lower Ferritin, C-reactive protein and LDH levels than the CAA carriers (P < 0.01). After further stratified by sex, these associations were also seen in the male patients, except for D-dimer. Interestingly, among men patients, we could observe associations between TLR8 haplotypes and Ferritin (P < 0.001), D-dimer (P = 0.04), C-reactive protein, and Lactate dehydrogenase in mild (P = 0.04) group. Our results suggest that even though TLR8 haplotypes show a significant association with COVID-19 outcomes, they are associated with clinical markers in COVID-19 severity.
Collapse
Affiliation(s)
- María Del Carmen Camacho-Rea
- Departamento de Nutrición Animal, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México
| | - Laura Edith Martínez-Gómez
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México
| | - Carlos Martinez-Armenta
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México
| | - Gabriela Angélica Martínez-Nava
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México
| | - Silvestre Ortega-Peña
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México
| | - Jessel Olea-Torres
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México
| | - Brígida Herrera-López
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México
| | - Carlos Suarez-Ahedo
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México
| | - Paola Vázquez-Cárdenas
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología E Infectología, Hospital General Dr. Manuel Gea González, CDMX, México
| | - Rosa P Vidal-Vázquez
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología E Infectología, Hospital General Dr. Manuel Gea González, CDMX, México
| | - Juan Pablo Ramírez-Hinojosa
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología E Infectología, Hospital General Dr. Manuel Gea González, CDMX, México
| | - Gilberto Vargas-Alarcón
- Centro de Innovación Médica Aplicada, Subdirección de Epidemiología E Infectología, Hospital General Dr. Manuel Gea González, CDMX, México
| | | | - José Manuel Fragoso
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, CDMX, México
| | - Felipe De Jesús Martínez-Ruiz
- Nuevo Hospital General Delegación Regional Sur de La Ciudad de México, Instituto de Seguridad y Servicios Sociales Para los Trabajadores del Estado (ISSSTE), CDMX, México
| | - Dulce María Zayago-Angeles
- Nuevo Hospital General Delegación Regional Sur de La Ciudad de México, Instituto de Seguridad y Servicios Sociales Para los Trabajadores del Estado (ISSSTE), CDMX, México
| | - Mónica Maribel Mata-Miranda
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Militar de Medicina, Universidad del Ejército y Fuerza Aérea, CDMX, México
| | - Gustavo Jesús Vazquez-Zapien
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Militar de Medicina, Universidad del Ejército y Fuerza Aérea, CDMX, México
| | - Adriana Martínez-Cuazitl
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Militar de Medicina, Universidad del Ejército y Fuerza Aérea, CDMX, México
| | - Armando Garcia-Galicia
- Servicio de Cirugía General, Hospital Central Norte Petróleos Mexicanos (PEMEX), CDMX, México
| | - Julio Granados
- Departamento de Trasplantes, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México
| | - Luis Ramos
- Departamento de Trasplantes, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México
| | | | - Carlos Pineda
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México
| | - Alberto López-Reyes
- Unidad de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Calz México-Xochimilco 289, Coapa, Col. Arenal de Guadalupe, Tlalpan, 14389, CDMX, México.
| |
Collapse
|
133
|
Tucker J, Eberly HW, Lighthall JG. Impact of a History of COVID-19 Infection on Postoperative Outcomes for Free Flap Patients. EAR, NOSE & THROAT JOURNAL 2024:1455613241255995. [PMID: 38804526 DOI: 10.1177/01455613241255995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024] Open
Abstract
Objectives: This study examines the impact of a history of coronavirus disease 2019 (COVID-19) infection on patients' outcomes after microvascular free flap surgery and to examine the recommendations on when to perform microvascular surgery after a COVID-19 infection. Methods: A retrospective chart review using the TriNetX database was completed on March 5, 2023. Two cohorts were created: (1) patients who had a positive COVID-19 diagnosis within 1 year before microvascular free flap surgery, and (2) patients with no history of COVID-19 who underwent free flap surgery. Current Procedural Terminology codes were used to identify procedures and International Classification of Diseases-10 codes were used to identify outcomes. Results: There was a total of 31,505 patients who underwent microvascular free flap surgery, 500 of whom had a diagnosis of COVID-19 within 1 year of free flap surgery and 31,005 without history of COVID-19. There was increased risk of sepsis, surgical site infection (SSI), generalized infection, gangrene, dehiscence, hematoma, seroma, intensive care unit admission, and death in patients who underwent free flap surgery within a year of COVID-19 infection. After propensity score matching, there were 498 patients in both groups. Increased risk remained for SSI and gangrene in patients with a history of COVID-19 after matching. When comparing surgical timing between 0 to 2 months after COVID-19 infection and 2 to 12 months after COVID-19 infection, there were no significant differences between groups. Conclusions: After propensity score matching, patients with a history of COVID-19 infection were at increased risk for SSI and gangrene. However, many flap surgeries cannot be delayed. This study may help counsel patients regarding the possible complications after surgery and provide a heightened awareness in the surgical team of a possible increase in infectious complications in this population. Additional studies should investigate optimal timing of free flap surgery after COVID-19 infection and ways to mitigate the risk of infectious complications.
Collapse
Affiliation(s)
| | - Hänel W Eberly
- Department of Otolaryngology-Head and Neck Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Jessyka G Lighthall
- College of Medicine, Surgery, The Pennsylvania State University, Hershey, PA, USA
| |
Collapse
|
134
|
Gavronova A, Hamerlik L, Bartkova M, Svrchokryl V, Kralikova V, Vranova K, Ondra P, Dobias M. Fat embolism and COVID-19 infection: autopsy and post-mortem laboratory findings in SARS-CoV-2 positive patients. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2024. [PMID: 38818791 DOI: 10.5507/bp.2024.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024] Open
Abstract
INTRODUCTION The article is one of the very first autopsy reports worldwide, which associates COVID-19 infection and pulmonary fat embolism. AIMS To point to a crucial connection between a severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2) infection and pulmonary fat embolism as one of the possible major mechanisms of severe COVID-19 symptoms. METHODS Lung, brain and kidney tissues examination of 16 full human autopsy cases. All deceased suffered from COVID-19 infection, none of them was admitted to hospital prior to death, immediate causes of death vary. Autopsies accompanied by microbiological examination and histological examination using Oil Red O staining were performed. Consequently, we have implemented a control cohort consisting of 16 deceased with no presence of pulmonary infection and various immediate causes of death. RESULTS Of the 16 autopsy cases, 11 (68.8%) were males and 5 (31.3%) females, with overall mean age 68.1 (39-86) years. Causes of death of studied subjects were natural, mostly from respiratory failure (in 12 cases, 75%). Cardiopulmonary resuscitation was performed in 7 cases (43.8%). None of dissected persons had larger signs of body trauma. Pulmonary fat embolism was found in 11 cases (68.8%), which generalised to kidneys in 8 patients (50% of all cases, 72.3% of cases with pulmonary fat embolism) and to brain tissue in 1 case. CONCLUSION We demonstrated a reasonable relation between a COVID-19 disease and a variously severe fat embolism, severity of which does not directly correlate with body weight. Further investigation or even change of medical treatment needs to be considered in patients with COVID-19.
