201
|
Ambereen A, Rahman SA, Rehman S, Zaidi K, Arif SH. Mandibular mucormycosis following SARS-CoV-2 infection - A case report and review of literature. CLINICAL INFECTION IN PRACTICE 2021; 12:100099. [PMID: 34568807 PMCID: PMC8450209 DOI: 10.1016/j.clinpr.2021.100099] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The second wave of COVID-19 pandemic has seen an unprecedented rise in the number of mucormycosis cases worldwide and in India particularly. This otherwise rare fungal infection has become an endemic among patients who have recovered from recent SARS-CoV-2 infection. Among the different types of mucormycosis, rhino-orbital-cerebral involvement has mainly been observed in the recent surge of cases. Very few cases of mucormycosis of mandible have been reported in literature and none in COVID-19 patients. We report a case of isolated mandibular mucormycosis in a COVID- 19 patient, with no other predisposing comorbidities.Case report.A 39 year old patient recently recovered from COVID-19 presented with typical symptoms of osteomyelitis which was confirmed using computed tomography of face. He underwent thorough debridement and curettage and tissue was sent for culture, special staining and biopsy. RESULT Diagnosis of mucormycosis was confirmed based on postoperative biopsy and special staining. He was further managed with complete course of appropriate antifungal therapy. CONCLUSION Mucormycosis is a fulminant and aggressive infection which requires prompt diagnosis and intervention. Early referral to a maxillofacial surgeon by physicians and general dental practitioners on seeing signs and symptoms of secondary fungal infections involving maxilla or mandible in patients with history of SARS-CoV-19 infection can improve prognosis.
Collapse
Affiliation(s)
- Aafiya Ambereen
- axillofacial Surgery Dr ZA Dental College Aligarh Muslim University, Indi
| | - Sajjad A Rahman
- axillofacial Surgery Dr ZA Dental College Aligarh Muslim University, Indi
| | - Suhailur Rehman
- Department of Pathology JN Medical College Aligarh Muslim University, India
| | - Kamran Zaidi
- axillofacial Surgery Dr ZA Dental College Aligarh Muslim University, Indi
| | - S H Arif
- Department of Pathology JN Medical College Aligarh Muslim University, India
| |
Collapse
|
202
|
The role of lipoprotein(a) in coronavirus disease 2019 (COVID-19) with relation to development of severe acute kidney injury. J Thromb Thrombolysis 2021; 53:581-585. [PMID: 34709533 PMCID: PMC8552425 DOI: 10.1007/s11239-021-02597-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 12/20/2022]
Abstract
Lipoprotein(a) (Lp(a)) is a prothrombotic and anti-fibrinolytic lipoprotein, whose role has not been clearly defined in the pathogenesis of coronavirus disease 2019 (COVID-19). In this prospective observational study, serum Lp(a) as well as outcomes were measured in 50 COVID-19 patients and 30 matched sick controls. Lp(a) was also assessed for correlation with a wide panel of biomarkers. Serum Lp(a) did not significantly differ between COVID-19 patients and sick controls, though its concentration was found to be significantly associated with severity of COVID-19 illness, including acute kidney failure stage (r = 0.380, p = 0.007), admission disease severity (r = 0.355, p = 0.013), and peak severity (r = 0.314; p = 0.03). Lp(a) was also positively correlated with interleukin (IL)-8 (r = 0.308; p = 0.037), fibrinogen (r = 0.344; p = 0.032) and creatinine (r = 0.327; p = 0.027), and negatively correlated with ADAMTS13 activity/VWF:Ag (r = − 0.335; p = 0.021); but not with IL-6 (r = 0.241; p = 0.106). These results would hence suggest that adverse outcomes in patients with COVID-19 may be aggravated by a genetically determined hyper-Lp(a) state rather than any inflammation induced elevations.
Collapse
|
203
|
Rando HM, MacLean AL, Lee AJ, Lordan R, Ray S, Bansal V, Skelly AN, Sell E, Dziak JJ, Shinholster L, D’Agostino McGowan L, Ben Guebila M, Wellhausen N, Knyazev S, Boca SM, Capone S, Qi Y, Park Y, Mai D, Sun Y, Boerckel JD, Brueffer C, Byrd JB, Kamil JP, Wang J, Velazquez R, Szeto GL, Barton JP, Goel RR, Mangul S, Lubiana T, COVID-19 Review Consortium, Gitter A, Greene CS. Pathogenesis, Symptomatology, and Transmission of SARS-CoV-2 through Analysis of Viral Genomics and Structure. mSystems 2021; 6:e0009521. [PMID: 34698547 PMCID: PMC8547481 DOI: 10.1128/msystems.00095-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 02/06/2023] Open
Abstract
The novel coronavirus SARS-CoV-2, which emerged in late 2019, has since spread around the world and infected hundreds of millions of people with coronavirus disease 2019 (COVID-19). While this viral species was unknown prior to January 2020, its similarity to other coronaviruses that infect humans has allowed for rapid insight into the mechanisms that it uses to infect human hosts, as well as the ways in which the human immune system can respond. Here, we contextualize SARS-CoV-2 among other coronaviruses and identify what is known and what can be inferred about its behavior once inside a human host. Because the genomic content of coronaviruses, which specifies the virus's structure, is highly conserved, early genomic analysis provided a significant head start in predicting viral pathogenesis and in understanding potential differences among variants. The pathogenesis of the virus offers insights into symptomatology, transmission, and individual susceptibility. Additionally, prior research into interactions between the human immune system and coronaviruses has identified how these viruses can evade the immune system's protective mechanisms. We also explore systems-level research into the regulatory and proteomic effects of SARS-CoV-2 infection and the immune response. Understanding the structure and behavior of the virus serves to contextualize the many facets of the COVID-19 pandemic and can influence efforts to control the virus and treat the disease. IMPORTANCE COVID-19 involves a number of organ systems and can present with a wide range of symptoms. From how the virus infects cells to how it spreads between people, the available research suggests that these patterns are very similar to those seen in the closely related viruses SARS-CoV-1 and possibly Middle East respiratory syndrome-related CoV (MERS-CoV). Understanding the pathogenesis of the SARS-CoV-2 virus also contextualizes how the different biological systems affected by COVID-19 connect. Exploring the structure, phylogeny, and pathogenesis of the virus therefore helps to guide interpretation of the broader impacts of the virus on the human body and on human populations. For this reason, an in-depth exploration of viral mechanisms is critical to a robust understanding of SARS-CoV-2 and, potentially, future emergent human CoVs (HCoVs).
Collapse
Affiliation(s)
- Halie M. Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, USA
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sandipan Ray
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Vikas Bansal
- Biomedical Data Science and Machine Learning Group, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Ashwin N. Skelly
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Sell
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John J. Dziak
- Edna Bennett Pierce Prevention Research Center, The Pennsylvania State University, University Park, Pennsylvania, USA
| | | | - Lucy D’Agostino McGowan
- Department of Mathematics and Statistics, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Marouen Ben Guebila
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Simina M. Boca
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
| | - Stephen Capone
- St. George’s University School of Medicine, St. George’s, Grenada
| | - Yanjun Qi
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
| | - YoSon Park
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Mai
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yuchen Sun
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
| | - Joel D. Boerckel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - James Brian Byrd
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Jeremy P. Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| | - Jinhui Wang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - John P. Barton
- Department of Physics and Astronomy, University of California-Riverside, Riverside, California, USA
| | - Rishi Raj Goel
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Serghei Mangul
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Tiago Lubiana
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - COVID-19 Review Consortium
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
- Biomedical Data Science and Machine Learning Group, German Center for Neurodegenerative Diseases, Tübingen, Germany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Edna Bennett Pierce Prevention Research Center, The Pennsylvania State University, University Park, Pennsylvania, USA
- Mercer University, Macon, Georgia, USA
- Department of Mathematics and Statistics, Wake Forest University, Winston-Salem, North Carolina, USA
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
- Georgia State University, Atlanta, Georgia, USA
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
- St. George’s University School of Medicine, St. George’s, Grenada
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Clinical Sciences, Lund University, Lund, Sweden
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
- Azimuth1, McLean, Virginia, USA
- Allen Institute for Immunology, Seattle, Washington, USA
- Department of Physics and Astronomy, University of California-Riverside, Riverside, California, USA
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, California, USA
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Casey S. Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| |
Collapse
|
204
|
Larijani B, Foroughi-Heravani N, Abedi M, Tayanloo-Beik A, Rezaei-Tavirani M, Adibi H, Arjmand B. Recent Advances of COVID-19 Modeling Based on Regenerative Medicine. Front Cell Dev Biol 2021; 9:683619. [PMID: 34760882 PMCID: PMC8573217 DOI: 10.3389/fcell.2021.683619] [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: 03/21/2021] [Accepted: 09/22/2021] [Indexed: 11/13/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has caused a pandemic since December 2019 that originated in Wuhan, China. Soon after that, the world health organization declared Coronavirus disease-2019 a global health concern. SARS-CoV-2 is responsible for a lethal respiratory infection as well as the involvement of other organs due to its large tropism spectrum such as neurologic, cardiovascular, endocrine, gastrointestinal, and renal systems. Since the behavior of the virus is not fully understood, a new manifestation of the infection is revealed every day. In order to be able to design more efficient drugs and vaccines to treat the infection, finding out the exact mechanism of pathogenicity would be necessary. Although there have been some big steps toward understanding the relevant process, there are still some deficiencies in this field. Accordingly, regenerative medicine (RM), can offer promising opportunities in discovering the exact mechanisms and specific treatments. For instance, since it is not always possible to catch the pathophysiology mechanisms in human beings, several modeling methods have been introduced in this field that can be studied in three main groups: stem cell-based models, organoids, and animal models. Regarding stem cell-based models, induced pluripotent stem cells are the major study subjects, which are generated by reprogramming the somatic stem cells and then directing them into different adult cell populations to study their behavior toward the infection. In organoid models, different cell lines can be guided to produce a 3D structure including liver, heart, and brain-like platforms. Among animal models, mice are the most common species in this field. However, in order for mice models to be permissive to the virus, angiotensin-converting enzyme 2 receptors, the main receptor involved in the pathogenicity of the virus, should be introduced to the host cells through different methods. Here, the current known mechanism of SARS-CoV-2 infection, different suggested models, the specific response toward different manipulation as well as challenges and shortcomings in each case have been reviewed. Finally, we have tried to provide a quick summary of the present available RM-based models for SARS-CoV-2 infection, as an essential part of developing drugs, for future therapeutic goals.
Collapse
Affiliation(s)
- Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Najmeh Foroughi-Heravani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Abedi
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Adibi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
205
|
A review of ischemic stroke in COVID-19: currently known pathophysiological mechanisms. Neurol Sci 2021; 43:67-79. [PMID: 34671854 PMCID: PMC8528653 DOI: 10.1007/s10072-021-05679-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19), the third type of coronavirus pneumonia after severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS), is spreading widely worldwide now. This pneumonia causes not only respiratory symptoms but also multiple organ dysfunction, including thrombotic diseases such as ischemic stroke. The purpose of this review is to explore whether COVID-19 is a risk factor for ischemic stroke and its related pathophysiological mechanisms. Based on the high thrombosis rate and frequent strokes of COVID-19 patients, combined with related laboratory indicators and pathological results, the discussion is mainly from two aspects: nerve invasion and endothelial dysfunction. SARS-CoV-2 can directly invade the CNS through blood-borne and neuronal retrograde pathways, causing cerebrovascular diseases. In addition, the endothelial dysfunction in COVID-19 is almost certain. Cytokine storm causes thromboinflammation, and downregulation of ACE2 leads to RAS imbalance, which eventually lead to ischemic stroke.
Collapse
|
206
|
Purkayastha P, Iftikhar MJ, Kostrubiec M. Ventricular Septal Defect in Delayed Presentation of ST-Elevation Myocardial Infarction (STEMI) Secondary to COVID-19 Pandemic. Cureus 2021; 13:e17913. [PMID: 34660108 PMCID: PMC8510695 DOI: 10.7759/cureus.17913] [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] [Accepted: 08/26/2021] [Indexed: 12/03/2022] Open
Abstract
Like many other countries at the moment, the United Kingdom (UK) is currently under national lockdown due to the coronavirus disease 2019 (COVID-19) pandemic. An unfortunate consequence of such social isolation measures is that patients with genuine acute medical emergencies may not present to a hospital in a timely manner. We present such a scenario, whereby a patient had a delayed presentation of ST-elevation myocardial infarction (STEMI) due to fear of breaching COVID-19 lockdown rules. As a result of the patient presenting well outside the optimal treatment window, her STEMI was complicated by a severe ventricular septal defect (VSD). We discuss how the COVID-19 pandemic has influenced the nature and management of STEMIs and associated issues.
Collapse
|
207
|
Ai J, Hong W, Wu M, Wei X. Pulmonary vascular system: A vulnerable target for COVID-19. MedComm (Beijing) 2021; 2:531-547. [PMID: 34909758 PMCID: PMC8662299 DOI: 10.1002/mco2.94] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 02/05/2023] Open
Abstract
The number of coronavirus disease 2019 (COVID‐19) cases has been increasing significantly, and the disease has evolved into a global pandemic, posing an unprecedented challenge to the healthcare community. Angiotensin‐converting enzyme 2, the binding and entry receptor of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) in hosts, is also expressed on pulmonary vascular endothelium; thus, pulmonary vasculature is a potential target in COVID‐19. Indeed, pulmonary vascular thickening is observed by early clinical imaging, implying a tropism of SARS‐CoV‐2 for pulmonary vasculature. Recent studies reported that COVID‐19 is associated with vascular endothelial damage and dysfunction along with inflammation, coagulopathy, and microthrombosis; all of these pathologic changes are the hallmarks of pulmonary vascular diseases. Notwithstanding the not fully elucidated effects of COVID‐19 on pulmonary vasculature, the vascular endotheliopathy that occurs after infection is attributed to direct infection and indirect damage mainly caused by renin‐angiotensin‐aldosterone system imbalance, coagulation cascade, oxidative stress, immune dysregulation, and intussusceptive angiogenesis. Degradation of endothelial glycocalyx exposes endothelial cell (EC) surface receptors to the vascular lumen, which renders pulmonary ECs more susceptible to SARS‐CoV‐2 infection. The present article reviews the potential pulmonary vascular pathophysiology and clinical presentations in COVID‐19 to provide a basis for clinicians and scientists, providing insights into the development of therapeutic strategies targeting pulmonary vasculature.
