1
|
Oliveira KB, de Souza FMA, de Sá LBM, Pacheco ALD, Prado MR, de Sousa Rodrigues CF, Bassi ÊJ, Santana-Melo I, Silva-Júnior A, Sabino-Silva R, Shetty AK, de Castro OW. Potential Mechanisms Underlying COVID-19-Mediated Central and Peripheral Demyelination: Roles of the RAAS and ADAM-17. Mol Neurobiol 2025; 62:1151-1164. [PMID: 38965171 DOI: 10.1007/s12035-024-04329-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/21/2024] [Indexed: 07/06/2024]
Abstract
Demyelination is among the most conspicuous neurological sequelae of SARS-CoV-2 infection (COVID-19) in both the central (CNS) and peripheral (PNS) nervous systems. Several hypotheses have been proposed to explain the mechanisms underlying demyelination in COVID-19. However, none have considered the SARS-CoV-2's effects on the renin-angiotensin-aldosterone system (RAAS). Therefore, our objective in this review is to evaluate how RAAS imbalance, caused by direct and indirect effects of SARS-CoV-2 infection, could contribute to myelin loss in the PNS and CNS. In the PNS, we propose that demyelination transpires from two significant changes induced by SARS-CoV-2 infection, which include upregulation of ADAM-17 and induction of lymphopenia. Whereas, in the CNS, demyelination could result from RAAS imbalance triggering two alterations: (1) a decrease in angiotensin type II receptor (AT2R) activity, responsible for restraining defense cells' action on myelin; (2) upregulation of ADAM-17 activity, leading to impaired maturation of oligodendrocytes and myelin formation. Thus, we hypothesize that increased ADAM-17 activity and decreased AT2R activity play roles in SARS-CoV-2 infection-mediated demyelination in the CNS.
Collapse
Affiliation(s)
- Kellysson Bruno Oliveira
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Fernanda Maria Araujo de Souza
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Letícia Barros Maurício de Sá
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Amanda Larissa Dias Pacheco
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Mariana Reis Prado
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Célio Fernando de Sousa Rodrigues
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Ênio José Bassi
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Igor Santana-Melo
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Abelardo Silva-Júnior
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA.
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX, 77843, USA.
| | - Olagide Wagner de Castro
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Av. Lourival de Melo Mota, Km 14, Campus A. C. Simões, Cidade Universitária, Maceió, AL, CEP, 57072-970, Brazil.
| |
Collapse
|
2
|
Gonzalez-Ochoa AJ, Szolnoky G, Hernandez-Ibarra AG, Fareed J. Treatment with Sulodexide Downregulates Biomarkers for Endothelial Dysfunction in Convalescent COVID-19 Patients. Clin Appl Thromb Hemost 2025; 31:10760296241297647. [PMID: 39763448 PMCID: PMC11705351 DOI: 10.1177/10760296241297647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Persistent elevation of biomarkers associated with endothelial dysfunction in convalescent COVID-19 patients has been linked to an increased risk of long-term cardiovascular complications, including long COVID syndrome. Sulodexide, known for its vascular endothelial affinity, has demonstrated pleiotropic protective properties. This study aims to evaluate the impact of sulodexide on serum levels of endothelial dysfunction biomarkers in patients during the convalescent phase of COVID-19. METHODS We conducted a double-blind, single-center, randomized, placebo-controlled trial in Mexico, comparing sulodexide (250 LRU orally, twice daily) with placebo over 8 weeks in adult patients during early COVID-19 convalescence. Differences in serum biomarkers between the groups were analyzed using repeated measures and post hoc tests, with Thrombomodulin (TM) as the primary endpoint. RESULTS Among 206 analyzed patients (103 in each group), at week 8, the sulodexide group exhibited significantly lower mean levels of Thrombomodulin (TM) (25.2 ± 7.9 ng/mL vs 29.9 ± 14.7 ng/mL, P = .03), von Willebrand Factor (vWF) (232 ± 131 U/dL vs 266 ± 122 U/dL, P = .02) and Interleukin-6 (IL-6) (12.5 ± 13.2 pg/mL vs 16.2 ± 16.5 pg/mL, P = .03) compared to the placebo group. D-dimer and C reactive protein (CRP) in the sulodexide group were also lowered. No significant differences were observed for P-selectin, fibrinogen, VCAM-1, or ICAM-1 levels. CONCLUSIONS Patients in the convalescent phase of COVID-19 who received sulodexide for eight weeks showed a reduction in TM, vWF, D-dimer, CRP, and IL-6 serum levels compared to placebo. These findings suggest a potential protective effect of sulodexide against thromboinflammation and endothelial damage.