Collapse
Affiliation(s)
- Adriana Gavronova
- Department of Forensic Medicine and Medical Law, University Hospital Olomouc, Czech Republic
- Department of Forensic Medicine and Medical Law, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Lukas Hamerlik
- Department of Forensic Medicine and Medical Law, University Hospital Olomouc, Czech Republic
- Department of Forensic Medicine and Medical Law, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Margita Bartkova
- Clinical laboratory, Psychiatric Hospital Sternberk, Czech Republic
| | - Vaclav Svrchokryl
- Department of Forensic Medicine and Medical Law, University Hospital Olomouc, Czech Republic
- Department of Forensic Medicine and Medical Law, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Veronika Kralikova
- Department of Forensic Medicine and Medical Law, University Hospital Olomouc, Czech Republic
- Department of Forensic Medicine and Medical Law, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Katerina Vranova
- Department of Forensic Medicine and Medical Law, University Hospital Olomouc, Czech Republic
- Department of Forensic Medicine and Medical Law, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Peter Ondra
- Department of Forensic Medicine and Medical Law, University Hospital Olomouc, Czech Republic
- Department of Forensic Medicine and Medical Law, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Martin Dobias
- Department of Forensic Medicine and Medical Law, University Hospital Olomouc, Czech Republic
- Department of Forensic Medicine and Medical Law, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| |
Collapse
|
135
|
Hogea B, Suba MI, Abu-Awwad SA, Cuntan P, Popa MV, Braescu RD, Abu-Awwad A. Exploring the Association between COVID-19 and Femoral Head Necrosis: A Comprehensive Review. Life (Basel) 2024; 14:671. [PMID: 38929655 PMCID: PMC11204444 DOI: 10.3390/life14060671] [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: 04/20/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
This study investigates the correlation between COVID-19 and avascular necrosis of the femoral head, considering the potential contribution of medication-induced effects. This research spans the period from August 2022 to January 2024 and includes 32 patients diagnosed with avascular necrosis. While steroid usage, particularly in high doses, is known to predispose individuals to this condition, this study aims to discern if COVID-19 itself plays a role beyond the influence of medication. Notably, COVID-19 is associated with disturbances in the coagulation system, potentially leading to thromboembolic complications. Of the patients, six did not have COVID-19, while seven had the virus but did not receive steroid treatment. However, 19 patients with COVID-19 exhibited severe pulmonary involvement and were administered both high-dose steroids and antiviral medication. Among the observed patients, 14 were female and 18 were male. Notably, three patients presented bilateral necrosis, all of whom had COVID-19 and significant pulmonary involvement. Diagnostic assessments included frontal and profile X-rays, as well as MRI scans for all patients.
Collapse
Affiliation(s)
- Bogdan Hogea
- Department XV—Discipline of Orthopedics—Traumatology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (B.H.); (A.A.-A.)
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld Liviu Rebreanu, No. 156, 300723 Timisoara, Romania; (S.-A.A.-A.); (P.C.); (M.-V.P.); (R.D.B.)
- Research Center University Professor Doctor Teodor Șora, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Madalina-Ianca Suba
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- Dr. Victor Babeș Infectious Diseases and Pneumophthisiology Hospital Timisoara, 300310 Timisoara, Romania
| | - Simona-Alina Abu-Awwad
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld Liviu Rebreanu, No. 156, 300723 Timisoara, Romania; (S.-A.A.-A.); (P.C.); (M.-V.P.); (R.D.B.)
- Department XII—Discipline of Obstetrics and Gynecology, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Paul Cuntan
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld Liviu Rebreanu, No. 156, 300723 Timisoara, Romania; (S.-A.A.-A.); (P.C.); (M.-V.P.); (R.D.B.)
| | - Mihai-Valetin Popa
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld Liviu Rebreanu, No. 156, 300723 Timisoara, Romania; (S.-A.A.-A.); (P.C.); (M.-V.P.); (R.D.B.)
| | - Ruben David Braescu
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld Liviu Rebreanu, No. 156, 300723 Timisoara, Romania; (S.-A.A.-A.); (P.C.); (M.-V.P.); (R.D.B.)
| | - Ahmed Abu-Awwad
- Department XV—Discipline of Orthopedics—Traumatology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania; (B.H.); (A.A.-A.)
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld Liviu Rebreanu, No. 156, 300723 Timisoara, Romania; (S.-A.A.-A.); (P.C.); (M.-V.P.); (R.D.B.)
- Research Center University Professor Doctor Teodor Șora, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
| |
Collapse
|
136
|
Sofi F, Dinu M, Reboldi G, Lotti S, Genovese L, Tritto I, Gensini G, Gibson CM, Ambrosio G. Worldwide impact of COVID-19 on hospital admissions for non-ST-elevation acute coronary syndromes (NSTACS): a systematic review with meta-analysis of 553 038 cases. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2024; 10:265-283. [PMID: 37580157 PMCID: PMC11112522 DOI: 10.1093/ehjqcco/qcad048] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND How coronavirus disease 2019 (COVID-19) impacted non-ST-segment elevation acute coronary syndromes (NSTACS) is an object of controversial reports. AIM To systematically review studies reporting NSTACS hospitalizations during the COVID-19 pandemic, and analyse whether differences in COVID-19 epidemiology, methodology of report, or public health-related factors could contribute to discrepant findings. METHODS Comprehensive search (Medline, Embase, Scopus, Web of Science, Cochrane Register), of studies reporting NSTACS hospitalizations during the COVID-19 pandemic compared with a reference period, following Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines. Data were independently extracted by multiple investigators and pooled using a random-effects model. Health-related metrics were from publicly available sources, and analysed through multiple meta-regression modelling. RESULTS We retrieved 102 articles (553 038 NSTACS cases, 40 countries). During peak COVID-19 pandemic, overall incidence rate ratio (IRR) of NSTACS hospitalizations over reference period decreased (0.70, 95% confidence interval (CI) 0.66-0.75; P < 0.00001). Significant heterogeneity was detected among studies (I2 = 98%; P < 0.00001). Importantly, wide variations were observed among, and within, countries. No significant differences were observed by study quality, whereas comparing different periods within 2020 resulted in greater decrease (IRR: 0.61; CI: 0.53-0.71) than comparing 2020 vs. previous years (IRR: 0.74; CI 0.69-0.79). Among many variables, major predictors of heterogeneity were severe acute respiratory syndrome coronavirus 2 reproduction rate/country, number of hospitals queried, and reference period length; country stringency index and socio-economical indicators did not contribute significantly. CONCLUSIONS During the COVID-19 pandemic, NSTACS hospitalizations decreased significantly worldwide. However, substantial heterogeneity emerged among countries, and within the same country. Factors linked to public health management, but also to methodologies to collect results may have contributed to this heterogeneity. TRIAL REGISTRATION The protocol was registered in the PROSPERO International Prospective Register of Systematic Reviews (ID: CRD42022308159).