Collapse
Affiliation(s)
- Jiayuan Ai
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu Sichuan PR China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu Sichuan PR China
| | - Min Wu
- Department of Biomedical Sciences School of Medicine and Health Sciences University of North Dakota Grand Forks North Dakota USA
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu Sichuan PR China
| |
Collapse
|
208
|
Lamponi S. Bioactive Natural Compounds with Antiplatelet and Anticoagulant Activity and Their Potential Role in the Treatment of Thrombotic Disorders. Life (Basel) 2021; 11:1095. [PMID: 34685464 PMCID: PMC8540276 DOI: 10.3390/life11101095] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/19/2022] Open
Abstract
Natural anticoagulant drugs can be obtained from plants, rich in secondary bioactive metabolites which, in addition to being effective antioxidants, also possess anticoagulant and antiplatelet properties and, for this reason, can be excellent candidates for the treatment of thrombotic diseases. This review reports an overview of the hemostatic process and thrombotic disorders together with data on plants, more and less common from around the world, containing bioactive compounds characterized by antiplatelet and anticoagulant activity. The reported literature was obtained from Medline, PubMed, Elsevier, Web of Science, Google Scholar considering only articles in the English language, published in peer-reviewed journals. The number of citations of the articles and the impact factor of the journals were other parameters used to select the scientific papers to be included in the review. The analysis of the literature data selected demonstrates that many plants' bioactive compounds show antiplatelet and anticoagulant activity that make them potential candidates to be used as new natural compounds able to interfere with both primary and secondary hemostasis. Moreover, they could be used together with anticoagulants currently administered in clinical practice to increase their efficacy and to reduce complications in the treatment of thrombotic disorders.
Collapse
Affiliation(s)
- Stefania Lamponi
- Department of Biotechnologies, Chemistry and Pharmacy and SienabioACTIVE, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
209
|
Evaluation of choroidal thickness in patients who have recovered from COVID-19. Int Ophthalmol 2021; 42:841-846. [PMID: 34611768 PMCID: PMC8491754 DOI: 10.1007/s10792-021-02049-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 09/22/2021] [Indexed: 12/23/2022]
Abstract
Purpose We aimed to evaluate choroidal thickness (CT) in patients who have recovered from COVID-19 by using enhanced depth imaging optical coherence tomography (EDI-OCT). Methods We included fifty-eight patients who have recovered from COVID-19 (group 1) and fifty healthy control subjects (group 2) in this prospective study. Best corrected visual acuity, anterior segment and posterior segment examinations of all subjects were performed. CT scan and measurements were taken with the EDI mode of the Spectral Domain OCT device. Results Of the 108 subjects included in this study, 57 were female and 51 were male. The mean age was similar in both groups (36.10 ± 7.12 and 35.58 ± 7.29, respectively, p = 0.276). Group 1 had the following characteristics: the mean time since diagnosis was 53.18 ± 2.84; it had been 38.48 ± 4.07 days since the PCR test was negative; and all subjects were outpatients. It was detected that the CT of the patients in group 1 decreased in all areas compared to group 2, and this decrease was significant in subfoveal, temporal and inferior areas (257.48 ± 32.79, 273.62 ± 45.04, p = 0.04; 232.96 ± 41.79, 252.76 ± 46.09, p = 0.02, and 245.22 ± 44.58, 271.54 ± 55.07, p = 0.01, respectively). In the retinal nerve fiber layer analysis for group 1, thickening was detected in all areas, although it was not statistically significant, except in the temporal area where it was (superotemporal, superonasal, nasal, inferonasal, inferotemporal, temporal, and global [p = 0 .08, p = 0.45, p = 0.73, p = 0.64, p = 0.74, p = 0.02, and p = 0.10, respectively]). Conclusion For individuals who had recovered from COVID-19, it was found that CT decreased in all areas in these patients. Therefore, this study in which we have demonstrated the decrease in the thickness of the choroidal tissue, a tissue with high blood flow, may contribute to the understanding of the systemic microvascular waste of this disease.
Collapse
|
210
|
Jacobi J. The pathophysiology of sepsis-2021 update: Part 1, immunology and coagulopathy leading to endothelial injury. Am J Health Syst Pharm 2021; 79:329-337. [PMID: 34605875 PMCID: PMC8500113 DOI: 10.1093/ajhp/zxab380] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Disclaimer In an effort to expedite the publication of articles related to the COVID-19 pandemic, AJHP is posting these manuscripts online as soon as possible after acceptance. Accepted manuscripts have been peer-reviewed and copyedited, but are posted online before technical formatting and author proofing. These manuscripts are not the final version of record and will be replaced with the final article (formatted per AJHP style and proofed by the authors) at a later time. Purpose To provide an overview of current literature on the pathophysiology of sepsis, with a focus on mediators of endothelial injury and organ dysfunction. Summary Sepsis is a dysregulated response to infection that triggers cascades of interconnected systems. Sepsis has been a significant cause of mortality worldwide, and the recent viral pandemic that may produce severe sepsis and septic shock has been a major contributor to sepsis-related mortality. Understanding of the pathophysiology of sepsis has changed dramatically over the last several decades. Significant insight into the components of the inflammatory response that contribute to endothelial injury and trigger coagulation pathways has been achieved. Similarly, characterization of anti-inflammatory pathways that may lead to secondary infections and poor outcome has illustrated opportunities for improved therapies. Description of an increasing number of important mediators and pathways has occurred and may point the way to novel therapies to address immune dysregulation. Pharmacists will need a fundamental understanding of the overlapping pathways of the immune response to fully prepare for use of novel treatment options. While pharmacists typically understand coagulation cascade how to utilize anticoagulants, the issues in sepsis related coagulopathy and role of mediators such as cytokines and complement and role of activated platelets and neutrophils require a different perspective. Conclusion Pharmacists can benefit from understanding both the cellular and organ system issues in sepsis to facilitate assessment of potential therapies for risk and benefit.
Collapse
|
211
|
Kartikasari U, Djajalaksana S, Martini H. Acute limb ischemia in a patient with Covid-19 pneumonia: a case report. J Thromb Thrombolysis 2021; 52:974-979. [PMID: 33797021 PMCID: PMC8016154 DOI: 10.1007/s11239-021-02434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has emerged as a pandemic across the world. Hypercoagulability status in COVID-19 is one of the causes of complication from severe COVID-19 with a high risk of arterial thrombosis. Acute Limb Ischemia is a vascular emergency caused by sudden decrease in the arterial perfusion. We report the case of a 53-year-old male patient with COVID-19 Pneumonia, diagnosed with Acute Limb Ischemia. From clinical examination, which included anamnesis, physical examination, and laboratory results as well as chest X-rays, a suspicion of Acute Limb Ischemia was found in a patient with COVID-19 pneumonia. The SARS-CoV-2 real time PCR examination showed positive results. In this patient, the diagnosis of Acute Limb Ischemia with Covid-19 Pneumonia was established through a multidisciplinary approach covering the fields of pulmonology, cardiology, and thoracic and cardiovascular surgery.
Collapse
Affiliation(s)
- Ulfah Kartikasari
- Dr Saiful Anwar General Hospital, Malang, Indonesia.
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Brawijaya University, Dr Saiful Anwar General Hospital, Malang, Indonesia.
| | - Susanthy Djajalaksana
- Dr Saiful Anwar General Hospital, Malang, Indonesia
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Brawijaya University, Dr Saiful Anwar General Hospital, Malang, Indonesia
| | - Heny Martini
- Dr Saiful Anwar General Hospital, Malang, Indonesia
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Dr Saiful Anwar General Hospital, Malang, Indonesia
| |
Collapse
|
212
|
Matucci-Cerinic M, Hughes M, Taliani G, Kahaleh B. Similarities between COVID-19 and systemic sclerosis early vasculopathy: A "viral" challenge for future research in scleroderma. Autoimmun Rev 2021; 20:102899. [PMID: 34274540 PMCID: PMC8280663 DOI: 10.1016/j.autrev.2021.102899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To review similarities between COVID-19 and systemic sclerosis (SSc) early vasculopathy to provide novel insights into both diseases. METHODS A narrative review of the literature supplemented with expert opinion. RESULTS There is clear evidence that the endothelium is at the centre stage in SSc and COVID-19, with endothelial cell activation/injury and dysfunction creating the crucial evolving step in the pathogenesis of both diseases. The angiotensin system has also been implicated in the early stages of both COVID-19 and SSc. Autoptic studies provide novel insights into the effects of SARS-CoV-2 on the endothelium. Normal endothelium and endothelial dysfunction in COVID-19 and SSc are discussed. It is debated whether SARS-CoV-2 infection triggers autoimmunity with production of autoantibodies which is of mechanistic interest because other viral illnesses are potentially involved in endothelial dysfunction and in SSc pathogenesis. CONCLUSION COVID-19 is due to a direct assault of SARS-CoV-2 on the vascular system as an acute infection, whereas SSc remains a chronic/sub-acute autoimmune disease of largely unknown etiology Further study and exploration of the SARS-CoV-2 pathogenic mechanisms might provide further useful milestones in the understanding of the early SSc pathogenesis.
Collapse
Affiliation(s)
- Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence & Division of Rheumatology AOUC, Florence, Italy; Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy.
| | - Michael Hughes
- Department of Rheumatology, Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Gloria Taliani
- Infectious Diseases Unit, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Bashar Kahaleh
- Division of Rheumatology, Allergy and Immunology, University of Toledo Medical Center, Toledo, OH, USA
| |
Collapse
|
213
|
Choudhary S, Sharma K, Singh PK. Von Willebrand factor: A key glycoprotein involved in thrombo-inflammatory complications of COVID-19. Chem Biol Interact 2021; 348:109657. [PMID: 34516971 PMCID: PMC8432980 DOI: 10.1016/j.cbi.2021.109657] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/25/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
COVID-19 is an ongoing public health emergency that has affected millions of people worldwide and is still a threat to many more. One of the pathophysiological features of COVID-19 is associated with the activation of vascular endothelial cells (ECs) leading to the disruption of vascular integrity, coagulation and inflammation. An interlink mechanism between coagulation and inflammatory pathways has been reported in COVID-19. Multiple components are involved in these pathological pathways. Out of all, Von Willebrand Factor (VWF) is one of the primary components of coagulation pathway and also a mediator of vascular inflammation that plays an important role in thrombo-inflammation that further leads to acute respiratory distress syndrome (ARDS). The thrombo-inflammatory co-morbidities such as hyper-coagulation, thrombosis, ARDS etc. have become the major cause of mortality in the patients of COVID-19 admitted to the ICU. Thus, VWF can be explored as a potential target to manage COVID-19 associated co-morbidities. Supporting this hypothesis, there are literature reports which disclose previous attempts to target VWF for the management of thrombo-inflammation in other pathological conditions. The current report summarizes emerging insights into the pathophysiology, mechanism(s), diagnosis, management and foundations for research on this less explored clinically relevant glycoprotein as coagulation biomarker in COVID-19.
Collapse
Affiliation(s)
- Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Kajal Sharma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Pankaj Kumar Singh
- Integrative Physiology and Pharmacology, Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.
| |
Collapse
|
214
|
Pérez-Campos Mayoral L, Hernández-Huerta MT, Papy-García D, Barritault D, Zenteno E, Sánchez Navarro LM, Pérez-Campos Mayoral E, Matias Cervantes CA, Martínez Cruz M, Mayoral Andrade G, López Cervantes M, Vázquez Martínez G, López Sánchez C, Pina Canseco S, Martínez Cruz R, Pérez-Campos E. Immunothrombotic dysregulation in chagas disease and COVID-19: a comparative study of anticoagulation. Mol Cell Biochem 2021; 476:3815-3825. [PMID: 34110554 PMCID: PMC8190527 DOI: 10.1007/s11010-021-04204-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 06/03/2021] [Indexed: 12/27/2022]
Abstract
Chagas and COVID-19 are diseases caused by Trypanosoma cruzi and SARS-CoV-2, respectively. These diseases present very different etiological agents despite showing similarities such as susceptibility/risk factors, pathogen-associated molecular patterns (PAMPs), recognition of glycosaminoglycans, inflammation, vascular leakage hypercoagulability, microthrombosis, and endotheliopathy; all of which suggest, in part, treatments with similar principles. Here, both diseases are compared, focusing mainly on the characteristics related to dysregulated immunothrombosis. Given the in-depth investigation of molecules and mechanisms related to microthrombosis in COVID-19, it is necessary to reconsider a prompt treatment of Chagas disease with oral anticoagulants.
Collapse
Affiliation(s)
- Laura Pérez-Campos Mayoral
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | | | | | | | - Edgar Zenteno
- Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, 04360, México
| | | | - Eduardo Pérez-Campos Mayoral
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | | | | | - Gabriel Mayoral Andrade
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | | | | | - Claudia López Sánchez
- Tecnológico Nacional de México / Instituto Tecnológico de Oaxaca, Oaxaca, 68030, México
| | - Socorro Pina Canseco
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | - Ruth Martínez Cruz
- Centro de Investigación Facultad de Medicina UNAM-UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, 68020, México
| | - Eduardo Pérez-Campos
- Tecnológico Nacional de México / Instituto Tecnológico de Oaxaca, Oaxaca, 68030, México.