Collapse
Affiliation(s)
- Alejandro J Gonzalez-Ochoa
- Vascular Surgery Department, Centro Médico del Noroeste, San Luis Rio Colorado, Sonora, México
- Vascular and Endovascular Surgery department, CLINEDEM, San Luis Rio Colorado, Sonora, México
| | - Gyozo Szolnoky
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | | | - Jawed Fareed
- Hemostasis and Thrombosis Research Laboratories, Loyola University Medical Center, Maywood, Illinois, USA
| |
Collapse
|
3
|
Azami P, Vafa RG, Heydarzadeh R, Sadeghi M, Amiri F, Azadian A, Khademolhosseini A, Yousefi M, Montaseri M, Hosseini N, Hosseini SA, Kojuri J. Evaluation of blood pressure variation in recovered COVID-19 patients at one-year follow-up: a retrospective cohort study. BMC Cardiovasc Disord 2024; 24:240. [PMID: 38714940 PMCID: PMC11075195 DOI: 10.1186/s12872-024-03916-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has various sequelae, one of which might be hypertension. We aimed to evaluate COVID-19's impact on blood pressure (BP) in non-hospitalized patients at one-year follow-up. METHOD A total of 7,950 consecutive COVID-19 patients regularly visiting our cardiology clinic were retrospectively screened. Patients' electronic medical records including demographics, comorbidities, vital signs, treatments, and outcomes, were reviewed by two physicians. Individuals with at least one BP measurement in the three months preceding COVID-19 and one measurement in 12 months or more following recovery were included. BP levels before and after COVID-19 were compared using the paired t-test. RESULTS 5,355 confirmed COVID-19 patients (mean age 55.51 ± 15.38 years) were included. Hypertension (56.9%) and diabetes mellitus (34%) were the predominant comorbidities, and 44.3% had prior major adverse cardiovascular events. Both systolic (126.90 ± 20.91 vs. 139.99 ± 23.94 mmHg, P < 0.001) and diastolic BP (80.54 ± 13.94 vs. 86.49 ± 14.40 mmHg, P < 0.001) were significantly higher post-COVID-19 vs. pre-COVID-19. Notably, 456 (14%) hypertensive patients experienced exacerbated hypertension, while 408 (17%) patients developed new-onset hypertension, overall 864 (16%) of patients had exacerbation or new hypertension. Linear regression analysis revealed that advanced age, smoking, previous cardiovascular events, hypertension, and diabetes mellitus predict increased BP following COVID-19 (P < 0.001). CONCLUSION COVID-19 raised systolic and diastolic BP in the long term in non-hospitalized patients, with over one-sixth developing new-onset or exacerbated hypertension. All patients should be evaluated regarding BP, following COVID-19 recovery, particularly those with the mentioned predictive factors. (clinicaltrial.gov: NCT05798208).
Collapse
Affiliation(s)
- Pouria Azami
- Shiraz University of Medical Sciences, Shiraz, Iran
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
- Cardiology Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Golchin Vafa
- Shiraz University of Medical Sciences, Shiraz, Iran
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Reza Heydarzadeh
- Shiraz University of Medical Sciences, Shiraz, Iran
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Mehrdad Sadeghi
- Shiraz University of Medical Sciences, Shiraz, Iran
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Farhang Amiri
- Shiraz University of Medical Sciences, Shiraz, Iran
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Alireza Azadian
- Shiraz University of Medical Sciences, Shiraz, Iran
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Amin Khademolhosseini
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Mina Yousefi
- Shahid sadoughi University of Medical sciences, Yazd, Iran
| | - Mohammad Montaseri
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Nazanin Hosseini
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Seyed Ali Hosseini
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran
| | - Javad Kojuri
- Shiraz University of Medical Sciences, Shiraz, Iran.
- Professor Kojuri Cardiology Clinic, Niayesh St. Niayesh Medical Complex, Shiraz, Iran.
- Cardiology Department, Shiraz University of Medical Sciences, Shiraz, Iran.
- Clinical Education Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Pai V, Bileck A, Hommer N, Janku P, Lindner T, Kauer V, Rumpf B, Haslacher H, Hagn G, Meier-Menches SM, Schmetterer L, Schmidl D, Gerner C, Garhöfer G. Impaired retinal oxygen metabolism and perfusion are accompanied by plasma protein and lipid alterations in recovered COVID-19 patients. Sci Rep 2024; 14:8395. [PMID: 38600099 PMCID: PMC11006918 DOI: 10.1038/s41598-024-56834-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 03/12/2024] [Indexed: 04/12/2024] Open
Abstract
The aim of the present study was to investigate retinal microcirculatory and functional metabolic changes in patients after they had recovered from a moderate to severe acute COVID-19 infection. Retinal perfusion was quantified using laser speckle flowgraphy. Oxygen saturation and retinal calibers were assessed with a dynamic vessel analyzer. Arterio-venous ratio (AVR) was calculated based on retinal vessel diameter data. Blood plasma samples underwent mass spectrometry-based multi-omics profiling, including proteomics, metabolomics and eicosadomics. A total of 40 subjects were included in the present study, of which 29 had recovered from moderate to severe COVID-19 within 2 to 23 weeks before inclusion and 11 had never had COVID-19, as confirmed by antibody testing. Perfusion in retinal vessels was significantly lower in patients (60.6 ± 16.0 a.u.) than in control subjects (76.2 ± 12.1 a.u., p = 0.006). Arterio-venous (AV) difference in oxygen saturation and AVR was significantly lower in patients compared to healthy controls (p = 0.021 for AVR and p = 0.023 for AV difference in oxygen saturation). Molecular profiles demonstrated down-regulation of cell adhesion molecules, NOTCH3 and fatty acids, and suggested a bisphasic dysregulation of nitric oxide synthesis after COVID-19 infection. The results of this study imply that retinal perfusion and oxygen metabolism is still significantly altered in patients well beyond the acute phase of COVID-19. This is also reflected in the molecular profiling analysis of blood plasma, indicating a down-regulation of nitric oxide-related endothelial and immunological cell functions.Trial Registration: ClinicalTrials.gov ( https://clinicaltrials.gov ) NCT05650905.