Collapse
Affiliation(s)
- Francesco Sofi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Monica Dinu
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - GianPaolo Reboldi
- Department of Medicine, University of Perugia School of Medicine, Perugia 06126, Italy
- Center for Clinical and Translational Research—CERICLET, University of Perugia School of Medicine, Perugia 06126, Italy
| | - Sofia Lotti
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Luca Genovese
- Department of Cardiology, IRCCS MultiMedica, Milan 20138, Italy
| | - Isabella Tritto
- Division of Cardiology, University of Perugia School of Medicine, Perugia 06126, Italy
| | | | | | - Giuseppe Ambrosio
- Center for Clinical and Translational Research—CERICLET, University of Perugia School of Medicine, Perugia 06126, Italy
- Division of Cardiology, University of Perugia School of Medicine, Perugia 06126, Italy
| |
Collapse
|
137
|
Huang S, Perry A, Sanchez Parra C, Gonzalez Torriente A, Ghumman H, Charkowick S, Colon J, Heide M, Jaglal M, Mhaskar R, Rico JF. Incidence of Thrombosis in COVID-19 Patients Compared to Non-COVID-19 Sepsis Patients in the Intensive Care Unit. J Clin Med 2024; 13:2974. [PMID: 38792515 PMCID: PMC11121895 DOI: 10.3390/jcm13102974] [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: 11/12/2023] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Background/Objectives: The hypercoagulable state associated with COVID-19 infection is associated with adverse outcomes and mortality. Studies have also demonstrated high rates of venous thromboembolism (VTE) events among patients with sepsis. We aimed to evaluate how the increase in thrombotic events in critically ill patients with COVID-19 infection compares to that of critically ill patients with non-COVID-19 sepsis. Methods: A chart review was performed of patients 18 years or older admitted to the intensive care unit (ICU) at Tampa General Hospital between 1 January 2020 and 31 December 2020 diagnosed with COVID-19 or sepsis secondary to other pathogens. Non-COVID-19 sepsis patients and COVID-19 patients were propensity-matched 3:1 on the Charlson Comorbidity Index. Multivariate analyses adjusting for confounding were conducted to report odds ratio (OR) and 95% confidence intervals (95% CIs) of predictors for thrombotic events and overall mortality. Results: After propensity score matching, 492 sepsis patients and 164 COVID-19 patients were included in the analysis. COVID-19 patients were significantly older (p = 0.021) and showed higher BMI (p < 0.001) than sepsis patients. COVID-19 patients did not show significantly higher odds of thrombosis after adjustment for confounders (OR 0.85, 95% CI 0.42-1.72), but had significantly lower odds of mortality than sepsis patients (OR 0.33, 95% CI 0.16-0.66). Conclusions: Our results suggest that further study is required to lower the rate of VTE in COVID-19 and non-COVID-19 sepsis patients admitted to the ICU; it is also reasonable to consider similar thromboembolism practices between these two patient groups.
Collapse
Affiliation(s)
- Sherri Huang
- Department of Internal Medicine and Pediatrics, University of South Florida, Tampa, FL 33606, USA
| | - Ashley Perry
- Department of Internal Medicine and Pediatrics, University of South Florida, Tampa, FL 33606, USA
| | - Carlos Sanchez Parra
- Department of Internal Medicine and Pediatrics, University of South Florida, Tampa, FL 33606, USA
- Division of Pediatric Cardiology, Texas Children’s Hospital, Houston, TX 77030, USA
| | | | - Haider Ghumman
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Shaun Charkowick
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Joshua Colon
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - McKenzi Heide
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Michael Jaglal
- Department of Hematology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Hematology and Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Rahul Mhaskar
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Juan Felipe Rico
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL 33606, USA
| |
Collapse
|
138
|
Zhu Y, Cao X, Ying R, Liu K, Chai Y, Luo M, Huang Q, Gao P, Zhang C. Mapping the vast landscape of multisystem complications of COVID-19: Bibliometric analysis. Heliyon 2024; 10:e30760. [PMID: 38765136 PMCID: PMC11098853 DOI: 10.1016/j.heliyon.2024.e30760] [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: 11/01/2023] [Revised: 04/16/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Background With the rapid global spread of COVID-19, it has become evident that the virus can lead to multisystem complications, leading to a significant increase in related publications. Bibliometrics serves as a valuable tool for identifying highly cited literature and research hotspots within specific areas. Objective The aim of this study is to identify current research hotspots and future trends in COVID-19 complications. Methods The dataset was obtained from the Web of Science Core Collection, covering COVID-19 complications from December 8, 2019, to October 31, 2022. Various aspects, including publication general information, authors, journals, co-cited authors, co-cited references, research hotspots, and future trends, were subjected to analysis. Visual analysis was conducted using VOSviewer, The Online Analysis Platform of Literature Metrology, and Charticulator. Results There were 4597 articles in the study. The top three countries with the most published articles are the USA (n = 1350, 29.4 %), China (n = 765, 16.6 %), and Italy (n = 623, 13.6 %). USA and China have the closest collaborative relationship. The institute with the largest number of publications is Huazhong University of Science and Technology, followed by Harvard Medical School. Nevertheless, half of the top 10 institutes belong to the USA. "Rezaei, Nima" published 13 articles and ranked first, followed by "Yaghi, Shadi" with 12 articles and "Frontera, Jennifer" with 12 articles. The journal with the largest number of publications is "Journal of Clinical Medicine". The top 3 co-cited authors are "Zhou, Fei", "Guan, Wei-Jie", "Huang, Chaolin". The top 3 co-cited references addressed COVID-19's clinical features in China and noticed that COVID-19 patients had a wide range of complications. We also list four research hotspots. Conclusions This study conducted a bibliometric visual analysis of the literature on COVID-19 complications and summarized the current research hotspots. This study may provide valuable insights into the complications of COVID-19.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyu Cao
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rongtao Ying
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Liu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yilu Chai
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maocai Luo
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingsong Huang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peiyang Gao
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
139
|