- Laboratorio de Patología Clínica "Eduardo Pérez Ortega", Oaxaca, 68000, México.
| |
Collapse
|
215
|
Rehabilitative management of post-acute COVID-19: clinical pictures and outcomes. Rheumatol Int 2021; 41:2167-2175. [PMID: 34580754 DOI: 10.1007/s00296-021-05003-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/16/2021] [Indexed: 12/16/2022]
Abstract
This study aimed to detect patients' characteristics who suffered severe and critical COVID-19 pneumonia admitted to the post-acute COVID-19 rehabilitation clinic in Ankara City Hospital, Physical Medicine and Rehabilitation Hospital and to share our experiences and outcomes of rehabilitation programmes applied. This study was designed as a single-centre, retrospective, observational study. Severe and critical COVID-19 patients, admitted to the post-acute COVID-19 rehabilitation clinic, were included in patient-based rehabilitation programmes, targeting neuromuscular and respiratory recovery. Functional status, oxygen (O2) requirement and daily living activities were assessed before and after rehabilitation. Eighty-five patients, of which 74% were male, were analysed, with the mean age of 58.27 ± 11.13 and mean body mass index of 25.29 ± 4.81 kg/m2. The most prevalent comorbidities were hypertension (49.4%) and diabetes mellitus (34.1%). Of the 85 patients, 84 received antiviral drugs, 81 low-molecular-weight heparin, 71 corticosteroids, 11 anakinra, 4 tocilizumab, 16 intravenous immunoglobulin and 6 plasmapheresis. 78.8% of the patients were admitted to the intensive care unit, with a mean length of stay of 19.41 ± 18.99 days, while those who needed O2 support with mechanic ventilation was 36.1%. Neurological complications, including Guillain-Barré syndrome, critical illness-related myopathy/neuropathy, cerebrovascular disease and steroid myopathy, were observed in 39 patients. On initial functional statuses, 55.3% were bedridden, 22.4% in wheelchair level and 20% mobilised with O2 support. After rehabilitation, these ratios were 2.4%, 4.7% and 8.2%, respectively. During admission, 71 (83.5%) patients required O2 support, but decreased to 7 (8.2%) post-rehabilitation. Barthel Index improved statistically from 44.82 ± 27.31 to 88.47 ± 17.56. Patient-based modulated rehabilitation programmes are highly effective in severe and critical COVID-19 complications, providing satisfactory well-being in daily living activities.
Collapse
|
216
|
Global haemostatic tests demonstrate the absence of parameters of hypercoagulability in non-hypoxic mild COVID-19 patients: a prospective matched study. J Thromb Thrombolysis 2021; 53:646-662. [PMID: 34581945 PMCID: PMC8476716 DOI: 10.1007/s11239-021-02575-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2021] [Indexed: 01/22/2023]
Abstract
Severe COVID-19 patients demonstrate hypercoagulability, necessitating thromboprophylaxis. However, less is known about the haemostatic profile in mild COVID-19 patients. We performed an age and gender-matched prospective study of 10 severe and 10 mild COVID-19 patients. Comprehensive coagulation profiling together with Thromboelastography and Clot Waveform Analysis were performed. FBC, PT, APTT, D-dimer, fibrinogen and CWA were repeated every 3 days for both groups and repeat TEG was performed for severe patients up till 15 days. On recruitment, severe patients had markers reflecting hypercoagulability including raised median D-dimer 1.0 μg/mL (IQR 0.6, 1.4) (p = 0.0004), fibrinogen 5.6 g/L (IQR 4.9, 6.6) (p = 0.002), Factor VIII 206% (IQR 171, 203) and vWF levels 265.5% (IQR 206, 321). Mild patients had normal values of PT, aPTT, fibrinogen and D-dimer, and slightly elevated median Factor VIII and von Willebrand factor (vWF) levels. Repeated 3-day assessments for both groups showed declining trends in D-dimer and Fibrinogen. CWA of severe COVID-19 group demonstrated hypercoagulability with an elevated median values of aPTT delta change 78.8% (IQR 69.8, 85.2) (p = 0.001), aPTT clot velocity (min1) 7.8%/s (IQR 6.7, 8.3) (p = 0.001), PT delta change 22.4% (IQR 19.4, 29.5) (p = 0.004), PT min1 7.1%/s (IQR 6.3, 9.0) (p = 0.02), PT clot acceleration (min 2) 3.6%/s2 (IQR 3.2, 4.5) (p = 0.02) and PT clot deceleration (max2) 2.9%/s2 (IQR 2.5, 3.5) (p = 0.02). TEG of severe patients reflected hypercoagulability with significant increases in the median values of CFF MA 34.6 mm (IQR 27.4,38.6) (p = 0.003), CRT Angle 78.9° (IQR 78.3, 80.0) (p = 0.0006), CRT A10 67.6 mm (IQR 65.8, 69.6) (p = 0.007) and CFF A10 32.0 mm (IQR 26.8, 34.0) (p = 0.003). Mild COVID-19 patients had absent hypercoagulability in both CWA and TEG. 2 severe patients developed thromboembolic events while none occurred in the mild COVID-19 group. Mild COVID-19 patients show absent parameters of hypercoagulability in global haemostatic tests while those with severe COVID-19 demonstrated parameters associated with hypercoagulability on the global haemostatic tests together with raised D-Dimer, fibrinogen, Factor VIII and vWF levels.
Collapse
|
217
|
Tomerak S, Khan S, Almasri M, Hussein R, Abdelati A, Aly A, Salameh MA, Saed Aldien A, Naveed H, Elshazly MB, Zakaria D. Systemic inflammation in COVID‐19 patients may induce various types of venous and arterial thrombosis: A systematic review. Scand J Immunol 2021; 94:e13097. [PMID: 34940978 PMCID: PMC8646950 DOI: 10.1111/sji.13097] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 01/08/2023]
Abstract
COVID‐19 is a global pandemic with a daily increasing number of affected individuals. Thrombosis is a severe complication of COVID‐19 that leads to a worse clinical course with higher rates of mortality. Multiple lines of evidence suggest that hyperinflammation plays a crucial role in disease progression. This review compiles clinical data of COVID‐19 patients who developed thrombotic complications to investigate the possible role of hyperinflammation in inducing hypercoagulation. A systematic literature search was performed using PubMed, Embase, Medline and Scopus to identify relevant clinical studies that investigated thrombotic manifestations and reported inflammatory and coagulation biomarkers in COVID‐19 patients. Only 54 studies met our inclusion criteria, the majority of which demonstrated significantly elevated inflammatory markers. In the cohort studies with control, D‐dimer was significantly higher in COVID‐19 patients with thrombosis as compared to the control. Pulmonary embolism, deep vein thrombosis and strokes were frequently reported which could be attributed to the hyperinflammatory response associated with COVID‐19 and/or to the direct viral activation of platelets and endothelial cells, two mechanisms that are discussed in this review. It is recommended that all admitted COVID‐19 patients should be assessed for hypercoagulation. Furthermore, several studies have suggested that anticoagulation may be beneficial, especially in hospitalized non‐ICU patients. Although vaccines against SARS‐CoV‐2 have been approved and distributed in several countries, research should continue in the field of prevention and treatment of COVID‐19 and its severe complications including thrombosis due to the emergence of new variants against which the efficacy of the vaccines is not yet clear.
Collapse
Affiliation(s)
- Sara Tomerak
- Weill Cornell Medicine QatarQatar Foundation Doha Qatar
| | - Safah Khan
- Weill Cornell Medicine QatarQatar Foundation Doha Qatar
| | - Muna Almasri
- Weill Cornell Medicine QatarQatar Foundation Doha Qatar
| | - Rawan Hussein
- Weill Cornell Medicine QatarQatar Foundation Doha Qatar
| | - Ali Abdelati
- Weill Cornell Medicine QatarQatar Foundation Doha Qatar
| | - Ahmed Aly
- Weill Cornell Medicine QatarQatar Foundation Doha Qatar
| | | | | | - Hiba Naveed
- Weill Cornell Medicine QatarQatar Foundation Doha Qatar
| | | | - Dalia Zakaria
- Weill Cornell Medicine QatarQatar Foundation Doha Qatar
| |
Collapse
|
218
|
Naqvi AR, Schwartz J, Brandini DA, Schaller S, Hussein H, Valverde A, Naqvi RA, Shukla D. COVID-19 and oral diseases: Assessing manifestations of a new pathogen in oral infections. Int Rev Immunol 2021; 41:423-437. [PMID: 34525891 DOI: 10.1080/08830185.2021.1967949] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022]
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is a recently identified virus responsible for life-threatening coronavirus disease 19 (COVID-19). The SARS-CoV-2 infected subjects can be asymptomatic or symptomatic; the later may present a wide spectrum of clinical manifestations. However, the impact of SARS-CoV-2 on oral diseases remain poorly studied. Detection of SARS-CoV-2 in saliva indicates existence of virus in the oral cavity. Recent studies demonstrating the expression of ACE-2, a SARS-CoV-2 entry receptor, in oral tissues further strengthens this observation. Cytokine storm in severe COVID-19 patients and copious secretion of pro-inflammatory cytokines (IL-6, IL-1β and TNF-α) in multiple symptomatic oral pathologies including periodontitis and periapical periodontitis suggests that inflammatory microenvironment is a hallmark of both COVID-19 and oral diseases. Hyperinflammation may provide conducive microenvironment for the growth of local oral pathogens or opportunistic microbes and exert detrimental impact on the oral tissue integrity. Multiple case reports have indicated uncharacterized oral lesions, symptomatic irreversible pulpitis, higher plaque index, necrotizing/desquamative gingivitis in COVID-19 patients suggesting that SARS-CoV-2 may worsen the manifestations of oral infections. However, the underlying factors and pathways remain elusive. Here we summarize current literature and suggest mechanisms for viral pathogenesis of oral dental pathology derived from oral microbiome and oral mucosa-dental tissue interactions. Longitudinal studies will reveal how the virus impairs disease progression and resolution post-therapy. Some relationships we suggest provide the basis for novel monitoring and treatment of oral viral disease in the era of SARS-CoV-2 pandemic, promoting evidence-based dentistry guidelines to diagnose virus-infected patients to improve oral health.
Collapse
Affiliation(s)
- Afsar R Naqvi
- Mucosal Immunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Joel Schwartz
- Molecular Pathology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daniela Atili Brandini
- Department of Diagnosis and Surgery, Araçatuba Dental School, Universidade Estadual Paulista/UNESP, Araçatuba, São Paulo, Brazil
| | - Samantha Schaller
- Mucosal Immunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Heba Hussein
- Department of Oral Medicine, Oral Diagnosis, and Periodontology, Faculty of Dentistry, Cairo University, Cairo, Egypt
| | - Araceli Valverde
- Mucosal Immunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Raza Ali Naqvi
- Mucosal Immunology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, Illinois, USA
| |
Collapse
|
219
|
Basic-Jukic N, Coric M, Bulimbasic S, Dika Z, Juric I, Furic-Cunko V, Katalinic L, Kos J, Fistrek M, Kastelan Z, Jelakovic B. Histopathologic findings on indication renal allograft biopsies after recovery from acute COVID-19. Clin Transplant 2021; 35:e14486. [PMID: 34532893 PMCID: PMC8646844 DOI: 10.1111/ctr.14486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/30/2022]
Abstract
Current knowledge on histopathological changes occurring after COVID‐19 in transplanted kidneys is limited. Herein, we present renal allograft pathology findings in patients recovered from COVID‐19. Six patients underwent indication biopsy, and one required allograft nephrectomy after acute COVID‐19. Demographic data, clinical characteristics, and laboratory findings were recorded. The histopathological analysis included light microscopy, immunostaining, and electron microscopy. Five patients were hospitalized for acute COVID‐19, and all were diagnosed with imaging‐confirmed pneumonia, one requiring mechanical ventilation, and two requiring dialysis. Two patients had mild form. Histopathologic examination of renal allograft specimens revealed collapsing, perihilar, tip‐lesion and secondary FSGS in one patient each. One patient had borderline acute cellular rejection, and two had chronic antibody‐mediated rejection. Histopathologic changes of glomerular tufts were accompanied by acute tubular injury in four patients. None of our patients had signs of viral inclusions in kidney cells. One patient died and one remained dialysis‐dependent after the good initial response to treatment. Patients with collapsing and perihilar FSGS had further progression of their chronic allograft nephropathy still without need for dialysis. In conclusion, diverse kidney pathology may be found in SARS‐CoV‐2–infected renal transplant patients. It seems that viral infection may affect the immune system with triggering of glomerular diseases, while the acute tubular injury is of multifactorial etiology. Direct viral effect is less likely.