Collapse
Affiliation(s)
- Viktoria Pai
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Vienna, Austria
| | - Nikolaus Hommer
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Patrick Janku
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Theresa Lindner
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Victoria Kauer
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Department of Medicine IV for Infectious Diseases and Tropical Medicine, Clinic Favoriten, Vienna, Austria
| | - Benedikt Rumpf
- Department of Medicine IV for Infectious Diseases and Tropical Medicine, Clinic Favoriten, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hagn
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090, Vienna, Austria
| | - Samuel M Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Leopold Schmetterer
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
- SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Doreen Schmidl
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Währinger Straße 38, 1090, Vienna, Austria.
- Joint Metabolome Facility, University of Vienna and Medical University Vienna, Vienna, Austria.
| | - Gerhard Garhöfer
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
5
|
EFE S, Demircan F, UÇAN A, İNAL V. The use of furosemide during Intravenous Immunoglobulin therapy should not always be considered contraindicated. MEDICINE IN DRUG DISCOVERY 2024; 21:100171. [DOI: 10.1016/j.medidd.2023.100171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2024] Open
|
6
|
Perico L, Benigni A, Remuzzi G. SARS-CoV-2 and the spike protein in endotheliopathy. Trends Microbiol 2024; 32:53-67. [PMID: 37393180 PMCID: PMC10258582 DOI: 10.1016/j.tim.2023.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 07/03/2023]
Abstract
SARS-CoV-2, the causative agent of COVID-19, primarily affects the epithelial compartment in the upper and lower airways. There is evidence that the microvasculature in both the pulmonary and extrapulmonary systems is a major target of SARS-CoV-2. Consistent with this, vascular dysfunction and thrombosis are the most severe complications in COVID-19. The proinflammatory milieu triggered by the hyperactivation of the immune system by SARS-CoV-2 has been suggested to be the main trigger for endothelial dysfunction during COVID-19. More recently, a rapidly growing number of reports have indicated that SARS-CoV-2 can interact directly with endothelial cells through the spike protein, leading to multiple instances of endothelial dysfunction. Here, we describe all the available findings showing the direct effect of the SARS-CoV-2 spike protein on endothelial cells and offer mechanistic insights into the molecular basis of vascular dysfunction in severe COVID-19.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy.
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Bergamo, Italy
| |
Collapse
|
7
|
Jin L, Wu L, Chen J, Zhang M, Sun J, Shen C, Du L, She X, Li Z. Uncoupling of the center-to-periphery arterial stiffness gradient and pulse pressure amplification in viral pneumonia infection. BMC Infect Dis 2023; 23:657. [PMID: 37798630 PMCID: PMC10552441 DOI: 10.1186/s12879-023-08650-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023] Open
Abstract
OBJECTIVES Arterial stiffness is a common manifestation of viral pneumonia infections, including COVID-19. Nevertheless, the relationship between the center-to-periphery arterial stiffness gradient and pulse pressure amplification (PPA) in infectious diseases remains unclear. This study aimed to investigate this relationship utilizing arterial pressure volume index (API) and arterial velocity pulse index (AVI) ratio. METHODS API/AVI and PPA were measured in 219 participants with COVID-19 and 374 normal participants. Multiple linear regression was used to assess the association of API/AVI and PPA, and restricted cubic spline was used to investigate the non-linear relationship between API/AVI and PPA. Receiver operating characteristic curve (ROC) analysis was used to evaluate the effects of API/AVI in identifying COVID-19 infection and severe stage. RESULTS There was a significant J-shaped relationship between API/AVI and PPA in COVID-19 group, while a M-shaped relationship was observed in normal group. API/AVI decreased rapidly as PPA decreased until API/AVI decreased slowly at PPA of 1.07, and then API/AVI decreased slowly again at PPA of 0.78. ROC results showed that API/AVI demonstrated excellent accuracy in identifying COVID-19 infection (AUC = 0.781) and a high specificity (84.88%) in identifying severe stage. CONCLUSIONS There was a J-shaped association between the API/AVI and PPA in viral infected patients, while a M-shaped relationship in the normal participants. API/AVI is better for identifying infected and uninfected patients, with a high specificity in identifying those in severe stages of the disease. The attenuation or reversal of API/AVI may be associated with the loss of PPA coupling.
Collapse
Affiliation(s)
- Lin Jin
- Department of Ultrasound, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200052, China
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Lingheng Wu
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
- Department of Ultrasound, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Jianxiong Chen
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
- Department of Ultrasound, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Mengjiao Zhang
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Jiali Sun
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Cuiqin Shen
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xiaoyin She
- Department of Emergency and Critical Care, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Zhaojun Li
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China.