Özer Aslan İ, Doğan K, Kural A, Baghaki S, Helvaci N, Ekin M, Yaşar L. Is it possible to identify COVID-19 infection-related biomarkers during pregnancy?: A prospective study in Turkish population. Medicine (Baltimore) 2024; 103:e38062. [PMID: 38728492 PMCID: PMC11081580 DOI: 10.1097/md.0000000000038062] [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: 12/31/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) has raised concerns about the potential complications it may cause in pregnant women. Therefore, biomarkers that can predict the course of COVID-19 in pregnant women may be of great benefit as they would provide valuable insights into the prognosis and, thus, the management of the disease. In this context, the objective of this study is to identify the biomarkers that can predict COVID-19 progression in pregnant women, focusing on composite hemogram parameters and systemic inflammatory and spike markers. The population of this single-center prospective case-control study consisted of all consecutive pregnant women with single healthy fetuses who tested positive for COVID-19 and who were admitted to Bakirköy Dr Sadi Konuk Training and Research Hospital in Istanbul, Turkey, a COVID-19 referral hospital, between April 2020 and March 2021, with an obstetric indication, during their second or third trimester. The control group consisted of consecutive pregnant women with a single healthy fetus who were admitted to the same hospital within the same date range, had demographic and obstetric characteristics matching the patient group, but tested negative for COVID-19. The patient and control groups were compared in terms of platelet-to-lymphocyte ratio (PLR), platelet-to-neutrophil ratio (PNR), and neutrophil-to-lymphocyte ratio (NLR), and systemic inflammatory and spike markers, including C-reactive protein (CRP), interleukin-6 (IL-6), interleukin-10 (IL-10), cluster of differentiation 26 (CD26), and B7 homolog 4 (B7H4). There were 45 (51.1%) and 43 (48.8%) pregnant women in the patient and control groups, respectively. There was no significant difference between the groups in demographic and obstetric characteristics (P > .05). The PNR, PLR, and CRP values were significantly higher in the patient group than in the control group (P < .05). On the other hand, there was no significant difference between the groups in IL-6, IL-10, CD26, and B7H4 levels (P > .05). The findings of our study showed that specific inflammatory markers, such as CRP, PLR, and PNR, can potentially predict the course of COVID-19 in pregnant women. However, more comprehensive, well-controlled studies are needed to corroborate our study's findings and investigate other potential inflammatory markers.
Collapse
Affiliation(s)
- İlke Özer Aslan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Tekirdag Namik Kemal University, Tekirdag, Türkiye
| | - Keziban Doğan
- Department of Obstetrics and Gynecology, University of Health Sciences, Bakirkoy Dr.Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Alev Kural
- Department of Biochemistry, University of Health Sciences, Bakirkoy Dr.Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Sema Baghaki
- Department of Obstetrics and Gynecology, University of Pittsburgh, PA
| | - Nazli Helvaci
- Department of Biochemistry, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Türkiye
| | - Murat Ekin
- Department of Obstetrics and Gynecology, University of Health Sciences, Bakirkoy Dr.Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Levent Yaşar
- Department of Obstetrics and Gynecology, University of Health Sciences, Bakirkoy Dr.Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| |
Collapse
|
140
|
Zhong L, Lin Q, He L, Liu D, Zhu L, Zeng Q, Song J. Time to maximum amplitude of thromboelastography can predict mortality in patients with severe COVID-19: a retrospective observational study. Front Med (Lausanne) 2024; 11:1356283. [PMID: 38756947 PMCID: PMC11097111 DOI: 10.3389/fmed.2024.1356283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Objective To predict mortality in severe patients with COVID-19 at admission to the intensive care unit (ICU) using thromboelastography (TEG). Methods This retrospective, two-center, observational study involved 87 patients with PCR-and chest CT-confirmed severe COVID-19 who were admitted to at Wuhan Huoshenshan Hospital and the 908th Hospital of Chinese PLA Logistic Support Force between February 2020 and February 2023. Clinic demographics, laboratory results, and outcomes were compared between those who survived and those who died during hospitalization. Results Thromboelastography showed that of the 87 patients, 14 were in a hypercoagulable state, 25 were in a hypocoagulable state, and 48 were normal, based on the time to maximum amplitude (TMA). Patients who died showed significantly lower α angle, but significantly longer R-time, K-time and TMA than patients who survived. Random forest selection showed that K-time, TMA, prothrombin time (PT), international normalized ratio (INR), D-dimer, C-reactive protein (CRP), aspartate aminotransferase (AST), and total bilirubin (Tbil) were significant predictors. Multivariate logistic regression identified that TMA and CRP were independently associated with mortality. TMA had a greater predictive power than CRP levels based on time-dependent AUCs. Patients with TMA ≥ 26.4 min were at significantly higher risk of mortality (hazard ratio 3.99, 95% Confidence Interval, 1.92-8.27, p < 0.01). Conclusion TMA ≥26.4 min at admission to ICU may be an independent predictor of in-hospital mortality for patients with severe COVID-19.
Collapse
Affiliation(s)
- Lincui Zhong
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Qingwei Lin
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Longping He
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Dongmei Liu
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
- Intensive Care Unit, The 940th Hospital of Chinese PLA Logistic Support Force, Lanzhou, Gansu, China
| | - Lin Zhu
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
- Department of Critical Care Medicine, The 944th Hospital of Chinese PLA Logistic Support Force, Jiuquan, Gansu, China
| | - Qingbo Zeng
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
| | - Jingchun Song
- Intensive Care Unit, The 908th Hospital of Chinese PLA Logistic Support Force, Nanchang, Jiangxi, China
- Intensive Care Unit, Huoshenshan Hospital, Wuhan, Hubei, China
| |
Collapse
|
141
|
Bijla M, Saini SK, Pathak AK, Bharadwaj KP, Sukhavasi K, Patil A, Saini D, Yadav R, Singh S, Leeuwenburgh C, Kumar P. Microbiome interactions with different risk factors in development of myocardial infarction. Exp Gerontol 2024; 189:112409. [PMID: 38522483 DOI: 10.1016/j.exger.2024.112409] [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: 02/16/2024] [Revised: 03/10/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Among all non-communicable diseases, Cardiovascular Diseases (CVDs) stand as the leading global cause of mortality. Within this spectrum, Myocardial Infarction (MI) strikingly accounts for over 15 % of all deaths. The intricate web of risk factors for MI, comprising family history, tobacco use, oral health, hypertension, nutritional pattern, and microbial infections, is firmly influenced by the human gut and oral microbiota, their diversity, richness, and dysbiosis, along with their respective metabolites. Host genetic factors, especially allelic variations in signaling and inflammatory markers, greatly affect the progression or severity of the disease. Despite the established significance of the human microbiome-nutrient-metabolite interplay in associations with CVDs, the unexplored terrain of the gut-heart-oral axis has risen as a critical knowledge gap. Moreover, the pivotal role of the microbiome and the complex interplay with host genetics, compounded by age-related changes, emerges as an area of vital importance in the development of MI. In addition, a distinctive disease susceptibility and severity influenced by gender-based or ancestral differences, adds a crucial insights to the association with increased mortality. Here, we aimed to provide an overview on interactions of microbiome (oral and gut) with major risk factors (tobacco use, alcohol consumption, diet, hypertension host genetics, gender, and aging) in the development of MI and therapeutic regulation.