Collapse
Affiliation(s)
- Nikolina Basic-Jukic
- Department of Nephrology, Arterial Hypertension and Dialysis, Klinički bolnički centar Zagreb, Zagreb, Croatia.,School of medicine, Sveučilište u Zagrebu Medicinski fakultet, Zagreb, Croatia
| | - Marijana Coric
- Department of pathology, Klinički bolnički centar Zagreb, Zagreb, Croatia
| | - Stela Bulimbasic
- Department of pathology, Klinički bolnički centar Zagreb, Zagreb, Croatia
| | - Zivka Dika
- Department of Nephrology, Arterial Hypertension and Dialysis, Klinički bolnički centar Zagreb, Zagreb, Croatia
| | - Ivana Juric
- Department of Nephrology, Arterial Hypertension and Dialysis, Klinički bolnički centar Zagreb, Zagreb, Croatia
| | - Vesna Furic-Cunko
- Department of Nephrology, Arterial Hypertension and Dialysis, Klinički bolnički centar Zagreb, Zagreb, Croatia
| | - Lea Katalinic
- Department of Nephrology, Arterial Hypertension and Dialysis, Klinički bolnički centar Zagreb, Zagreb, Croatia
| | - Jelena Kos
- Department of Nephrology, Arterial Hypertension and Dialysis, Klinički bolnički centar Zagreb, Zagreb, Croatia
| | - Margareta Fistrek
- Department of Nephrology, Arterial Hypertension and Dialysis, Klinički bolnički centar Zagreb, Zagreb, Croatia
| | - Zeljko Kastelan
- Department of Urology, University Hospital center Zagreb, Zagreb, Croatia
| | - Bojan Jelakovic
- Department of Nephrology, Arterial Hypertension and Dialysis, Klinički bolnički centar Zagreb, Zagreb, Croatia.,School of medicine, Sveučilište u Zagrebu Medicinski fakultet, Zagreb, Croatia
| |
Collapse
|
220
|
Saleh M, Alkofide A, Alshammari A, Siddiqui K, Owaidah T. Changes in Hematological, Clinical and Laboratory Parameters for Children with COVID-19: Single-Center Experience. J Blood Med 2021; 12:819-826. [PMID: 34512062 PMCID: PMC8427837 DOI: 10.2147/jbm.s321372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/06/2021] [Indexed: 12/23/2022] Open
Abstract
Background COVID-19 wreaked havoc on the healthcare system, with more than 36 million cases reported globally. Although the pediatric population makes up a lesser proportion of total COVID-19 patients than adults, the clinical status, age and comorbidities warrant identifying possible prognostic factors associated with disease severity in this group. The current study aimed to explore the incidence of thrombosis, overall outcome, and different hematological and coagulation markers in children with COVID-19. Methods This is a single-center prospective study of 43 patients (age < 14 years) with confirmed COVID-19 diagnosis recruited from April to August 2020. Data for clinical presentation were collected and analyzed. The samples were tested for different hematological and coagulation markers. Results Twenty-nine (67.4%) were symptomatic at presentation, with fever being the most common symptom (n = 23, 53.5%), followed by respiratory (n = 5, 11.6%) and gastrointestinal symptoms (n = 3, 7%). Co-morbid conditions were recorded in 26 (60.5%) patients, with malignancy being the commonest (n = 9, 20.9%). In this cohort of patients with age <14 years, hypertension, respiratory symptoms and ABO group-A were significantly associated with pediatric intensive care unit (PICU) admission during the course of treatment. Patients with elevated FVIII and fibrinogen levels at presentation were more likely to have an extended length of hospital stay (LOS) (P-value =0.036 and 0.032 respectively). No thrombotic event was observed in our cohort. D-dimer values were higher (above 0.5 µg/mL) in 24 (55.8%) patients at admission. We found an association between high D-dimer and PICU admission and LOS. Conclusion Although we did not observe thrombosis in our cohort, serial measurements of D-dimer and elevated FVIII bear a prognostic value in predicting the need for critical care in children with COVID-19. Further studies with larger sample size can aid in the establishment of prognostic factors for the pediatric COVID-19 population.
Collapse
Affiliation(s)
- Mahasen Saleh
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Amani Alkofide
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Anfal Alshammari
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Khawar Siddiqui
- Department of Pediatric Hematology Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Tarek Owaidah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Kingdom of Saudi Arabia.,Department of Pathology, Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| |
Collapse
|
221
|
Shibeeb S, Ahmad MN. Thrombotic and Hypercoagulability Complications of COVID-19: An Update. J Blood Med 2021; 12:785-793. [PMID: 34512059 PMCID: PMC8421041 DOI: 10.2147/jbm.s316014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/20/2021] [Indexed: 12/27/2022] Open
Abstract
The current COVID-19 pandemic emerged in December 2019, in China, affecting millions of people worldwide. COVID-19 is mainly a disease of the respiratory system, yet systematic complications have also been reported among SARS-CoV-2 infected patients. Thrombotic complications are one of the severe clinical outcomes of COVID-19, especially among critically ill patients, and are associated with poor prognosis. To date, many studies have concluded that COVID-19 increases the incidence of thrombotic events and coagulopathies; however, the exact mechanism behind such a disease outcome is not well known. Various pathophysiological mechanisms for thrombotic events in COVID-19 have been proposed, these include virus-induced endothelial cell damage, inflammation, and excess production of pro-inflammatory cytokines. As a result, most critically diseased COVID-19 patients are managed with prophylactic anticoagulant, yet some still develop thrombotic episodes. Therefore, better understanding of the mechanisms behind the thrombotic complications is needed to develop treatments that specifically target such pathways, which may aid in better disease management and improve the prognosis.
Collapse
Affiliation(s)
- Sapha Shibeeb
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Muneera Naseer Ahmad
- Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
222
|
Al Otair H, AlSaleh K, AlQahtany FS, Al Ayed K, Al Ammar H, Al Mefgai N, Al Zeer F. The Level of vWF Antigen and Coagulation Markers in Hospitalized Patients with Covid-19. J Blood Med 2021; 12:809-817. [PMID: 34512061 PMCID: PMC8416187 DOI: 10.2147/jbm.s318940] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 06/22/2021] [Indexed: 12/25/2022] Open
Abstract
Background The coagulopathy of COVID-19 still awaits more clarification, and one approach that has not been investigated is to compare the hemostatic changes between COVID-19 and non-COVID-19 infected patients. Objective This study aims to study COVID-19 coagulopathy by measuring markers of endothelial injury and coagulation, including anticoagulants (TFPI, protein C, protein S, and AT) in COVID-19 patients and compare them with non-COVID-19 patients early in the course of the disease. Methodology This is an observational, prospective cross-sectional study comparing the levels of protein C, protein S, antithrombin (AT) III, clotting factor (F) VIII, von Willebrand factor (vWF) and coagulation screening tests (PT and a PTT), fibrinogen, D-dimer in COVID-19 patients admitted during the same time with non-COVID-19 infections. The demographic and clinical data of the patients were collected from electronic medical records during admission. Blood tests were extracted within 24 hours of admission for both groups. Results Fifty-four (66.7% males) consecutive COVID-19 patients and 24 (59% males) non-COVID-19 controls were enrolled in the study from October 2020 till December 2020. COVID-19 patients were significantly older than non-COVID-19 (57.7±14.2 vs 50±19.8 years, p=0.005). Fibrinogen level was significantly higher in COVID-19 patients compared to controls (5.9±1.48 vs 3.9±1.57, p<0.001). There was no statistically significant difference in the level of FVIII, protein C, S, ATIII, and D-dimer between the two groups. The level of vWF Ag was statistically higher in COVID-19 patients (276.7±91.1 vs 184.7±89.4, p=0.0001). There was significant thrombocytopenia and lymphopenia among COVID-19 patients. Inflammatory markers, CRP, ferritin, and LDH, were increased in COVID-19 patients compared to non-COVID-19, but the difference was not statistically significant. High fibrinogen and vWF AG levels were the two independent variables found in COVID-19 patients. Conclusion The level of vWF Ag is increased early in the course of COVID-19 infection. This can be used as a biomarker for endothelial injury, which is peculiar to COVID-19 infection.
Collapse
Affiliation(s)
- Hadeel Al Otair
- Department of Medicine, King Saud University Medical city (KSUMC), King Saud University, Riyadh, Saudi Arabia
| | - Khalid AlSaleh
- Department of Medicine, Hematology/ oncology division, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fatmah S AlQahtany
- Department of Pathology, Hematopathology Unit, College of Medicine, King Saud University, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Khalid Al Ayed
- Department of Medicine, King Saud University Medical city (KSUMC), King Saud University, Riyadh, Saudi Arabia
| | - Hessah Al Ammar
- Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Noura Al Mefgai
- Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Faisal Al Zeer
- College of Medicine, King Saud University, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
223
|
Bobescu E, Marceanu LG, Covaciu A, Vladau LA. Thrombosis, an important piece in the COVID-19 puzzle: From pathophysiology to therapy. Anatol J Cardiol 2021; 25:601-608. [PMID: 34498590 DOI: 10.5152/anatoljcardiol.2021.475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A lot of data about coronavirus disease 2019 (COVID-19) have been already published; however, these still form only a part of the pandemic puzzle. Once we have all the pieces of the puzzle, we will be able to successfully treat this disease with its multiple challenges. COVID-19 has a high thrombogenic potential. In this study, we aimed to review published data about COVID-19 associated thrombosis from pathophysiology to treatment and the role in patient evolution. We searched for articles and studies published online through MEDLINE/PubMed database, Google Scholar, ScienceDirect, Wiley Online Library, and Nature Public Health Emergency Collection. We found numerous articles regarding COVID-19 infection but selected only those focused on thrombosis. D-dimers have a predictive value in identifying thrombosis and a high level correlates with the severity of the infection and death. Most patients who were on chronic anticoagulant therapy before contracting the virus had a better prognosis. Heparin has other favorable effects such as a direct antiviral and anti-inflammatory effect in addition to its anticoagulant effect. COVID-19 infections are frequently complicated by thrombotic pathology. High plasma level of D-dimers is a predictive factor for severe prognosis, and the recommended anticoagulant, associated with low mortality, is heparin followed by a direct oral anticoagulant. Randomized studies in large groups of patients and therapeutic guidelines are still needed on this subject.
Collapse
Affiliation(s)
- Elena Bobescu
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov; Brasov- Romania;Department of Cardiology, Clinical County Emergency Hospital Brasov; Brasov-Romania
| | - Luigi Geo Marceanu
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov; Brasov- Romania
| | - Alexandru Covaciu
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov; Brasov- Romania;Department of Cardiology, Clinical County Emergency Hospital Brasov; Brasov-Romania
| | - Larisa Alexandra Vladau
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov; Brasov- Romania;Department of Cardiology, Clinical County Emergency Hospital Brasov; Brasov-Romania
| |
Collapse
|
224
|
Sharma V, Singh AP, Singh AP. Therapeutic approaches in COVID-19 followed before arrival of any vaccine. MATERIALS TODAY. PROCEEDINGS 2021; 48:1258-1264. [PMID: 34493973 PMCID: PMC8413457 DOI: 10.1016/j.matpr.2021.08.265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/02/2021] [Accepted: 08/23/2021] [Indexed: 01/08/2023]
Abstract
In present times covid-19 is spreading and is showing very destructive effects. It does not only affected the physical health but mental health as well as the economy of the major affected countries. Corona viruses are group of related RNA viruses. The first case of this virus was observed in China and then this virus got spread in the many countries and different strategies were made to stop the spread of this virus. Since no particular vaccine was available to cure this so different strategies were made. Due to the emergence of pandemic diseases, drug development and control strategies have been re-examined. One of the most important factors that scientists have to consider is the effectiveness of their drugs. This virus causes the respiratory tract infections which can range from mild to lethal. COVID-19 is a major cause of death in advanced countries. It is due to the absence of any particular vaccine that can effectively treat this condition. So in this review we will discuss about the therapeutic approaches followed to combat this deadly virus. Ayurveda, nitric oxide, nanoparticles and enzymes played a very important role in boosting the immunity and treatment of corona. Many herbs and some tips of using a combination of herbs proved to be very efficient while facing problems in breathing. Giving the dose of nitric oxide at some particular level and chloroquine the drug showed the antiviral activity against the virus. Developing methods to identify and contain COVID-19 is essential to successfully manage the virus. Various strains of the SARS-COV-2 were detected and were found more dangerous. The therapeutic approaches followed actually were efficient and can be used to combat the other variants also. This review focuses on the latest developments in the field of therapeutics and the strategies which were followed before any vaccine.
Collapse
Affiliation(s)
- Vasundhra Sharma
- Department of Chemistry, Chandigarh University, Mohali, Punjab, India
| | - Atul Pratap Singh
- Department of Chemistry, Chandigarh University, Mohali, Punjab, India
| | | |
Collapse
|
225
|
Abacioglu OO, Yildirim A. The ATRIA score is superior to the m-CHA2DS2-Vasc score in predicting in-hospital mortality in COVID-19. ACTA ACUST UNITED AC 2021; 67:443-448. [PMID: 34468612 DOI: 10.1590/1806-9282.20200983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Coronavirus disease 2019 (COVID-19) has become a health and social problem all over the world. Most of the deaths occur from embolism and thrombus formation. We aimed to compare the predictive value of the anticoagulation and risk factors in atrial fibrillation (ATRIA) and m-CHA2DS2-Vasc scores in in-hospital mortality in COVID-19. METHODS Three-hundred and ninety-four patients who were hospitalized due to COVID-19 between 10 June 2020 and 10 September 2020 were included. Three-hundred and sixty patients who survived were defined as the non-mortality group and the remaining 34 whose hospitalizations resulted in death were defined as the mortality group. The anticoagulation and risk factors in atrial fibrillation and m-CHA2DS2-Vasc scores of the patients were calculated. RESULTS A total of 394 patients, mean age 66.2±9.7 (221 male [56.1%]) were included in this retrospective study. The median values of the anticoagulation and risk factors in atrial fibrillation and m-CHA2DS2-Vasc scores were different between the groups (p<0.000 for both). The multivariate logistic regression analysis showed that both the m-CHA2DS2-Vasc and anticoagulation and risk factors in atrial fibrillation scores were independent predictors of in-hospital mortality (p=0.024, 95%CI 1.039-1.704 for anticoagulation and risk factors in atrial fibrillation and p=0.043, 95%CI 1.012-2.088 for m-CHA2DS2-Vasc). In the receiver operating characteristic curve analysis, the anticoagulation and risk factors in atrial fibrillation score was superior to the m-CHA2DS2-Vasc score with an AUC 0.774 and SE:0.037, and p<0.001. CONCLUSIONS In our study, we showed that the anticoagulation and risk factors in atrial fibrillation and m-CHA2DS2-Vasc scores can be used as predictors of thrombosis and mortality in COVID-19 patients. In addition, the predictive value of the anticoagulation and risk factors in atrial fibrillation score was higher than that of m-CHA2DS2-Vasc. The use of the anticoagulation and risk factors in atrial fibrillation score to assess high-risk patients in COVID-19 may be recommended.