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
8
|
Roche R, Odeh NH, Andar AU, Tulapurkar ME, Roche JA. Protection against Severe Illness versus Immunity-Redefining Vaccine Effectiveness in the Aftermath of COVID-19. Microorganisms 2023; 11:1963. [PMID: 37630523 PMCID: PMC10459411 DOI: 10.3390/microorganisms11081963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/03/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Anti-SARS-CoV-2 vaccines have played a pivotal role in reducing the risk of developing severe illness from COVID-19, thus helping end the COVID-19 global public health emergency after more than three years. Intriguingly, as SARS-CoV-2 variants emerged, individuals who were fully vaccinated did get infected in high numbers, and viral loads in vaccinated individuals were as high as those in the unvaccinated. However, even with high viral loads, vaccinated individuals were significantly less likely to develop severe illness; this begs the question as to whether the main effect of anti-SARS-CoV-2 vaccines is to confer protection against severe illness or immunity against infection. The answer to this question is consequential, not only to the understanding of how anti-SARS-CoV-2 vaccines work, but also to public health efforts against existing and novel pathogens. In this review, we argue that immune system sensitization-desensitization rather than sterilizing immunity may explain vaccine-mediated protection against severe COVID-19 illness even when the SARS-CoV-2 viral load is high. Through the lessons learned from COVID-19, we make the case that in the disease's aftermath, public health agencies must revisit healthcare policies, including redefining the term "vaccine effectiveness."
Collapse
Affiliation(s)
- Renuka Roche
- Occupational Therapy Program, School of Health Sciences, College of Health and Human Services, Eastern Michigan University, Ypsilanti, MI 48197, USA;
| | - Nouha H. Odeh
- Ph.D. Program in Immunology and Microbiology, Department of Biochemistry, Microbiology & Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA;
| | - Abhay U. Andar
- Baltimore County, Translational Life Science Technology, University of Maryland, Rockville, MD 20850, USA;
| | - Mohan E. Tulapurkar
- Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph A. Roche
- Physical Therapy Program, Department of Health Care Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
9
|
Petrache I, Pujadas E, Ganju A, Serban KA, Borowiec A, Babbs B, Bronova IA, Egersdorf N, Hume PS, Goel K, Janssen WJ, Berdyshev EV, Cordon-Cardo C, Kolesnick R. Marked elevations in lung and plasma ceramide in COVID-19 linked to microvascular injury. JCI Insight 2023; 8:e156104. [PMID: 37212278 PMCID: PMC10322682 DOI: 10.1172/jci.insight.156104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 04/05/2023] [Indexed: 05/23/2023] Open
Abstract
The pathogenesis of the marked pulmonary microvasculature injury, a distinguishing feature of COVID-19 acute respiratory distress syndrome (COVID-ARDS), remains unclear. Implicated in the pathophysiology of diverse diseases characterized by endothelial damage, including ARDS and ischemic cardiovascular disease, ceramide and in particular palmitoyl ceramide (C16:0-ceramide) may be involved in the microvascular injury in COVID-19. Using deidentified plasma and lung samples from COVID-19 patients, ceramide profiling by mass spectrometry was performed. Compared with healthy individuals, a specific 3-fold C16:0-ceramide elevation in COVID-19 patient plasma was identified. Compared with age-matched controls, autopsied lungs of individuals succumbing to COVID-ARDS displayed a massive 9-fold C16:0-ceramide elevation and exhibited a previously unrecognized microvascular ceramide-staining pattern and markedly enhanced apoptosis. In COVID-19 plasma and lungs, the C16-ceramide/C24-ceramide ratios were increased and reversed, respectively, consistent with increased risk of vascular injury. Indeed, exposure of primary human lung microvascular endothelial cell monolayers to C16:0-ceramide-rich plasma lipid extracts from COVID-19, but not healthy, individuals led to a significant decrease in endothelial barrier function. This effect was phenocopied by spiking healthy plasma lipid extracts with synthetic C16:0-ceramide and was inhibited by treatment with ceramide-neutralizing monoclonal antibody or single-chain variable fragment. These results indicate that C16:0-ceramide may be implicated in the vascular injury associated with COVID-19.