Collapse
Affiliation(s)
- Manisha Bijla
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, India
| | - Sunil Kumar Saini
- Department of Zoology, Swami Shraddhanand College, Delhi University, India
| | - Ajai Kumar Pathak
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia; Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | | | - Katyayani Sukhavasi
- Department of Cardiac Surgery and The Heart Clinic, Tartu University Hospital & Department of Cardiology, Institute of Clinical Medicine, Tartu University, Tartu, Estonia
| | - Ayurshi Patil
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | - Diksha Saini
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | - Rakesh Yadav
- Department of Cardiology, AIIMS, New Delhi, India
| | - Shalini Singh
- ICMR-National Institute of Cancer Prevention and Research, Noida, India
| | | | - Pramod Kumar
- ICMR-National Institute of Cancer Prevention and Research, Noida, India.
| |
Collapse
|
142
|
Fu H, Cai X, Cui L, Nong W, Li W, Mei H, Yang T, Yue H, Huang Q, An Z, Wu Y, Huang X, Zhang X. The evolution of preexisting primary immune thrombocytopenia after COVID-19 onset: A nationally representative, prospective, multicentre, observational study. Ann Hematol 2024; 103:1549-1559. [PMID: 38526649 DOI: 10.1007/s00277-024-05720-0] [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: 12/15/2023] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
The symptoms in patients with primary immune thrombocytopenia (ITP) after COVID-19 onset remain largely unclear. The aim of this study was to describe the platelet count fluctuations in ITP patients following the diagnosis of COVID-19. A prospective multicentre observational study was conducted from December 15th, 2022, to January 31st, 2023 in 39 general hospitals across China. Patients with preexisting primary ITP who were newly diagnosed with COVID-19 were enrolled. A total of 1216 ITP patients with newly-diagnosed COVID-19 were enrolled. 375 (30.8%) patients experienced ITP exacerbation within eight weeks after the diagnosis of COVID-19, and most exacerbation (266/375, 70.9%) developed in the first two weeks. Immunosuppressive therapy for ITP and severe/critical COVID-19 infection were independent variables associated with ITP exacerbation. Overall the platelet count had a transient increasing trend, and the platelet peak value occurred at two weeks after COVID-19 infection. Then, the platelet count decreased to the baseline level in the following weeks. The platelet count had a transient increasing trend in ITP patients following the diagnosis of COVID-19. ITP exacerbation only occurred in less than one-third of ITP patients. Nonimmunosuppressive therapy may have an advantage to prevent ITP exacerbation during COVID-19.
Collapse
Affiliation(s)
- Haixia Fu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Xuan Cai
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Lijuan Cui
- General Hospital of Ningxia Medical University, Lan Zhou, Ningxia, China
| | - Weixia Nong
- Department of Hematology, First Affiliated Hospital, School of Medicine, Shihezi University, Xinjiang, China
| | - Wenqian Li
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tonggji Medical Colloege, Huazhong University of Science and Technology, Wuhan, China
| | - Tonghua Yang
- Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Han Yue
- Department of Hematology, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiusha Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Zhuoyu An
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Yejun Wu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, China.
| |
Collapse
|
143
|
Albabtain MS, Alyousef KA, Alharbi ZM, Almutairi MN, Jawdat D. Characteristics, Outcomes, and Associations of Venous Thromboembolism in Diabetic Patients Infected With COVID-19 in Riyadh, Saudi Arabia. Cureus 2024; 16:e59468. [PMID: 38826952 PMCID: PMC11142384 DOI: 10.7759/cureus.59468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2024] [Indexed: 06/04/2024] Open
Abstract
Background The associations and risk factors for venous thromboembolism (VTE) among hospitalized COVID-19 patients remain ambiguous in the literature, with some conflicting findings, especially in Saudi Arabia. In this study, we aim to elaborate on these data by examining regional patient populations and exploring the incidence, lab findings, and outcomes of VTE among hospitalized COVID-19 patients known to have diabetes mellitus (DM). Methodology This cross-sectional study was conducted at King Abdulaziz Medical City in Riyadh. The BestCare system was used to collect patients' data between September 2020 and February 2022. JMP15 was used for data analysis. Frequencies and percentages were used for categorical data, and median and interquartile ranges were used for quantitative data. The chi-square and Kruskal-Wallis rank-sum tests were used to assess the difference between categorical and quantitative variables, respectively. Nominal logistical regression was used to assess diabetes as a risk factor for developing VTE among COVID-19 patients. Results Data from 153 admitted patients were collected after they satisfied the inclusion criteria. Of these patients, 39 (25.49%) developed VTE. The demographic data included age group, gender, and DM status presented as frequencies and percentages. Through bivariate analysis, patients with longer hospital stays had at least one episode of VTE (p = 0.0072). Using nominal logistic regression analysis, diabetes as a risk factor (odds ratio = 4.11, confidence interval = 0.955-5.05, p = 0.0287) was significantly associated with the development of VTE in COVID-19 patients. Conclusions Based on our study, diabetes proved significant when evaluating the possible factors regarding VTE development in COVID-19 patients. In addition, the length of stay also played a critical role in the severity of VTE in COVID-19 patients. Similar studies should be conducted on a national scale in Saudi Arabia to accomplish two goals: first, to gain further understanding of the impact of the variables investigated in our population, and second, to publish data that are more generalizable to the larger population of Saudi Arabia.