Collapse
Affiliation(s)
| | - Arafat Yildirim
- Adana City Training and Research Hospital, Cardiology - Adana, Turkey
| |
Collapse
|
226
|
A Case of Post-COVID-19-Associated Paracentral Acute Middle Maculopathy and Giant Cell Arteritis-Like Vasculitis. J Neuroophthalmol 2021; 41:351-355. [PMID: 34415268 DOI: 10.1097/wno.0000000000001348] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
ABSTRACT A 47-year-old man with a history of COVID-19 infection 2 months before presentation, presented with a scotoma of the paracentral visual field of the right eye. After thorough testing and evaluation, a diagnosis of paracentral acute middle maculopathy (PAMM) was established. Two months later, the patient developed temporal headache and jaw claudication. High-dose steroids were initiated, and workup for giant cell arteritis (GCA) was undertaken. The patient experienced resolution of the symptoms within 24 hours of steroid initiation. ESR, CRP, and temporal artery biopsy results were normal, although all were obtained more than 2 weeks after steroid initiation. To the best of our knowledge, our patient represents the first individual to date to potentially implicate COVID-19 in both small and large vessel vasculitis in the ophthalmic setting.
Collapse
|
227
|
Seth S, Rashid F, Khera K. An overview of the COVID-19 complications in paediatric population: A pandemic dilemma. Int J Clin Pract 2021; 75:e14494. [PMID: 34115913 PMCID: PMC8420266 DOI: 10.1111/ijcp.14494] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
AIM The primary objective of this article is to understand the various complications caused by the coronavirus in the paediatric population. METHOD An electronic search was conducted using PubMed and incorporated forward and backward research methods on clinical trials, case reports, case series, guidelines and reports from the centre for disease control and prevention (CDC), and the keywords included COVID-19, paediatrics, multisystem inflammatory syndrome in children (MIS-C), complications, acute kidney injury and heart failure. Secondary resources included one study from preprint servers (www.preprints.org), last search 8 May 2021, with notion of nonpeer review status. Data were collected and analysed to stay current with the most recent alerts and guidelines for the best care for children during the COVID-19 pandemic. RESULTS Evaluation and analysis of literature revealed MIS-C to be the most prevalent followed by neurological complications. Whereas the least prevalent were septic shock and ophthalmic complications. CONCLUSION Even though COVID-19 is known to be a less severe in the paediatric population, the complications of the virus have caused a great deal of stress to the paediatric patients' parents and paediatricians worldwide, and hence, emphasis should be given to the management of coronavirus complications in paediatrics.
Collapse
Affiliation(s)
- Shrey Seth
- Department of Pharmacy PracticeManipal College of Pharmaceutical Sciences, Manipal Academy of Higher EducationManipalIndia
| | - Femida Rashid
- Department of Pharmacy PracticeManipal College of Pharmaceutical Sciences, Manipal Academy of Higher EducationManipalIndia
| | - Kanav Khera
- Department of Pharmacy PracticeManipal College of Pharmaceutical Sciences, Manipal Academy of Higher EducationManipalIndia
| |
Collapse
|
228
|
Santosa YP, Yuwono A. Two Different Clinical Presentations of Acute Limb Ischemia Caused by Acute Thrombotic Events in COVID-19. Cureus 2021; 13:e17916. [PMID: 34660110 PMCID: PMC8511142 DOI: 10.7759/cureus.17916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2021] [Indexed: 01/15/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Coagulopathy is frequently found in severe cases of COVID-19 and is usually manifested as a prothrombotic state. Hyperinflammation, endotheliitis, and immobilization during illness are hypothesized to play a role. Acute limb ischemia (ALI) is one of the presentations of arterial thrombosis in COVID-19. We present two cases of middle-aged men with COVID-19 infection, who developed ALI. The first patient developed ALI after 16 days from the initial COVID-19 diagnosis, and the second patient was admitted to the emergency ward due to sudden discoloration of his right lower limb, and COVID-19 was diagnosed during the evaluation.
Collapse
Affiliation(s)
- Yudistira P Santosa
- Department of Internal Medicine, Atma Jaya Catholic University of Indonesia, Jakarta, IDN
| | - Angelina Yuwono
- Department of Internal Medicine, Atma Jaya Catholic University of Indonesia, Jakarta, IDN
| |
Collapse
|
229
|
Jing W, Procko E. ACE2-based decoy receptors for SARS coronavirus 2. Proteins 2021; 89:1065-1078. [PMID: 33973262 PMCID: PMC8242511 DOI: 10.1002/prot.26140] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/16/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
SARS coronavirus 2 is neutralized by proteins that block receptor-binding sites on spikes that project from the viral envelope. In particular, substantial research investment has advanced monoclonal antibody therapies to the clinic where they have shown partial efficacy in reducing viral burden and hospitalization. An alternative is to use the host entry receptor, angiotensin-converting enzyme 2 (ACE2), as a soluble decoy that broadly blocks SARS-associated coronaviruses with limited potential for viral escape. Here, we summarize efforts to engineer higher affinity variants of soluble ACE2 that rival the potency of affinity-matured antibodies. Strategies have also been used to increase the valency of ACE2 decoys for avid spike interactions and to improve pharmacokinetics via IgG fusions. Finally, the intrinsic catalytic activity of ACE2 for the turnover of the vasoconstrictor angiotensin II may directly address COVID-19 symptoms and protect against lung and cardiovascular injury, conferring dual mechanisms of action unachievable by monoclonal antibodies. Soluble ACE2 derivatives therefore have the potential to be next generation therapeutics for addressing the immediate needs of the current pandemic and possible future outbreaks.
Collapse
Affiliation(s)
- Wenyang Jing
- Center for Biophysics and Quantitative BiologyUniversity of IllinoisUrbanaIllinoisUSA
| | - Erik Procko
- Center for Biophysics and Quantitative BiologyUniversity of IllinoisUrbanaIllinoisUSA
- Department of Biochemistry and Cancer Center at IllinoisUniversity of IllinoisUrbanaIllinoisUSA
| |
Collapse
|
230
|
Kohansal Vajari M, Shirin M, Pourbagheri‐Sigaroodi A, Akbari ME, Abolghasemi H, Bashash D. COVID-19-related coagulopathy: A review of pathophysiology and pharmaceutical management. Cell Biol Int 2021; 45:1832-1850. [PMID: 33945651 PMCID: PMC8239905 DOI: 10.1002/cbin.11623] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/20/2021] [Accepted: 05/01/2021] [Indexed: 01/08/2023]
Abstract
December 2019 will never be forgotten in the history of medicine when an outbreak of pneumonia of unknown etiology in Wuhan, China sooner or later prompted the World Health Organization to issue a public health warning emergency. This is not the first nor will it be the last time that a member of β-coronaviruses (CoVs) is waging a full-scale war against human health. Notwithstanding the fact that pneumonia is the primary symptom of the novel coronavirus (2019nCoV; designated as SARS-CoV-2), the emergence of severe disease mainly due to the injury of nonpulmonary organs at the shadow of coagulopathy leaves no choice, in some cases, rather than a dreadful death. Multiple casual factors such as inflammation, endothelial dysfunction, platelet and complement activation, renin-angiotensin-aldosterone system derangement, and hypoxemia play a major role in the pathogenesis of coagulopathy in coronavirus disease 2019 (COVID-19) patients. Due to the undeniable role of coagulation dysfunction in the initiation of several complications, assessment of coagulation parameters and the platelet count would be beneficial in early diagnosis and also timely prediction of disease severity. Although low-molecular-weight heparin is considered as the first-line of treatment in COVID-19-associated coagulopathy, several possible therapeutic options have also been proposed for better management of the disease. In conclusion, this review would help us to gain insight into the pathogenesis, clinical manifestation, and laboratory findings associated with COVID-19 coagulopathy and would summarize management strategies to alleviate coagulopathy-related complications.
Collapse
Affiliation(s)
- Mahdi Kohansal Vajari
- Department of Hematology and Blood Banking, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Mahsa Shirin
- Department of Hematology and Blood Banking, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Atieh Pourbagheri‐Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Mohammad Esmaeil Akbari
- Cancer Research Center, Department of General SurgeryShahid Beheshti University of Medical SciencesTehranIran
| | - Hassan Abolghasemi
- Pediatric Congenital Hematologic Disorders Research Center, Mofid HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
231
|
Tokarek T, Dziewierz A, Malinowski KP, Rakowski T, Bartuś S, Dudek D, Siudak Z. Treatment Delay and Clinical Outcomes in Patients with ST-Segment Elevation Myocardial Infarction during the COVID-19 Pandemic. J Clin Med 2021; 10:jcm10173920. [PMID: 34501369 PMCID: PMC8432080 DOI: 10.3390/jcm10173920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 02/02/2023] Open
Abstract
Pandemic-specific protocols require additional time to prepare medical staff and catheterization laboratories. Thus, we sought to investigate treatment delay and clinical outcomes in COVID-19 positive and negative patients with ST-segment elevation myocardial infarction (STEMI) treated with percutaneous coronary intervention (PCI) during on- and off-hours. All consecutive patients with STEMI treated with PCI between 1 March and 31 December 2020 were enrolled in the analysis. A propensity score match was used to compare COVID-19 positive and negative patients for on- and off-hours. The study group was comprised of 877 paired patients treated during regular hours (every day 7:00 a.m. to 16:59 p.m.) and 418 matched pairs with PCI performed during off-hours (every day 17:00 p.m. to 06:59 a.m.) (ORPKI Polish National Registry). No difference in periprocedural mortality was observed between the two groups (on-hours: COVID-19 negative vs. COVID-19 positive: 17 (1.9%) vs. 11 (1.3%); p = 0.3; off-hours: COVID-19 negative vs. COVID-19 positive: 4 (1.0%) vs. 7 (1.7%); p = 0.5). Additionally, a similar rate of periprocedural complications was reported. Patients diagnosed with COVID-19 were exposed to longer time from first medical contact to angiography (on-hours: 133.8 (±137.1) vs. 117.1 (±135.8) (min); p = 0.001) (off-hours: 148.1 (±201.6) vs. 112.2 (±138.7) (min); p = 0.003). However, there was no influence of COVID-19 diagnosis on mortality and the prevalence of other periprocedural complications irrespective of time of intervention.
Collapse
Affiliation(s)
- Tomasz Tokarek
- Department of Cardiology and Cardiovascular Interventions, University Hospital, 2 Jakubowskiego St., 30-688 Krakow, Poland; (A.D.); (T.R.); (S.B.)
- Center for Intensive Care and Perioperative Medicine, Jagiellonian University Medical College, 31-008 Krakow, Poland
- Correspondence: ; Tel.: +48-12-400-22-62
| | - Artur Dziewierz
- Department of Cardiology and Cardiovascular Interventions, University Hospital, 2 Jakubowskiego St., 30-688 Krakow, Poland; (A.D.); (T.R.); (S.B.)
- 2nd Department of Cardiology, Jagiellonian University Medical College, 31-008 Krakow, Poland; (K.P.M.); (D.D.)
| | - Krzysztof Piotr Malinowski
- 2nd Department of Cardiology, Jagiellonian University Medical College, 31-008 Krakow, Poland; (K.P.M.); (D.D.)
| | - Tomasz Rakowski
- Department of Cardiology and Cardiovascular Interventions, University Hospital, 2 Jakubowskiego St., 30-688 Krakow, Poland; (A.D.); (T.R.); (S.B.)
- 2nd Department of Cardiology, Jagiellonian University Medical College, 31-008 Krakow, Poland; (K.P.M.); (D.D.)
| | - Stanisław Bartuś
- Department of Cardiology and Cardiovascular Interventions, University Hospital, 2 Jakubowskiego St., 30-688 Krakow, Poland; (A.D.); (T.R.); (S.B.)
- 2nd Department of Cardiology, Jagiellonian University Medical College, 31-008 Krakow, Poland; (K.P.M.); (D.D.)
| | - Dariusz Dudek
- 2nd Department of Cardiology, Jagiellonian University Medical College, 31-008 Krakow, Poland; (K.P.M.); (D.D.)
| | - Zbigniew Siudak
- Faculty of Medicine and Health Science, Jan Kochanowski University, 25-369 Kielce, Poland;
| |
Collapse
|
232
|
Liu D, Zeng X, Ding Z, Lv F, Mehta JL, Wang X. Adverse Cardiovascular Effects of Anti-COVID-19 Drugs. Front Pharmacol 2021; 12:699949. [PMID: 34512335 PMCID: PMC8424204 DOI: 10.3389/fphar.2021.699949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or COVID-19 infection is the cause of the ongoing global pandemic. Mortality from COVID-19 infection is particularly high in patients with cardiovascular diseases. In addition, COVID-19 patients with preexisting cardiovascular comorbidities have a higher risk of death. Main cardiovascular complications of COVID-19 are myocardial infarction, myocarditis, acute myocardial injury, arrhythmias, heart failure, stroke, and venous thromboembolism. Therapeutic interventions in terms of drugs for COVID-19 have many cardiac adverse effects. Here, we review the relative therapeutic efficacy and adverse effects of anti-COVID-19 drugs.