Collapse
Affiliation(s)
- Irina Petrache
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Elisabet Pujadas
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aditya Ganju
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Karina A. Serban
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Alexander Borowiec
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Beatrice Babbs
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Irina A. Bronova
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Nicholas Egersdorf
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Patrick S. Hume
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Khushboo Goel
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - William J. Janssen
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Evgeny V. Berdyshev
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Richard Kolesnick
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
10
|
Beckman JD, DaSilva A, Aronovich E, Nguyen A, Nguyen J, Hargis G, Reynolds D, Vercellotti GM, Betts B, Wood DK. JAK-STAT inhibition reduces endothelial prothrombotic activation and leukocyte-endothelial proadhesive interactions. J Thromb Haemost 2023; 21:1366-1380. [PMID: 36738826 PMCID: PMC10246778 DOI: 10.1016/j.jtha.2023.01.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Vascular activation is characterized by increased proinflammatory, pro thrombotic, and proadhesive signaling. Several chronic and acute conditions, including Bcr-abl-negative myeloproliferative neoplasms (MPNs), graft-vs-host disease, and COVID-19 have been noted to have increased activation of the janus kinase (JAK)-signal transducer and downstream activator of transcription (STAT) pathways. Two notable inhibitors of the JAK-STAT pathway are ruxolitinib (JAK1/2 inhibitor) and fedratinib (JAK2 inhibitor), which are currently used to treat MPN patients. However, in some conditions, it has been noted that JAK inhibitors can increase the risk of thromboembolic complications. OBJECTIVES We sought to define the anti-inflammatory and antithrombotic effects of JAK-STAT inhibitors in vascular endothelial cells. METHODS We assessed endothelial activation in the presence or absence of ruxolitinib or fedratinib by using immunoblots, immunofluorescence, qRT-PCR, and function coagulation assays. Finally, we used endothelialized microfluidics perfused with blood from normal and JAK2V617F+ individuals to evaluate whether ruxolitinib and fedratinib changed cell adhesion. RESULTS We found that both ruxolitinib and fedratinib reduced endothelial cell phospho-STAT1 and STAT3 signaling and attenuated nuclear phospho-NK-κB and phospho-c-Jun localization. JAK-STAT inhibition also limited secretion of proadhesive and procoagulant P-selectin and von Willebrand factor and proinflammatory IL-6. Likewise, we found that JAK-STAT inhibition reduced endothelial tissue factor and urokinase plasminogen activator expression and activity. CONCLUSIONS By using endothelialized microfluidics perfused with whole blood samples, we demonstrated that endothelial treatment with JAK-STAT inhibitors prevented rolling of both healthy control and JAK2V617F MPN leukocytes. Together, these findings demonstrate that JAK-STAT inhibitors reduce the upregulation of critical prothrombotic pathways and prevent increased leukocyte-endothelial adhesion.
Collapse
Affiliation(s)
- Joan D Beckman
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA.
| | - Angelica DaSilva
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Elena Aronovich
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Aithanh Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geneva Hargis
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - David Reynolds
- Department of Biomedical Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gregory M Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brian Betts
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
11
|
Gao Y, Li M, Wang B, Ma Y. Prognostic value of Nrf2/HO-1 expression and its correlation with occurrence in esophageal squamous cell carcinoma. Genes Genomics 2023; 45:723-739. [PMID: 37043130 DOI: 10.1007/s13258-023-01371-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/16/2023] [Indexed: 04/13/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is thought to be started and developed by genes associated with inflammation. A cancer's ability to spread and grow can be aided by nuclear factor erythroid-2 related factor 2 (Nrf2) hyperactivation, which can also make a tumor more resistant to chemotherapy and radiation treatment. However, it is still unknown how Nrf2 gene expression affects ESCC prognosis and controls function throughout ESCC advancement. OBJECTIVE The expression of Nrf2 and HO-1 in ESCC and precancerous esophageal precancerous lesions was analyzed, and their relationship with esophageal squamous cell carcinoma was analyzed. METHODS Immunohistochemistry (IHC) was used to confirm the expression of Nrf2 and heme oxygenase-1 (HO-1) proteins in tissue microarrays from Chinese populations with ESCC. We looked at the connections between Nrf2/HO-1 expression and invading immune cells using the TIMER database. RESULTS Ethnicity and N stage are associated with Nrf2 overexpression. Differentiation, N stage, vascular invasion, distant metastasis, and American Joint Committee on Cancer (AJCC) staging are all associated with HO-1 overexpression. The expression of Nrf2 and HO-1 had a favorable correlation. Patients with elevated Nrf2 and HO-1 expression had lower progression-free survival (PFS) and overall survival (OS). In high-grade intraepithelial neoplasia, Nrf2 and HO-1 expression generally occurred, partially in low-grade intraepithelial neoplasia specimens, and rarely in normal mucosa. We further show that Nrf2 suppression is linked to higher immunological marker expression and lower immune cell infiltration. CONCLUSION The prognosis of ESCC may be improved by inhibiting the expression of Nrf2 and HO-1. A lack of immune cells was seen in ESCC with Nrf2 impairment.
Collapse
Affiliation(s)
- Yongmei Gao
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Mengyan Li
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Bo Wang
- Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yuqing Ma
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| |
Collapse
|
12
|
Oliveira KB, de Melo IS, da Silva BRM, Oliveira KLDS, Sabino-Silva R, Anhezini L, Katayama PL, Santos VR, Shetty AK, de Castro OW. SARS-CoV-2 and Hypertension: Evidence Supporting Invasion into the Brain Via Baroreflex Circuitry and the Role of Imbalanced Renin-Angiotensin-Aldosterone-System. Neurosci Insights 2023; 18:26331055231151926. [PMID: 36756280 PMCID: PMC9900164 DOI: 10.1177/26331055231151926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023] Open
Abstract
Hypertension is considered one of the most critical risk factors for COVID-19. Evidence suggests that SARS-CoV-2 infection produces intense effects on the cardiovascular system by weakening the wall of large vessels via vasa-vasorum. In this commentary, we propose that SARS-CoV-2 invades carotid and aortic baroreceptors, leading to infection of the nucleus tractus solitari (NTS) and paraventricular hypothalamic nucleus (PVN), and such dysregulation of NTS and PVN following infection causes blood pressure alteration at the central level. We additionally explored the hypothesis that SARS-CoV-2 favors the internalization of membrane ACE2 receptors generating an imbalance of the renin-angiotensin-aldosterone system (RAAS), increasing the activity of angiotensin II (ANG-II), disintegrin, and metalloproteinase 17 domain (ADAM17/TACE), eventually modulating the integration of afferents reaching the NTS from baroreceptors and promoting increased blood pressure. These mechanisms are related to the increased sympathetic activity, which leads to transient or permanent hypertension associated with SARS-CoV-2 invasion, contributing to the high number of deaths by cardiovascular implications.