Collapse
Affiliation(s)
- Mansour S Albabtain
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| | - Khalid A Alyousef
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| | - Ziad M Alharbi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| | - Mohammed N Almutairi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| | - Dunia Jawdat
- Cellular Therapy Services, King Abdullah International Medical Research Center, Riyadh, SAU
| |
Collapse
|
144
|
Bruno AM, Allshouse AA, Benson AE, Yost CC, Metz TD, Varner MW, Silver RM, Branch DW. Thrombotic Markers in Pregnant Patients with and without SARS-CoV-2 Infection. Am J Perinatol 2024; 41:e3202-e3209. [PMID: 37967868 PMCID: PMC11427751 DOI: 10.1055/a-2211-5052] [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] [Indexed: 11/17/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is associated with coagulation abnormalities and increased risk for venous and arterial thrombi. This study aimed to evaluate D-dimer levels and lupus anticoagulant (LAC) positivity in pregnant individuals with and without Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. STUDY DESIGN This was a prospective cohort study of pregnant individuals delivering at a single academic institution from April 2020 to March 2022. Individuals with a positive SARS-CoV-2 result during pregnancy were compared with a convenience sample of those without a positive SARS-CoV-2 result. For individuals with SARS-CoV-2 infection, severity was assessed based on the National Institutes of Health classification system. The primary outcome was D-dimer level measured during delivery admission. The secondary outcomes were LAC positivity and thromboembolic events. Outcomes were compared between individuals with and without a positive SARS-CoV-2 result, and further by disease severity. RESULTS Of 98 participants, 77 (78.6%) were SARS-CoV-2 positive during pregnancy. Among individuals with SARS-CoV-2 infection, severity was asymptomatic in 20 (26.0%), mild in 13 (16.9%), moderate in 4 (5.2%), severe in 38 (49.4%), and critical in 2 (2.6%). The D-dimer concentration at delivery did not significantly differ between those with a SARS-CoV-2 positive result compared with those without (mean 2.03 µg/mL [95% confidence interval {CI} 1.72-2.40] vs. 2.37 µg/mL [95% CI 1.65-3.40]; p = 0.43). Three individuals (4%) with SARS-CoV-2 infection and none (0%) without infection were LAC positive (p = 0.59). There were no clinically apparent thromboses in either group. D-dimer concentrations and LAC positive results did not differ by COVID-19 severity. CONCLUSION Thrombotic markers did not differ in pregnant individuals by SARS-CoV-2 infection; however, high rates of LAC positivity were detected. KEY POINTS · Thrombotic markers did not differ in pregnant individuals by SARS-CoV-2 infection.. · Higher than expected rates of LAC positivity were detected.. · There were no clinically apparent thromboses..
Collapse
Affiliation(s)
- Ann M Bruno
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
- Department of Obstetrics & Gynecology, Intermountain Health, Salt Lake City, Utah
| | - Amanda A Allshouse
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
| | - Ashley E Benson
- Department of Obstetrics & Gynecology, Oregon Health and Science University, Portland, Oregon
| | - Christian Con Yost
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
- Molecular Medicine Program, Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Torri D Metz
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
- Department of Obstetrics & Gynecology, Intermountain Health, Salt Lake City, Utah
| | - Michael W Varner
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
| | - Robert M Silver
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
| | - D Ware Branch
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah
- Department of Obstetrics & Gynecology, Intermountain Health, Salt Lake City, Utah
| |
Collapse
|
145
|
Riou M, Coste F, Meyer A, Enache I, Talha S, Charloux A, Reboul C, Geny B. Mechanisms of Pulmonary Vasculopathy in Acute and Long-Term COVID-19: A Review. Int J Mol Sci 2024; 25:4941. [PMID: 38732160 PMCID: PMC11084496 DOI: 10.3390/ijms25094941] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Despite the end of the pandemic, coronavirus disease 2019 (COVID-19) remains a major public health concern. The first waves of the virus led to a better understanding of its pathogenesis, highlighting the fact that there is a specific pulmonary vascular disorder. Indeed, COVID-19 may predispose patients to thrombotic disease in both venous and arterial circulation, and many cases of severe acute pulmonary embolism have been reported. The demonstrated presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) within the endothelial cells suggests that direct viral effects, in addition to indirect effects of perivascular inflammation and coagulopathy, may contribute to pulmonary vasculopathy in COVID-19. In this review, we discuss the pathological mechanisms leading to pulmonary vascular damage during acute infection, which appear to be mainly related to thromboembolic events, an impaired coagulation cascade, micro- and macrovascular thrombosis, endotheliitis and hypoxic pulmonary vasoconstriction. As many patients develop post-COVID symptoms, including dyspnea, we also discuss the hypothesis of pulmonary vascular damage and pulmonary hypertension as a sequela of the infection, which may be involved in the pathophysiology of long COVID.
Collapse
Affiliation(s)
- Marianne Riou
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Florence Coste
- EA4278, Laboratoire de Pharm-Ecologie Cardiovasculaire, UFR Sciences Technologies Santé, Pôle Sport et Recherche, 74 rue Louis Pasteur, 84000 Avignon, France; (F.C.); (C.R.)
| | - Alain Meyer
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Irina Enache
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Samy Talha
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Anne Charloux
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| | - Cyril Reboul
- EA4278, Laboratoire de Pharm-Ecologie Cardiovasculaire, UFR Sciences Technologies Santé, Pôle Sport et Recherche, 74 rue Louis Pasteur, 84000 Avignon, France; (F.C.); (C.R.)
| | - Bernard Geny
- Translational Medicine Federation of Strasbourg (FMTS), University of Strasbourg, CRBS, Team 3072 “Mitochondria, Oxidative Stress and Muscle Protection”, 1 rue Eugène Boeckel, CS 60026, 67084 Strasbourg, France; (M.R.); (A.M.); (I.E.); (S.T.); (A.C.)
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, 1 Place de l’hôpital, 67091 Strasbourg, France
| |
Collapse
|
146
|
Larrosa-García M, Garcia-Garcia S, Louro J, Sánchez-Montalvá A, Sampol Sirvent J, Augustín Recio S, Guillén Del Castillo A, Riera-Arnau J, Gorgas MQ, Miarons M. Use of chronic medications and risk of death due to COVID-19 in hospitalised patients. Eur J Hosp Pharm 2024; 31:247-252. [PMID: 36302612 PMCID: PMC11042468 DOI: 10.1136/ejhpharm-2021-003186] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 10/11/2022] [Indexed: 06/16/2023] Open
Abstract
OBJECTIVES To evaluate the potential association between chronic exposure to medication and death related to COVID-19. METHODS This is a retrospective cross-sectional study that included all patients hospitalised due to COVID-19 from 11 March to 4 June 2020 in our centre. Chronic patient medication was classified by the Anatomical Therapeutic Chemical (ATC) classification; demographic and clinical data were analysed. Multivariate logistic regression models were used to estimate the adjusted odds ratios (aOR) of death for each drug exposure; each aOR represents an independent model adjusted by clinical factors related to COVID-19 mortality. RESULTS The study included 978 patients with a mean (SD) age of 64.5 (17.7) years who were predominantly male (531, 54.3%). Of all 978 patients, 182 (18.61%) died during the follow-up of the study. The most common Charlson Comorbidity Index (CCI) was 0, 4.2% were smokers, 16.7% were obese, 47.4% had hypertension, and 19.4% were diabetic. Most patients (70.8%) were prescribed at least one treatment, 32.5% used >5 treatments, and 8.6% >10. Our data suggest that COVID-19 hospitalised patients taking trimethoprim and analogues, leukotriene receptor antagonists, calcineurin inhibitors, aldosterone antagonists, selective immunosuppressants, propulsives, insulins and analogues, and benzodiazepine derivatives have a higher risk of death. CONCLUSIONS This study investigated the association between chronic exposure to drugs and the risk of death in COVID-19 patients. Our results have shed some light on the impact of chronic drug exposure on the risk of severe COVID-19; however, further research is needed to increase the understanding about its relevance.