Collapse
Affiliation(s)
- Dongling Liu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xiang Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, China
| | - Zufeng Ding
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States
| | - Fenghua Lv
- Department of Cardiology, Xinxiang Medical University First Affiliated Hospital, Weihui, China
| | - Jawahar L. Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, United States
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
- Department of Cardiology, Xinxiang Medical University First Affiliated Hospital, Weihui, China
| |
Collapse
|
233
|
Association Between Novel Coronavirus Disease 2019 (COVID-19) and Idiopathic Intracranial Hypertension. ARCHIVES OF PEDIATRIC INFECTIOUS DISEASES 2021. [DOI: 10.5812/pedinfect.115171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction: Idiopathic intracranial hypertension (IIH), also known as pseudotumor cerebri, is a rare neurological manifestation of coronavirus disease 2019 (COVID-19) infection. Case Presentation: In this case study, we present a 10-year-old girl with headache, diplopia, bilateral sixth nerve palsy, and papilledema following a history of seven days of fever about two weeks ago. In lumbar puncture, the opening pressure was 56 cmH2O. Cerebrospinal fluid analysis was normal. Brain magnetic resonance imaging (MRI) was suggestive of IIH. Nasopharyngeal reverse transcription-polymerase chain reaction (RT-PCR) was positive for COVID-19. Oral acetazolamide was prescribed, and the patient improved completely after three weeks of continuous medical treatment. Conclusions: Since IIH is a rare neurological complication of COVID-19, we have to suspect COVID-19 infection in any patient with IIH.
Collapse
|
234
|
Valle EDO, Cabrera CPS, Albuquerque CCCD, Silva GVD, Oliveira MFAD, Sales GTM, Smolentzov I, Reichert BV, Andrade L, Seabra VF, Lins PRG, Rodrigues CE. Continuous renal replacement therapy in COVID-19-associated AKI: adding heparin to citrate to extend filter life-a retrospective cohort study. Crit Care 2021; 25:299. [PMID: 34412667 PMCID: PMC8375288 DOI: 10.1186/s13054-021-03729-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/12/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) may predispose patients to thrombotic events. The best anticoagulation strategy for continuous renal replacement therapy (CRRT) in such patients is still under debate. The purpose of this study was to evaluate the impact that different anticoagulation protocols have on filter clotting risk. METHODS This was a retrospective observational study comparing two different anticoagulation strategies (citrate only and citrate plus intravenous infusion of unfractionated heparin) in patients with acute kidney injury (AKI), associated or not with COVID-19 (COV + AKI and COV - AKI, respectively), who were submitted to CRRT. Filter clotting risks were compared among groups. RESULTS Between January 2019 and July 2020, 238 patients were evaluated: 188 in the COV + AKI group and 50 in the COV - AKI group. Filter clotting during the first filter use occurred in 111 patients (46.6%). Heparin use conferred protection against filter clotting (HR = 0.37, 95% CI 0.25-0.55), resulting in longer filter survival. Bleeding events and the need for blood transfusion were similar between the citrate only and citrate plus unfractionated heparin strategies. In-hospital mortality was higher among the COV + AKI patients than among the COV - AKI patients, although it was similar between the COV + AKI patients who received heparin and those who did not. Filter clotting was more common in patients with D-dimer levels above the median (5990 ng/ml). In the multivariate analysis, heparin was associated with a lower risk of filter clotting (HR = 0.28, 95% CI 0.18-0.43), whereas an elevated D-dimer level and high hemoglobin were found to be risk factors for circuit clotting. A diagnosis of COVID-19 was marginally associated with an increased risk of circuit clotting (HR = 2.15, 95% CI 0.99-4.68). CONCLUSIONS In COV + AKI patients, adding systemic heparin to standard regional citrate anticoagulation may prolong CRRT filter patency by reducing clotting risk with a low risk of complications.
Collapse
Affiliation(s)
- Eduardo de Oliveira Valle
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Carla Paulina Sandoval Cabrera
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Claudia Coimbra César de Albuquerque
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Giovanio Vieira da Silva
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Márcia Fernanda Arantes de Oliveira
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Gabriel Teixeira Montezuma Sales
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Igor Smolentzov
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Bernardo Vergara Reichert
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Lucia Andrade
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Victor Faria Seabra
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Paulo Ricardo Gessolo Lins
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil
| | - Camila Eleuterio Rodrigues
- Hospital das Clínicas, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º andar, sala 3310, São Paulo, SP, CEP 01246-903, Brazil.
| |
Collapse
|
235
|
Sen S, Kannan NB, Kumar J, Rajan RP, Kumar K, Baliga G, Reddy H, Upadhyay A, Ramasamy K. Retinal manifestations in patients with SARS-CoV-2 infection and pathogenetic implications: a systematic review. Int Ophthalmol 2021; 42:323-336. [PMID: 34379290 PMCID: PMC8356207 DOI: 10.1007/s10792-021-01996-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
Introduction The pandemic of COVID-19 has been caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. Apart from respiratory malfunction, COVID-19 causes a system-wide thromboembolic state, leading to serious cardiovascular, cerebrovascular and peripheral vascular manifestations. However, our knowledge regarding retinal manifestations due to systemic COVID-19 is minimal. This systematic review has comprehensively summarized all retinal manifestations secondary to COVID-19 disease recorded till date since the beginning of the pandemic. Methods All studies published till November 27, 2020, which have reported retinal manifestations in COVID-19 patients were systematically reviewed using the PRISMA statement. Results We included 15 articles: 11 case reports and four cross-sectional case series. The most commonly reported manifestations which did not affect visual acuity were retinal hemorrhages and cotton wool spots. The most common vision threatening manifestation was retinal vein occlusion with associated macular edema. Rarely, patients may also present with retinal arterial occlusions and ocular inflammation. These manifestations may occur from as soon as within a week after the onset of COVID-19 symptoms to more than 6 weeks after. Conclusion Mostly causing milder disease, COVID-19 may however lead to severe life-threatening thromboembolic complications, and systemic antithrombotic therapy has been suggested as a prophylactic and therapeutic management strategy for patients affected with serious systemic disease. However, both sick and apparently healthy patients may suffer from various retinal complications which may lead to loss of vision as well. No consensus regarding management of retinal complications with anticoagulants or anti-inflammatory medications have been proposed; however, they may be tackled on individual basis.
Collapse
Affiliation(s)
- Sagnik Sen
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, India.
| | | | - Jayant Kumar
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, India
| | - Renu P Rajan
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, India
| | - Karthik Kumar
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, India
| | - Girish Baliga
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, India
| | | | - Anubhav Upadhyay
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, India
| | - Kim Ramasamy
- Department of Retina and Vitreous, Aravind Eye Hospital, Madurai, India
| |
Collapse
|
236
|
Becker RC, Sexton T, Smyth S. COVID-19 and biomarkers of thrombosis: focus on von Willebrand factor and extracellular vesicles. J Thromb Thrombolysis 2021; 52:1010-1019. [PMID: 34350541 PMCID: PMC8336902 DOI: 10.1007/s11239-021-02544-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/15/2021] [Indexed: 12/19/2022]
Abstract
COVID-19, caused by the SARS-CoV-2 virus, is responsible for a pandemic of unparalleled portion over the past century. While the acute phase of infection causes significant morbidity and mortality, post-acute sequelae that can affect essentially any organ system is rapidly taking on an equally large part of the overall impact on human health, quality of life, attempts to return to normalcy and the global economy. Herein, we summarize the potential role of von Willebrand Factor and extracellular vesicles toward understanding the pathophysiology, clinical presentation, duration of illness, diagnostic approach and management of COVID-19 and its sequelae.
Collapse
Affiliation(s)
- Richard C Becker
- Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267, USA.
| | - Travis Sexton
- The Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY, USA
| | - Susan Smyth
- University of Arkansas for Medical Sciences, Little Rock, AK, USA
| |
Collapse
|
237
|
Burgos-Blasco B, Güemes-Villahoz N, Vidal-Villegas B, Garcia-Feijoo J, Donate-Lopez J, Martin-Sanchez FJ, Gonzalez-Armengol JJ, Mendez-Hernandez CD. Optic Nerve Head Vessel Density Assessment in Recovered COVID-19 Patients: A Prospective Study Using Optical Coherence Tomography Angiography. J Glaucoma 2021; 30:711-717. [PMID: 33927148 PMCID: PMC8366516 DOI: 10.1097/ijg.0000000000001858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/06/2021] [Indexed: 01/08/2023]
Abstract
PRECIS Vascular diseases have been linked to alterations in optic nerve head perfusion. PURPOSE The main objective was to investigate the changes in peripapillary vessel density (VD) in post coronavirus disease (COVID-19) patients. METHODS In this prospective pilot exploratory study, patients with COVID-19 that were attended in the Emergency Department of Hospital Clinico San Carlos (Madrid) were included. All patients underwent optic nerve head optical coherence tomography angiography using the Cirrus HD-OCT 500 with AngioPlex OCTA (Zeiss, Dublin, CA) 4 and 12 weeks after diagnosis by positive reverse transcriptase-polymerase chain reaction test from nasopharyngeal swab at the Emergency Department. Sociodemographic data, medical history, disease severity, and laboratory work-up were collected. RESULTS One hundred and eighty eyes of 90 patients with severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection were included. None of the patients reported visual changes. Severe patients were older, more frequently hispanic, dyslipidemic, and presented lower lymphocytes counts, as well as increased ferritin, D-dimer, fibrinogen, and international normalized ratio levels. No changes in optic nerve head vascularization were observed when both visits were compared. No correlation was found between VD and clinical parameters, disease severity and laboratory work-up. CONCLUSIONS Changes to peripapillary VD were not observed in patients with COVID-19 in the early months following diagnosis.
Collapse
Affiliation(s)
- Barbara Burgos-Blasco
- Department of Ophthalmology, Instituto de investigación sanitaria del Hospital Clínico San Carlos (IsISSC), Instituto de Investigaciones Oftalmológicas Ramón Castroviejo
- Departamento de Inmunología, Oftalmología y ORL, Facultad de Medicina, Universidad Complutense de Madrid, ISCIII (OFTARED)
| | - Noemi Güemes-Villahoz
- Department of Ophthalmology, Instituto de investigación sanitaria del Hospital Clínico San Carlos (IsISSC), Instituto de Investigaciones Oftalmológicas Ramón Castroviejo
- Departamento de Inmunología, Oftalmología y ORL, Facultad de Medicina, Universidad Complutense de Madrid, ISCIII (OFTARED)
| | - Beatriz Vidal-Villegas
- Department of Ophthalmology, Instituto de investigación sanitaria del Hospital Clínico San Carlos (IsISSC), Instituto de Investigaciones Oftalmológicas Ramón Castroviejo
- Departamento de Inmunología, Oftalmología y ORL, Facultad de Medicina, Universidad Complutense de Madrid, ISCIII (OFTARED)
| | - Julian Garcia-Feijoo
- Department of Ophthalmology, Instituto de investigación sanitaria del Hospital Clínico San Carlos (IsISSC), Instituto de Investigaciones Oftalmológicas Ramón Castroviejo
- Departamento de Inmunología, Oftalmología y ORL, Facultad de Medicina, Universidad Complutense de Madrid, ISCIII (OFTARED)
| | - Juan Donate-Lopez
- Department of Ophthalmology, Instituto de investigación sanitaria del Hospital Clínico San Carlos (IsISSC), Instituto de Investigaciones Oftalmológicas Ramón Castroviejo
- Departamento de Inmunología, Oftalmología y ORL, Facultad de Medicina, Universidad Complutense de Madrid, ISCIII (OFTARED)
| | - Francisco J. Martin-Sanchez
- Emergency Department, Hospital Clínico San Carlos, Instituto de investigación sanitaria del Hospital Clínico San Carlos (IsISCC), Madrid, Spain
| | - Juan J. Gonzalez-Armengol
- Emergency Department, Hospital Clínico San Carlos, Instituto de investigación sanitaria del Hospital Clínico San Carlos (IsISCC), Madrid, Spain
| | - Carmen D. Mendez-Hernandez
- Department of Ophthalmology, Instituto de investigación sanitaria del Hospital Clínico San Carlos (IsISSC), Instituto de Investigaciones Oftalmológicas Ramón Castroviejo
- Departamento de Inmunología, Oftalmología y ORL, Facultad de Medicina, Universidad Complutense de Madrid, ISCIII (OFTARED)
| |
Collapse
|
238
|
Kurian SJ, Unnikrishnan MK, Miraj SS, Bagchi D, Banerjee M, Reddy BS, Rodrigues GS, Manu MK, Saravu K, Mukhopadhyay C, Rao M. Probiotics in Prevention and Treatment of COVID-19: Current Perspective and Future Prospects. Arch Med Res 2021; 52:582-594. [PMID: 33785208 PMCID: PMC7972717 DOI: 10.1016/j.arcmed.2021.03.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/02/2021] [Accepted: 03/12/2021] [Indexed: 02/06/2023]
Abstract
Saving lives and flattening the curve are the foremost priorities during the ongoing pandemic spread of SARS-CoV-2. Developing cutting-edge technology and collating available evidence would support frontline health teams. Nutritional adequacy improves general health and immunity to prevent and assuage infections. This review aims to outline the potential role of probiotics in fighting the COVID-19 by covering recent evidence on the association between microbiota, probiotics, and COVID-19, the role of probiotics as an immune-modulator and antiviral agent. The high basic reproduction number (R0) of SARS-CoV-2, absence of conclusive remedies, and the pleiotropic effect of probiotics in fighting influenza and other coronaviruses together favour probiotics supplements. However, further support from preclinical and clinical studies and reviews outlining the role of probiotics in COVID-19 are critical. Results are awaited from many ongoing clinical trials investigating the benefits of probiotics in COVID-19.