Collapse
Affiliation(s)
- Kellysson Bruno Oliveira
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Igor Santana de Melo
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Bianca Rodrigues Melo da Silva
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Keylla Lavínia da Silva Oliveira
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of
Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlândia, Minas
Gerais, Brazil
| | - Lucas Anhezini
- Department of Histology, Institute of
Biological Sciences and Health, Federal University of Alagoas, Maceió, Alagoas,
Brazil
| | - Pedro Lourenco Katayama
- Department of Physiology and Pathology,
Dentistry School of Araraquara, São Paulo State University, Araraquara, São Paulo,
Brazil
| | - Victor Rodrigues Santos
- Department of Morphology, Institute of
Biological Science, Federal University of Minas Gerais (UFMG), Belo Horizonte, Minas
Gerais, Brazil
| | - Ashok K Shetty
- Institute for Regenerative Medicine,
Department of Cell Biology and Genetics, Texas A&M University School of
Medicine, College Station, TX, USA
| | - Olagide Wagner de Castro
- Department of Physiology, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió,
Alagoas, Brazil,Olagide Wagner de Castro, Institute of
Biological Sciences and Health, Federal University of Alagoas (UFAL), Av.
Lourival de Melo Mota, km 14, Campus A. C. Simões, Cidade Universitária, Maceió,
Alagoas CEP 57072-970, Brazil.
| |
Collapse
|
13
|
Jankauskas SS, Kansakar U, Sardu C, Varzideh F, Avvisato R, Wang X, Matarese A, Marfella R, Ziosi M, Gambardella J, Santulli G. COVID-19 Causes Ferroptosis and Oxidative Stress in Human Endothelial Cells. Antioxidants (Basel) 2023; 12:326. [PMID: 36829885 PMCID: PMC9952002 DOI: 10.3390/antiox12020326] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/03/2023] Open
Abstract
Oxidative stress and endothelial dysfunction have been shown to play crucial roles in the pathophysiology of COVID-19 (coronavirus disease 2019). On these grounds, we sought to investigate the impact of COVID-19 on lipid peroxidation and ferroptosis in human endothelial cells. We hypothesized that oxidative stress and lipid peroxidation induced by COVID-19 in endothelial cells could be linked to the disease outcome. Thus, we collected serum from COVID-19 patients on hospital admission, and we incubated these sera with human endothelial cells, comparing the effects on the generation of reactive oxygen species (ROS) and lipid peroxidation between patients who survived and patients who did not survive. We found that the serum from non-survivors significantly increased lipid peroxidation. Moreover, serum from non-survivors markedly regulated the expression levels of the main markers of ferroptosis, including GPX4, SLC7A11, FTH1, and SAT1, a response that was rescued by silencing TNFR1 on endothelial cells. Taken together, our data indicate that serum from patients who did not survive COVID-19 triggers lipid peroxidation in human endothelial cells.
Collapse
Affiliation(s)
- Stanislovas S. Jankauskas
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Urna Kansakar
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Celestino Sardu
- University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | - Fahimeh Varzideh
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Roberta Avvisato
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- “Federico II” University, 80131 Naples, Italy
| | - Xujun Wang
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | | | | | - Jessica Gambardella
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- “Federico II” University, 80131 Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- “Federico II” University, 80131 Naples, Italy
- Department of Molecular Pharmacology, Einstein Institute for Neuroimmunology and Inflammation (INI), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
14
|
COVID-19 Pathology Sheds Further Light on Balance between Neutrophil Proteases and Their Inhibitors. Biomolecules 2022; 13:biom13010082. [PMID: 36671467 PMCID: PMC9855895 DOI: 10.3390/biom13010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Excessive neutrophil influx and activation in lungs during infections, such as manifest during the ongoing SARS CoV-2 pandemic, have brought neutrophil extracellular traps (NETs) and the concomitant release of granule contents that damage surrounding tissues into sharp focus. Neutrophil proteases, which are known to participate in NET release, also enable the binding of the viral spike protein to cellular receptors and assist in the spread of infection. Blood and tissue fluids normally also contain liver-derived protease inhibitors that balance the activity of proteases. Interestingly, neutrophils themselves also express the protease inhibitor alpha-1-antitrypsin (AAT), the product of the SERPINA-1 gene, and store it in neutrophil cytoplasmic granules. The absence of AAT or mutations in the SERPINA-1 gene promotes lung remodeling and fibrosis in diseases such as chronic obstructive pulmonary disease (COPD), and acute respiratory distress syndrome (ARDS) and increases the risk of allergic responses. Recent observations point to the fact that reduced activity of AAT presents a major susceptibility factor for severe COVID-19. Here, we focus attention on the mechanism of neutrophil elastase (NE) in NET release and its inhibition by AAT as an additional factor that may determine the severity of COVID-19.