Collapse
Affiliation(s)
| | | | - Javier Louro
- Department of Epidemiology and Evaluation, IMIM, Hospital del Mar INAD, Barcelona, Spain
| | | | | | | | | | - Judit Riera-Arnau
- Pharmacology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | - Marta Miarons
- Pharmacy Department, Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
147
|
刘 鑫, 石 雪, 李 军. [A case of COVID-19 associated ischemic colitis]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2024; 56:362-365. [PMID: 38595259 PMCID: PMC11004963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Indexed: 04/11/2024]
Abstract
Ischemic colitis is a disease in which local tissue in the intestinal wall dies to varying degrees due to insufficient blood supply to the colon. Risk factors include cardiovascular disease, diabetes, chronic kidney disease, chronic obstructive pulmonary disease, etc. Typical clinical manifestations of the disease are abdominal pain and hematochezia. The most common locations are the watershed areas of splenic flexure and rectosigmoid junction. The lesions are segmental and clearly demarcated from normal mucosa under endoscopy. The digestive tract is a common extra-pulmonary organ affected by the novel coronavirus, which can be directly damaged by the virus or indirectly caused by virus-mediated inflammation and hypercoagulability. The corona virus disease 2019 (COVID-19) associated intestinal injury can be characterized by malabsorption, malnutrition, intestinal flora shift, etc. CT can show intestinal ischemia, intestinal wall thickening, intestinal wall cystoid gas, intestinal obstruction, ascites, intussusception and other signs. In this study, we reported a case of ischemic colitis in a moderate COVID-19 patient. The affected area was atypical and the endoscope showed diffuse lesions from the cecum to the rectosigmoid junction. No signs of intestinal ischemia were found on imaging and clear thrombosis in small interstitial vessels was found in pathological tissue. Combined with the fact that the patient had no special risk factors in his past history, the laboratory tests indicated elevated ferritin and D-dimer, while the autoantibodies and fecal etiology results were negative, we speculated that the hypercoagulability caused by novel coronavirus infection was involved in the occurrence and development of the disease in this patient. After prolonged infusion support and prophylactic anti-infection therapy, the patient slowly resumed diet and eventually went into remission. Finally, we hoped to attract clinical attention with the help of this case of moderate COVID-19 complicated with ischemic colitis which had a wide range of lesions and a slow reco-very. For patients with abdominal pain and blood in the stool after being diagnosed as COVID-19, even if they are not severe COVID-19, they should be alert to the possibility of ischemic colitis, so as not to be mistaken for gastrointestinal reactions related to COVID-19.
Collapse
Affiliation(s)
- 鑫 刘
- 北京大学第三医院消化科,北京 100191Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - 雪迎 石
- 北京大学第三医院病理科,北京 100191Department of Pathology, Peking University Third Hospital, Beijing 100191, China
| | - 军 李
- 北京大学第三医院消化科,北京 100191Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
148
|
Wainwright BS, Chihade DB, Costanza MJ, Feghali AC, Shaw PM. Paradigm Shift of Interventional Strategies and Outcomes for Acute Limb Ischemia Post-Pandemic. J Endovasc Ther 2024:15266028241246162. [PMID: 38606923 DOI: 10.1177/15266028241246162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
PURPOSE We performed a large-scale comparison of patients treated for acute limb ischemia (ALI) in the pre-COVID (2017-2019) and COVID (2020-2022) eras to evaluate changes in interventional strategies and compare factors associated with adverse outcomes. We sought to characterize patient outcomes in an evolving ALI treatment algorithm in response to pandemic-associated presentation delays and rapid technological advancements in mechanical thrombectomy (MT). METHODS Using the TriNetX global research network, we conducted a multicenter query across 80 health care organizations (HCOs) spanning 4 countries for patients treated for ALI. Propensity score matching was performed to account for comorbidities. Risk of adverse outcomes within 30 days was calculated for each era, including re-intervention (RI30), major/minor amputation, and death. Patients were then stratified by initial intervention: open revascularization (OR), MT, or catheter-directed thrombolysis and adjunctive endovascular procedures alone (CDT/EP). Risk of adverse outcomes was compared between treatment groups of the same era. RESULTS After propensity score matching, the pre-COVID era and COVID era cohorts included 7344 patients each. COVID era patients experienced a statistically significant higher risk of 30-day mortality (RR=1.211, p=0.027). Mechanical thrombectomy interventions were performed more frequently in the COVID era (RR=1.314, p<0.0001). Comparing outcomes between treatment groups, MT patients required RI30 more than OR patients (pre-COVID: RR=2.074, p=0.006; COVID: RR=1.600, p=0.025). Open revascularization patients had higher 30-day mortality (pre-COVID: RR=2.368, p<0.0001; COVID: RR=2.013, p<0.0001) and major amputations (pre-COVID: RR=2.432, p<0.0001; COVID: RR=2.176, p<0.0001) than CDT/EP. Pre-COVID CDT/EP patients were at higher risk for RI30 (RR=1.449, p=0.005) and minor amputations (RR=1.500, p=0.010) than OR. The MT group had higher major amputation rates than CDT/EP (pre-COVID: RR=2.043, p=0.019; COVID: RR=1.914, p=0.007). COVID-era MT patients had greater 30-day mortality (RR=1.706, p=0.031) and RI30 (RR=1.544, p=0.029) than CDT/EP. CONCLUSION Significant shifts toward an MT-based approach have been observed in the last 3 years. Although MT required more RI30 than OR, there was no associated consequence of mortality and limb salvage. The increased mortality seen among COVID-era patients could be explained by delayed presentation, as well as poorly understood pro-thrombogenic or pro-inflammatory mechanisms related to the first waves of COVID. More research is necessary to determine an optimal treatment algorithm. CLINICAL IMPACT Comorbid risk factors and severity of ischemia must be carefully considered before selecting an interventional strategy to prevent adverse outcomes and maximize limb salvage. Open revascularization strategies are associated with increased mortality and limb loss compared to less-invasive thrombolytic therapy alone. Mechanical thrombectomy (MT)-based approaches have been increasingly used in the last 3 years. Patients receiving MT are more likely to require reintervention within 30 days.