Collapse
Affiliation(s)
- Shilia Jacob Kurian
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India; Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | - Sonal Sekhar Miraj
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India; Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| | - Debasis Bagchi
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, USA
| | - Mithu Banerjee
- Department of Biochemistry, All India Institute of Medical Sciences,Jodhpur, Rajasthan, India
| | - B Shrikar Reddy
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Gabriel Sunil Rodrigues
- Department of Surgery, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mohan K Manu
- Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India; Department of Respiratory Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kavitha Saravu
- Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India; Department of Infectious Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Chiranjay Mukhopadhyay
- Department of Microbiology and Center for Emerging and Tropical Diseases, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
239
|
Mohamed M, Smith J, Parajuli S, Garg N, Aziz F, Mandelbrot D, Djamali A, Zhong W. Successful management of T-cell mediated rejection in a recent kidney transplant recipient with COVID-19 associated severe acute respiratory syndrome. Transpl Infect Dis 2021; 23:e13598. [PMID: 33780103 PMCID: PMC8250251 DOI: 10.1111/tid.13598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 02/21/2021] [Indexed: 12/11/2022]
Abstract
COVID-19-associated vasculitis has been reported as a defining feature of systemic disease including acute kidney injury. However, the understanding of COVID-19 kidney transplant-related injuries is still evolving. We report a case of AKI with isolated vasculitis (v2 lesion) in a new kidney transplant recipient with COVID-19 pneumonia.
Collapse
Affiliation(s)
- Maha Mohamed
- Department of MedicineDivision of NephrologyUniversity of Wisconsin School of MedicineMadisonWIUSA
| | - Jeannina Smith
- Department of MedicineDivision of Infectious DiseaseUniversity of Wisconsin School of MedicineMadisonWIUSA
| | - Sandesh Parajuli
- Department of MedicineDivision of NephrologyUniversity of Wisconsin School of MedicineMadisonWIUSA
| | - Neetika Garg
- Department of MedicineDivision of NephrologyUniversity of Wisconsin School of MedicineMadisonWIUSA
| | - Fahad Aziz
- Department of MedicineDivision of NephrologyUniversity of Wisconsin School of MedicineMadisonWIUSA
| | - Didier Mandelbrot
- Department of MedicineDivision of NephrologyUniversity of Wisconsin School of MedicineMadisonWIUSA
| | - Arjang Djamali
- Department of MedicineDivision of NephrologyUniversity of Wisconsin School of MedicineMadisonWIUSA
| | - Weixiong Zhong
- Department of Pathology and Laboratory MedicineUniversity of Wisconsin School of MedicineMadisonWIUSA
| |
Collapse
|
240
|
Sullivan KD, Galbraith MD, Kinning KT, Bartsch KW, Levinsky NC, Araya P, Smith KP, Granrath RE, Shaw JR, Baxter RM, Jordan KR, Russell SA, Dzieciatkowska ME, Reisz JA, Gamboni F, Cendali FI, Ghosh T, Monte AA, Bennett TD, Miller MG, Hsieh EWY, D'Alessandro A, Hansen KC, Espinosa JM. The COVIDome Explorer researcher portal. Cell Rep 2021; 36:109527. [PMID: 34348131 PMCID: PMC8316015 DOI: 10.1016/j.celrep.2021.109527] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/30/2021] [Accepted: 07/22/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 pathology involves dysregulation of diverse molecular, cellular, and physiological processes. To expedite integrated and collaborative COVID-19 research, we completed multi-omics analysis of hospitalized COVID-19 patients, including matched analysis of the whole-blood transcriptome, plasma proteomics with two complementary platforms, cytokine profiling, plasma and red blood cell metabolomics, deep immune cell phenotyping by mass cytometry, and clinical data annotation. We refer to this multidimensional dataset as the COVIDome. We then created the COVIDome Explorer, an online researcher portal where the data can be analyzed and visualized in real time. We illustrate herein the use of the COVIDome dataset through a multi-omics analysis of biosignatures associated with C-reactive protein (CRP), an established marker of poor prognosis in COVID-19, revealing associations between CRP levels and damage-associated molecular patterns, depletion of protective serpins, and mitochondrial metabolism dysregulation. We expect that the COVIDome Explorer will rapidly accelerate data sharing, hypothesis testing, and discoveries worldwide.
Collapse
Affiliation(s)
- Kelly Daniel Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Matthew Dominic Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kohl Thomas Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kyle William Bartsch
- Information Services, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nik Caldwell Levinsky
- Information Services, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Keith Patrick Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ross Erich Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jessica Rose Shaw
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ryan Michael Baxter
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kimberly Rae Jordan
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Seth Aaron Russell
- Data Science to Patient Value, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Monika Ewa Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Julie Ann Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Francesca Isabelle Cendali
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tusharkanti Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Andrew Albert Monte
- Department of Emergency Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tellen Demeke Bennett
- Department of Pediatrics, Sections of Informatics and Data Science and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael George Miller
- Information Services, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elena Wen-Yuan Hsieh
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pediatrics, Division of Allergy/Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kirk Charles Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Joaquin Maximiliano Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
241
|
Motolese F, Ferrante M, Rossi M, Magliozzi A, Sbarra M, Ursini F, Marano M, Capone F, Travaglino F, Antonelli Incalzi R, Di Lazzaro V, Pilato F. Posterior Reversible Encephalopathy Syndrome and brain haemorrhage as COVID-19 complication: a review of the available literature. J Neurol 2021; 268:4407-4414. [PMID: 34291313 PMCID: PMC8294241 DOI: 10.1007/s00415-021-10709-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023]
Abstract
Background SARS-CoV-2 infection has been associated with various neurological manifestations. Since patients affected by SARS-CoV-2 infection present coagulation and immune system dysregulation, ischemic or haemorragic stroke is not uncommon, irrespective of respiratory distress. However, the occurrence of focal neurological deficits together with other symptoms like headache, cortical blindness, seizure and altered mental status should prompt the diagnosis of Posterior Reversible Encephalopathy Syndrome (PRES). Antithrombotic treatment, the alteration of endothelial function, and coagulopathy due to COVID-19 and PRES leading to the breakdown of blood–brain barrier may then contribute to the occurrence of a brain haemorrhage. Methods We describe the case of a COVID-19 patient who developed bilateral occipital lobe haemorrhages suggestive of haemorrhagic PRES. We then reviewed the available literature about haemorrhagic evolution of PRES in COVID-19. Results We describe the clinical and radiological features of five COVID-19 patients who developed haemorrhagic PRES. Conclusions Coagulopathy and endothelial dysfunction resulting from the massive release of cytokines during the host immune response may be key factors in the pathogenesis of COVID-19-related PRES. Antithrombotic therapy and the leakage of the blood–brain barrier can subsequently increase the risk of haemorrhagic transformation of the lesioned brain tissue. A prompt diagnosis of PRES is mandatory, since the timely interruption/reversal of antithrombotic therapy may be a key determinant for a good prognosis.
Collapse
Affiliation(s)
- Francesco Motolese
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy.
| | - Mario Ferrante
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Mariagrazia Rossi
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Alessandro Magliozzi
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Martina Sbarra
- Departmental Faculty of Medicine and Surgery, Unit of Diagnostic Imaging and Interventional Radiology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesca Ursini
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Massimo Marano
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Fioravante Capone
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy
| | | | | | - Vincenzo Di Lazzaro
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Fabio Pilato
- Neurology, Neurophysiology and Neurobiology Unit, Department of Medicine, Università Campus Bio-Medico di Roma, Viale Alvaro del Portillo, 21, 00128, Rome, Italy
| |
Collapse
|
242
|
Sierra-Hidalgo F, Aragón Revilla E, Arranz García P, Martínez-Acebes E, Gómez-Moreno SM, Muñoz-Rivas N, Esquivel López A. Increased Incidence of In-Hospital Ischemic Stroke During SARS-CoV-2 Outbreak: A Single-Center Study. Neurocrit Care 2021; 36:208-215. [PMID: 34268645 PMCID: PMC8281805 DOI: 10.1007/s12028-021-01286-1] [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: 10/07/2020] [Accepted: 05/21/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Meta-analyses of observational studies report a 1.1-1.7% pooled risk of stroke among patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection requiring hospitalization, but consultations for stroke and reperfusion procedures have decreased during the outbreak that occurred during the first half of the year 2020. It is still unclear whether a true increase in the risk of stroke exists among patients with coronavirus disease 2019 (COVID-19). In-hospital ischemic stroke (IHIS) complicated the 0.04-0.06% of all admissions in the pre-COVID-19 era, but its incidence has not been assessed among inpatients with COVID-19. We aimed to compare IHIS incidence among patients with SARS-CoV-2 infection with that of inpatients with non-COVID-19 illnesses from the same outbreak period and from previous periods. METHODS This historical cohort study belongs to the COVID-19@Vallecas cohort. The incidence of IHIS was estimated for patients with SARS-CoV-2 hospitalized during March-April 2020 [COVID-19 cohort (CC)], for patients with non-COVID-19 medical illness hospitalized during the same outbreak period [2020 non-COVID-19 cohort (20NCC)], and for inpatients with non-COVID-19 illness admitted during March-April of the years 2016-2019 [historical non-COVID-19 cohort (HNCC)]. Unadjusted risk of IHIS was compared between the three cohorts, and adjusted incidence rate ratio (IRR) of IHIS between cohorts was obtained by means of Poisson regression. RESULTS Overall, 8126 inpatients were included in this study. Patients in the CC were younger and more commonly men than those from the HNCC and 20NCC. Absolute risk of IHIS was 0.05% for HNCC, 0.23% for 20NCC, and 0.36% for CC, (p = 0.004 for HNCC vs. CC). Cumulative incidence for IHIS by day nine after admission, with death as a competing risk, was 0.09% for HNCC, 0.23% for 20NCC, and 0.50% for CC. In an adjusted Poisson regression model with sex, age, needing of intensive care unit admission, and cohort (HNCC as reference) as covariates, COVID-19 was an independent predictor for IHIS (IRR 6.76, 95% confidence interval 1.66-27.54, p = 0.01). A nonsignificant increase in the risk of IHIS was observed for the 20NCC (IRR 5.62, 95% confidence interval 0.93-33.9, p = 0.06). CONCLUSIONS SARS-CoV-2 outbreak was associated with an increase in the incidence of IHIS when compared with inpatients from a historical cohort. Viral infection itself may be related to the increased risk of IHIS among patients with COVID-19, but in view of our results from the 20NCC, it is likely that other factors, such as hospital saturation and overwhelming of health systems, may have played a role in the increased frequency of IHIS.
Collapse
Affiliation(s)
- Fernando Sierra-Hidalgo
- Department of Neurology, Hospital Universitario Infanta Leonor, Avenida Gran Vía del Este 80, 28031, Madrid, Spain.
| | - Esther Aragón Revilla
- Department of Neurology, Hospital Universitario Infanta Leonor, Avenida Gran Vía del Este 80, 28031, Madrid, Spain
| | - Paz Arranz García
- Department of Medical Administration, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Eva Martínez-Acebes
- Department of Neurology, Hospital Universitario Infanta Leonor, Avenida Gran Vía del Este 80, 28031, Madrid, Spain
| | - Sonia Mayra Gómez-Moreno
- Department of Neurology, Hospital Universitario Infanta Leonor, Avenida Gran Vía del Este 80, 28031, Madrid, Spain
| | - Nuria Muñoz-Rivas
- Department of Internal Medicine, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Alberto Esquivel López
- Department of Neurology, Hospital Universitario Infanta Leonor, Avenida Gran Vía del Este 80, 28031, Madrid, Spain
| | | |
Collapse
|
243
|
Gusev E, Sarapultsev A, Hu D, Chereshnev V. Problems of Pathogenesis and Pathogenetic Therapy of COVID-19 from the Perspective of the General Theory of Pathological Systems (General Pathological Processes). Int J Mol Sci 2021; 22:7582. [PMID: 34299201 PMCID: PMC8304657 DOI: 10.3390/ijms22147582] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/30/2021] [Accepted: 07/12/2021] [Indexed: 01/18/2023] Open
Abstract
The COVID-19 pandemic examines not only the state of actual health care but also the state of fundamental medicine in various countries. Pro-inflammatory processes extend far beyond the classical concepts of inflammation. They manifest themselves in a variety of ways, beginning with extreme physiology, then allostasis at low-grade inflammation, and finally the shockogenic phenomenon of "inflammatory systemic microcirculation". The pathogenetic core of critical situations, including COVID-19, is this phenomenon. Microcirculatory abnormalities, on the other hand, lie at the heart of a specific type of general pathological process known as systemic inflammation (SI). Systemic inflammatory response, cytokine release, cytokine storm, and thrombo-inflammatory syndrome are all terms that refer to different aspects of SI. As a result, the metabolic syndrome model does not adequately reflect the pathophysiology of persistent low-grade systemic inflammation (ChSLGI). Diseases associated with ChSLGI, on the other hand, are risk factors for a severe COVID-19 course. The review examines the role of hypoxia, metabolic dysfunction, scavenger receptors, and pattern-recognition receptors, as well as the processes of the hemophagocytic syndrome, in the systemic alteration and development of SI in COVID-19.
Collapse
Affiliation(s)
- Evgenii Gusev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (E.G.); (V.C.)
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (E.G.); (V.C.)