Collapse
|
15
|
Moran TE, Hammers DE, Lee SW. The Role of Host-Cellular Responses in COVID-19 Endothelial Dysfunction. Curr Drug Targets 2022; 23:1555-1566. [PMID: 35748550 DOI: 10.2174/1389450123666220624094940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/11/2022] [Accepted: 05/11/2022] [Indexed: 01/25/2023]
Abstract
SARS-CoV2, Severe acute respiratory syndrome coronavirus 2, is a novel member of the human coronavirus family that has recently emerged worldwide to cause COVID-19 disease. COVID-19 disease has been declared a worldwide pandemic with over 270 million total cases, and >5 million deaths as of this writing. Although co-morbidities and preexisting conditions have played a significant role in the severity of COVID-19, the hallmark feature of severe disease associated with SARS-CoV2 is respiratory failure. Recent findings have demonstrated a key role for endothelial dysfunction caused by SARS-CoV2 in these clinical outcomes, characterized by endothelial inflammation, the persistence of a pro-coagulative state, and major recruitment of leukocytes and other immune cells to localized areas of endothelial dysfunction. Though it is generally recognized that endothelial impairment is a major contributor to COVID-19 disease, studies to examine the initial cellular events involved in triggering endothelial dysfunction are needed. In this article, we review the general strategy of pathogens to exploit endothelial cells and the endothelium to cause disease. We discuss the role of the endothelium in COVID-19 disease and highlight very recent findings that identify key signaling and cellular events that are associated with the initiation of SARS-CoV2 infection. These studies may reveal specific molecular pathways that can serve as potential means of therapeutic development against COVID-19 disease.
Collapse
Affiliation(s)
- Thomas E Moran
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Daniel E Hammers
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA
| | - Shaun W Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA.,W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN, USA.,Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
16
|
Miceli G, Basso MG, Rizzo G, Pintus C, Tuttolomondo A. The Role of the Coagulation System in Peripheral Arterial Disease: Interactions with the Arterial Wall and Its Vascular Microenvironment and Implications for Rational Therapies. Int J Mol Sci 2022; 23:14914. [PMID: 36499242 PMCID: PMC9739112 DOI: 10.3390/ijms232314914] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
Peripheral artery disease (PAD) is a clinical manifestation of atherosclerotic disease with a large-scale impact on the economy and global health. Despite the role played by platelets in the process of atherogenesis being well recognized, evidence has been increasing on the contribution of the coagulation system to the atherosclerosis formation and PAD development, with important repercussions for the therapeutic approach. Histopathological analysis and some clinical studies conducted on atherosclerotic plaques testify to the existence of different types of plaques. Likely, the role of coagulation in each specific type of plaque can be an important determinant in the histopathological composition of atherosclerosis and in its future stability. In this review, we analyze the molecular contribution of inflammation and the coagulation system on PAD pathogenesis, focusing on molecular similarities and differences between atherogenesis in PAD and coronary artery disease (CAD) and discussing the possible implications for current therapeutic strategies and future perspectives accounting for molecular inflammatory and coagulation targets. Understanding the role of cross-talking between coagulation and inflammation in atherosclerosis genesis and progression could help in choosing the right patients for future dual pathway inhibition strategies, where an antiplatelet agent is combined with an anticoagulant, whose role, despite pathophysiological premises and trials' results, is still under debate.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| |
Collapse
|
17
|
Qin R, Kurz E, Chen S, Zeck B, Chiribogas L, Jackson D, Herchen A, Attia T, Carlock M, Rapkiewicz A, Bar-Sagi D, Ritchie B, Ross TM, Mahal LK. α2,6-Sialylation Is Upregulated in Severe COVID-19, Implicating the Complement Cascade. ACS Infect Dis 2022; 8:2348-2361. [PMID: 36219583 PMCID: PMC9578644 DOI: 10.1021/acsinfecdis.2c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Indexed: 01/29/2023]
Abstract
Better understanding of the molecular mechanisms underlying COVID-19 severity is desperately needed in current times. Although hyper-inflammation drives severe COVID-19, precise mechanisms triggering this cascade and what role glycosylation might play therein are unknown. Here we report the first high-throughput glycomic analysis of COVID-19 plasma samples and autopsy tissues. We find that α2,6-sialylation is upregulated in the plasma of patients with severe COVID-19 and in autopsied lung tissue. This glycan motif is enriched on members of the complement cascade (e.g., C5, C9), which show higher levels of sialylation in severe COVID-19. In the lung tissue, we observe increased complement deposition, associated with elevated α2,6-sialylation levels, corresponding to elevated markers of poor prognosis (IL-6) and fibrotic response. We also observe upregulation of the α2,6-sialylation enzyme ST6GAL1 in patients who succumbed to COVID-19. Our work identifies a heretofore undescribed relationship between sialylation and complement in severe COVID-19, potentially informing future therapeutic development.