Collapse
Affiliation(s)
- Brandon S Wainwright
- Norton College of Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Deena B Chihade
- Division of Vascular Surgery & Endovascular Services, Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Michael J Costanza
- Division of Vascular Surgery & Endovascular Services, Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Anthony C Feghali
- Division of Vascular Surgery & Endovascular Services, Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Palma M Shaw
- Division of Vascular Surgery & Endovascular Services, Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
149
|
Hafez W, Rashid A, Abuelsaoud HM, Jose M, Kishk S, Gador M, Emoshe T, Abdulaal F, Nair N, Ahmad M, Rashid VJ, Faheem Y, John S, Ahmed S, Daraghmi A, Soliman R, Abdelrahman A, Mohamed AA, Ghanem M. Evaluating the potential mediating role of ADAMTS13 activity in the relationship between obesity and the severity of COVID-19: A retrospective cohort study. Medicine (Baltimore) 2024; 103:e37806. [PMID: 38608066 PMCID: PMC11018207 DOI: 10.1097/md.0000000000037806] [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: 08/17/2023] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
Obesity and low enzyme A disintegrin and metalloproteinase with thrombospondin type-1 motif-13 (ADAMTS13) activity have been linked to poor coronavirus disease 2019 (COVID-19). Given that obesity may influence ADAMTS13 activity, it is feasible; however, it remains unclear whether ADAMTS13 activity acts as a mediator between obesity and COVID-19 outcomes. We investigated the link between body mass index (BMI) and COVID-19 outcomes, using ADAMTS13 activity as a mediator. ADAMTS13 activity was measured in 86 hospitalized COVID-19 patients. BMI, ADAMTS13 activity, and COVID-19 outcomes were assessed. Obese patients had a high odds ratio for low ADAMTS13 levels. When different levels of ADAMTS13 activity were considered, the severity of COVID-19 in obese patients was 4.5 times that in the normal BMI group. Furthermore, increased coagulopathy indicators correlated with low ADAMTS13 activity. Patients with elevated ALT and AST levels showed a 3 to 4-fold increase in the chances of low ADAMTS13 activity (OR:3.19, 95% CI:1.22-8.90, P = .021; OR:2.17, 95% CI:0.91-5.27, P = .082, respectively). When ADAMTS13 activity was considered, obese patients had greater COVID-19 severity and slower viral clearance than those with normal BMI. Low ADAMTS13 activity and impaired liver function are associated with poor COVID-19 outcomes. These findings encourage researchers to use molecular component identification to study the effects of obesity on the von Willebrand factor (VWF)/ADAMTS13 axis, COVID-19 pathogenesis, and outcomes.
Collapse
Affiliation(s)
- Wael Hafez
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
- Internal Medicine Department, Medical Research and Clinical Studies Institute; The National Research Centre, Cairo, Egypt
| | - Asrar Rashid
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | | | - Mohan Jose
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Samy Kishk
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Muneir Gador
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | | | - Fatema Abdulaal
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Nivedita Nair
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Muhammad Ahmad
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | | | - Youmna Faheem
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Steffi John
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Sabah Ahmed
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Ahmed Daraghmi
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| | - Rami Soliman
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
- National Institute of Chest and Allergy, Egypt
| | - Ahmed Abdelrahman
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
- Internal Medicine Department, Zagazig Faculty of Medicine, Zagazig, Egypt
| | - Ahmed Ali Mohamed
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
- Intensive Care Department, Theodor Bilharz Research Institute, AL Warak, Giza Governorate, Egypt
| | - Mirvat Ghanem
- NMC Royal Hospital, Khalifa City, Abu Dhabi, United Arab Emirates
| |
Collapse
|
150
|
Altin N, Tiğlioğlu P, Ulusoy TU, Aydin FN, Kar İ, Karakoc B, Utebey G. A challenging issue in COVID-19 infection: The relationship between PA1-1 and TAFI levels in patients with coagulation disorder: A retrospective and observational study. Medicine (Baltimore) 2024; 103:e37802. [PMID: 38608056 PMCID: PMC11018242 DOI: 10.1097/md.0000000000037802] [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: 01/12/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
COVID-19 disrupts the balance between coagulation and fibrinolysis. Especially in the clinical course of serious disease, plasminogen activator inhibitor-1 (PAI-1), thrombin activatable fibrinolysis inhibitor (TAFI), and tissue plasminogen activator levels increase in association with hypercoagulable state and hypofibrinolysis. This explains the increased incidence of thrombosis seen in COVID-19 infection. In this study, we aimed to examine the changes in PAI-1 and TAFI levels of COVID-19 patients. Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Dişkapi Yildirim Beyazit Training and Research Hospital-Ankara Turkey, between April 1 and May 7, 2021. Patients who were diagnosed with COVID-19 were included in this retrospective study. TAFI and PAI-1 levels were analyzed from the samples that had been stored at -80 °C formerly. One hundred thirty-five patients diagnosed with COVID-19 and followed up in the service or intensive care unit were included in the study. Thirty-four (25.2%) patients required follow-up in the intensive care unit. Mortality rate was 10.4%, the coagulation tests of these patients were also compared. PA1-1 levels were found to be statistically significantly higher in intensive care unit patients (median: 133 pg/mL vs 31 pg/mL; P < .001), and there was no significant difference in TAFI levels (median:7.31 ng/mL vs 9.80 ng/mL; P = .171) between the 2 groups. TAFI levels were found to be higher in patients who died. In COVID-19 infection, as the severity of the disease increases, the coagulation balance deteriorates and eventually a hypercoagulable state occurs with an increase in PAI-1 and TAFI levels. Markers such as PAI and TAFI can be illuminating in further studies in determining prognosis and mortality and developing new treatment options.
Collapse
Affiliation(s)
- Nilgun Altin
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Dişkapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Pinar Tiğlioğlu
- Department of Hematology, Sancaktepe Prof. Dr. Sehit Ilhan Varank Training and Research Hospital, Istanbul, Turkey
| | - Tulay Unver Ulusoy
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Dişkapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Fevzi Nuri Aydin
- Department of Biochemistry, University of Health Sciences, Dişkapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - İrem Kar
- Department of Biostatistics, Ankara University, Ankara, Turkey
| | - Busra Karakoc
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Dişkapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Gulten Utebey
- Department of Anesthesia and Reanimation, University of Health Sciences, Dişkapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| |
Collapse
|