- School of Medical Biology, South Ural State University, 454080 Chelyabinsk, Russia
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 200092, China;
| | - Valeriy Chereshnev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia; (E.G.); (V.C.)
| |
Collapse
|
244
|
Martinot M, Eyriey M, Gravier S, Bonijoly T, Kayser D, Ion C, Mohseni-Zadeh M, Camara S, Dubois J, Haerrel E, Drouaine J, Kaiser J, Ongagna JC, Schieber-Pachart A, Kempf C. Predictors of mortality, ICU hospitalization, and extrapulmonary complications in COVID-19 patients. Infect Dis Now 2021; 51:518-525. [PMID: 34242842 PMCID: PMC8260549 DOI: 10.1016/j.idnow.2021.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/08/2021] [Accepted: 07/01/2021] [Indexed: 01/08/2023]
Abstract
OBJECTIVE A major coronavirus disease 2019 (COVID-19) outbreak occurred in Northeastern France in spring 2020. This single-center retrospective observational cohort study aimed to compare patients with severe COVID-19 and those with non-severe COVID-19 (survivors vs. non-survivors, ICU patients vs. non-ICU patients) and to describe extrapulmonary complications. PATIENTS AND METHODS We included all patients with a confirmed diagnosis of COVID-19 admitted to Colmar Hospital in March 2020. RESULTS We examined 600 patients (median age 71.09 years; median body mass index: 26.9 kg/m2); 57.7% were males, 86.3% had at least one comorbidity, 153 (25.5%) required ICU hospitalization, and 115 (19.1%) died. Baseline independent factors associated with death were older age (>75 vs. ≤75 years), male sex, oxygen supply, chronic neurological, renal, and pulmonary diseases, diabetes, cancer, low platelet and hemoglobin counts, and high levels of C-reactive protein (CRP) and serum creatinine. Factors associated with ICU hospitalization were age <75 years, oxygen supply, chronic pulmonary disease, absence of dementia, and high levels of CRP, hemoglobin, and serum creatinine. Among the 600 patients, 80 (13.3%) had an acute renal injury, 33 (5.5%) had a cardiovascular event, 27 (4.5%) had an acute liver injury, 24 (4%) had venous thromboembolism, eight (1.3%) had a neurological event, five (0.8%) had rhabdomyolysis, and one had acute pancreatitis. Most extrapulmonary complications occurred in ICU patients. CONCLUSION This study highlighted the main risk factors for ICU hospitalization and death caused by severe COVID-19 and the frequency of numerous extrapulmonary complications in France.
Collapse
Affiliation(s)
- M Martinot
- Infectious Diseases Department, Hôpitaux Civils, 39, avenue de la liberté, 68024 Colmar, France.
| | - M Eyriey
- Clinical Research Department, Hôpitaux Civils de Colmar, Colmar, France
| | - S Gravier
- Infectious Diseases Department, Hôpitaux Civils, 39, avenue de la liberté, 68024 Colmar, France
| | - T Bonijoly
- Infectious Diseases Department, Hôpitaux Civils, 39, avenue de la liberté, 68024 Colmar, France
| | - D Kayser
- Infectious Diseases Department, Hôpitaux Civils, 39, avenue de la liberté, 68024 Colmar, France
| | - C Ion
- Infectious Diseases Department, Hôpitaux Civils, 39, avenue de la liberté, 68024 Colmar, France
| | - M Mohseni-Zadeh
- Infectious Diseases Department, Hôpitaux Civils, 39, avenue de la liberté, 68024 Colmar, France
| | - S Camara
- Clinical Research Department, Hôpitaux Civils de Colmar, Colmar, France
| | - J Dubois
- Clinical Research Department, Hôpitaux Civils de Colmar, Colmar, France
| | - E Haerrel
- Clinical Research Department, Hôpitaux Civils de Colmar, Colmar, France
| | - J Drouaine
- Clinical Research Department, Hôpitaux Civils de Colmar, Colmar, France
| | - J Kaiser
- Clinical Research Department, Hôpitaux Civils de Colmar, Colmar, France; Pharmacy Department, Hôpitaux Civils de Colmar, Colmar, France
| | - J C Ongagna
- Clinical Research Department, Hôpitaux Civils de Colmar, Colmar, France
| | | | - C Kempf
- Clinical Research Department, Hôpitaux Civils de Colmar, Colmar, France
| |
Collapse
|
245
|
Ionescu MA. COVID-19 skin lesions are rarely positive at RT-PCR test: the macrophage activation with vascular impact and SARS-CoV-2-induced cytokine storm. Int J Dermatol 2021; 61:3-6. [PMID: 34213786 PMCID: PMC8444652 DOI: 10.1111/ijd.15749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/11/2021] [Accepted: 06/10/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Several skin manifestations have been reported since the start of the COVID-19 pandemic: chilblains-like, livedoid lesions, urticaria-like, pseudo-Kawasaki disease, and others. Histopathologic images of these lesions most often show aspects of endothelitis, images similar to autoimmune vasculitis. Cutaneous lesions are often negative at RT-PCR for SARS-CoV-2 virus. METHOD AND RESULTS We reviewed recent articles on the mechanisms of COVID-19 and we synthesized main pathways of inflammatory cascade. After the penetration into the cells of the respiratory epithelium, SARS-CoV-2 virus initiates a "cytokine storm" well described in previous publications: the expression of interferon type I (IFN-I) is one of the key elements of the antiviral response in COVID-19 patients, IFN-I expression seems to play an important role in the induction of interleukin 6 (IL-6), chemotactic factors such as Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) and the consequent activation of monocyte-macrophage system followed by the expression of TNF-alpha, and finally by the induction of coagulation factors by both extrinsic and intrinsic pathways. CONCLUSIONS The simplified synthesis of the main pathophysiological mechanisms of COVID-19 could help us to understand at least partially the importance of macrophage activation and its vascular involvement in many skin lesions that remain often negative at in␣situ tests for SARS-CoV-2.
Collapse
|
246
|
Kamenskaya OV, Loginova IY, Klinkova AS, Lomivorotov VV, Chernyavsky AM. Clinical observations of COVID-19 infection in patients with chronic thromboembolic pulmonary hypertension. ACTA ACUST UNITED AC 2021; 61:28-34. [PMID: 34311685 DOI: 10.18087/cardio.2021.6.n1564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/07/2021] [Accepted: 05/01/2021] [Indexed: 11/18/2022]
Abstract
Aim To present clinical observations of the novel coronavirus infection (COVID-19) in patients with chronic thromboembolic pulmonary hypertension (CTEPH) after a surgical intervention in the form of thromobendarterectomy from pulmonary artery branches.Material and methods The Acad. E.N. Meshalkin National Medical Research Center performed 127 open surgical interventions for CTEPH in the form of thromobendarterectomy from 2016 through 2020. The present study enrolled 113 patients included into the follow-up care group and into the Center Registry who were followed up for more than 6 months after the surgery. Clinical and functional features of COVID-19 were evaluated in the studied group.Results In the follow-up care group, 5 (4.4%) postoperative CTEPH patients had COVID-19. One patient had asymptomatic disease, and others had typical clinical symptoms and bilateral polysegmental pneumonia. There were no cases requiring artificial ventilation and no lethal outcomes. All patients with COVID-19 received anticoagulants as a basis therapy for CTEPH, and two patients who had residual pulmonary arterial hypertension (PAH) additionally received a PAH-specific therapy. During the treatment of COVID-19, no adjustment of the anticoagulant or PAH-specific therapy was required.Conclusion The group of patients with CTEPH is a unique pathophysiological model for studying the effect of COVID-19 under the conditions of compromised pulmonary circulation. In the studied follow-up care group, the COVID-19 morbidity was 4.4 % without fatal outcomes. Evaluation of the role of chronic anticoagulant and PAH-specific therapy in COVID-19 postoperative patients as well as evaluation of the role of COVID-19 in CTEPH progression merit further investigation.
Collapse
Affiliation(s)
- O V Kamenskaya
- National medical research center named after E. Meshalkin of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - I Yu Loginova
- National medical research center named after E. Meshalkin of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - A S Klinkova
- National medical research center named after E. Meshalkin of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - V V Lomivorotov
- National medical research center named after E. Meshalkin of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - A M Chernyavsky
- National medical research center named after E. Meshalkin of the Ministry of Health of the Russian Federation, Novosibirsk, Russia
| |
Collapse
|
247
|
Hong LZ, Shou ZX, Zheng DM, Jin X. The most important biomarker associated with coagulation and inflammation among COVID-19 patients. Mol Cell Biochem 2021; 476:2877-2885. [PMID: 33742367 PMCID: PMC7978444 DOI: 10.1007/s11010-021-04122-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/25/2021] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) induced by SARS-Cov-2 can be related to coagulopathy. Also, the infection-induced inflammatory changes are found in patients with disseminated intravascular coagulopathy (DIC). The lack of previous immunity to COVID-19 has caused infection of a large number of patients worldwide and unpredictability regarding the management of the complications that appear in the course of this viral illness. Lungs are the most important target organ of the SARS-COV-2. In COVID-19 patients, acute lung injury leads to respiratory failure. However, multiorgan failure can also occur in these patients. The primary coagulopathy of COVID-19 is marked by a considerable elevation of D-dimer, ferritin, and fibrinogen degradation products. In comparison, abnormalities in platelet count, prothrombin time, and partial thromboplastin time are partly uncommon in initial presentations. Inflammatory biomarkers including CRP, LDH, and IL-6 are significantly elevated in the early stages of the disease. In this regard, inflammation-associated biomarkers and coagulation test screening, including the assessment of IL-6, CRP, LDH, D-dimer, platelet count, PT&PTT time, ferritin, and fibrinogen levels are suggested for detecting infection by this virus. Overall, COVID-19-associated coagulopathy should be managed like other patients with critical conditions, and supportive care and thromboembolic prophylaxis should be used for severe patients.
Collapse
Affiliation(s)
- Ling-Zhi Hong
- Emergency Department, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch), Hangzhou, 311700, Zhejiang, China
| | - Zhang-Xuan Shou
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - De-Ming Zheng
- Infectious Diseases Department, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch), Hangzhou, 311700, Zhejiang, China
| | - Xue Jin
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
248
|
Arslan G. Celiac Artery Thrombosis and Splenic Infarction as a Consequence of Mild COVID-19 Infection: Report of an Unusual Case. Hamostaseologie 2021; 42:193-194. [PMID: 34198348 DOI: 10.1055/a-1508-7388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
COVID-19 has been associated with the hypercoagulable state in the literature. Patients who are admitted to the hospital with severe COVID-19 may have some thrombotic complications. These patients have a high risk for venous and arterial thrombosis of large and small vessels. Here, a 42-year-old female with celiac artery thrombosis and splenic infarction after a history of mild COVID-19 was presented.
Collapse
Affiliation(s)
- Gokhan Arslan
- Department of Cardiovascular Surgery, Gulhane Faculty of Medicine, University of Health Sciences, Ankara, Turkey
| |
Collapse
|
249
|
Moon CY, Schilder BM, Raj T, Huang KL. Phenome-wide and expression quantitative trait locus associations of coronavirus disease 2019 genetic risk loci. iScience 2021; 24:102550. [PMID: 34027315 PMCID: PMC8129787 DOI: 10.1016/j.isci.2021.102550] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/24/2021] [Accepted: 05/13/2021] [Indexed: 12/15/2022] Open
Abstract
While several genes and clinical traits have been associated with higher risk of severe coronavirus disease 2019 (COVID-19), how host genetic variants may interact with these parameters and contribute to severe disease is still unclear. Herein, we performed phenome-wide association study, tissue and immune-cell-specific expression quantitative trait locus (eQTL)/splicing quantitative trait locus, and colocalization analyses for genetic risk loci suggestively associated with severe COVID-19 with respiratory failure. Thirteen phenotypes/traits were associated with the severe COVID-19-associated loci at the genome-wide significance threshold, including monocyte counts, fat metabolism traits, and fibrotic idiopathic interstitial pneumonia. In addition, we identified tissue and immune subtype-specific eQTL associations affecting 48 genes, including several ones that may directly impact host immune responses, colocalized with the severe COVID-19 genome-wide association study associations, and showed altered expression in single-cell transcriptomes. Collectively, our work demonstrates that host genetic variations associated with multiple genes and traits show genetic pleiotropy with severe COVID-19 and may inform disease etiology.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian M. Schilder
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Towfique Raj
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
250
|
Putilina MV, Grishin DV. SARS-CoV-2 (COVID-19) as a Predictor of Neuroinflammation and Neurodegeneration: Potential Treatment Strategies. ACTA ACUST UNITED AC 2021; 51:577-582. [PMID: 34176996 PMCID: PMC8219508 DOI: 10.1007/s11055-021-01108-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 (COVID-19) pandemic has attracted attention to the challenge of neuroinflammation as an unavoidable component of viral infections. Acute neuroinflammatory responses include activation of resident tissue macrophages in the CNS followed by release of a variety of cytokines and chemokines associated with activation of oxidative stress and delayed neuron damage. This makes the search for treatments with indirect anti-inflammatory properties relevant. From this point of view, attention is focused on further study of the treatment of patients with COVID-19 with dipyridamole (Curantil) which, having antiviral and anti-inflammatory effects, can inhibit acute inflammatory activity and progression of fibrosis, is a drug with potential, especially among patients with early increases in the D-dimer concentration and severe signs of microangiopathy.
Collapse
Affiliation(s)
- M V Putilina
- Pirogov Russian National Research Medical University, Russian Ministry of Health, Moscow, Russia
| | - D V Grishin
- Pirogov Russian National Research Medical University, Russian Ministry of Health, Moscow, Russia.,Filatov City Clinical Hospital No. 15, Moscow Health Department, Moscow, Russia
| |
Collapse
|