Collapse
Affiliation(s)
- Rui Qin
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Emma Kurz
- Department
of Cell Biology, NYU Grossman School of
Medicine, 550 First Avenue, New York, New York 10016, United
States
| | - Shuhui Chen
- Department
of Chemistry, Biomedical Research Institute, New York University, New York, New York10003, United States
| | - Briana Zeck
- Center
for Biospecimen Research and Development, NYU Langone, New York, New York 10016, United
States
| | - Luis Chiribogas
- Center
for Biospecimen Research and Development, NYU Langone, New York, New York 10016, United
States
| | - Dana Jackson
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Alex Herchen
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Tyson Attia
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Michael Carlock
- Center for
Vaccines and Immunology, University of Georgia, Athens, Georgia 30605, United States
| | - Amy Rapkiewicz
- Department
of Pathology, NYU Long Island School of
Medicine, Mineola, New York 11501, United
States
| | - Dafna Bar-Sagi
- Department
of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Bruce Ritchie
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Ted M. Ross
- Center for
Vaccines and Immunology, University of Georgia, Athens, Georgia 30605, United States
| | - Lara K. Mahal
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
18
|
Abel T, Moodley J, Khaliq OP, Naicker T. Vascular Endothelial Growth Factor Receptor 2: Molecular Mechanism and Therapeutic Potential in Preeclampsia Comorbidity with Human Immunodeficiency Virus and Severe Acute Respiratory Syndrome Coronavirus 2 Infections. Int J Mol Sci 2022; 23:ijms232213752. [PMID: 36430232 PMCID: PMC9691176 DOI: 10.3390/ijms232213752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
This review explored the role of vascular endothelial growth factor receptor-2 (VEGFR-2) in the synergy of preeclampsia (PE), human immunodeficiency virus (HIV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Downregulation of VEGFR-2 in PE promotes endothelial dysfunction and prevents endothelial cell (EC) migration, proliferation, and differentiation. The HIV-1 accessory protein, tat (trans-activator of transcription), prevents VEGFR-2 signaling via the vascular endothelial growth factor A (VEGF-A) ligand. Combined antiretroviral therapy (cART) may cause immune reconstitution, impaired decidualization, and endothelial injury, thus may be a risk factor for PE development. The VEGF/VEGFR-2 interaction may be associated with SARS-CoV-2-related pulmonary oedema. Endothelial dysfunction and heightened inflammation are both associated with PE, HIV, and SARS-CoV-2 infection; therefore, it is plausible that both characteristics may be exacerbated in the synergy of these events. In addition, this review explored microRNAs (miR) regulating VEGFR-2. An overexpression of miR-126 is evident in PE, HIV, and SARS-CoV-2 infection; thus, modulating the expression of miR-126 may be a therapeutic strategy. However, the involvement of microRNAs in PE, HIV, and SARS-CoV-2 infection needs further investigating. Since these conditions have been evaluated independently, this review attempts to predict their clinical manifestations in their synergy, as well as independently; thereby providing a platform for early diagnosis and therapeutic potential in PE, HIV, and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Tashlen Abel
- Women’s Health and HIV Research Group, Department of Obstetrics & Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Jagidesa Moodley
- Women’s Health and HIV Research Group, Department of Obstetrics & Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Olive P. Khaliq
- Department of Paediatrics and Child Health, Faculty of Health Sciences, The University of the Free State, Bloemfontein 9300, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institution, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
- Correspondence:
| |
Collapse
|
19
|
Naidoo N, Moodley J, Khaliq OP, Naicker T. Neuropilin-1 in the pathogenesis of preeclampsia, HIV-1, and SARS-CoV-2 infection: A review. Virus Res 2022; 319:198880. [PMID: 35905790 PMCID: PMC9316720 DOI: 10.1016/j.virusres.2022.198880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 12/25/2022]
Abstract
This review explores the role of transmembrane neuropilin-1 (NRP-1) in pregnancy, preeclampsia (PE), human immunodeficiency virus type 1 (HIV-1) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Since these conditions are assessed independently, this review attempts to predict their comorbid clinical manifestations. Dysregulation of NRP-1 contributes to the pathogenesis of PE by (a) impairing vascular endothelial growth factor (VEGF) signaling for adequate spiral artery remodeling and placentation, (b) inducing syncytiotrophoblast (ST) cell apoptosis and increasing ST-derived microparticle circulation and (c) by decreasing regulatory T cell activity predisposing maternal immune intolerance. Although NRP-1 is upregulated in SARS-CoV-2 placentae, its exploitation for SARS-CoV-2 internalization and increased infectivity may alter angiogenesis through the competitive inhibition of VEGF. The anti-inflammatory nature of NRP-1 may aid its upregulation in HIV-1 infection; however, the HIV-accessory protein, tat, reduces NRP-1 expression. Upregulated NRP-1 in macrophages and dendritic cells also demonstrated HIV-1 resistance/reduced infectivity. Notably, HIV-1-infected pregnant women receiving antiretroviral therapy (ART) to prevent vertical transmission may experience immune reconstitution, impaired decidualization, and elevated markers of endothelial injury. Since endothelial dysfunction and altered immune responses are central to PE, HIV-1 infection, ART usage and SARS-CoV-2 infection, it is plausible that an exacerbation of both features may prevail in the synergy of these events. Additionally, this review identifies microRNAs (miRNAs) mediating NRP-1 expression. MiR-320 and miR-141 are overexpressed in PE, while miR-206 and miR-124-3p showed increased expression in PE and HIV-1 infection. Additionally, miR-214 is overexpressed in PE, HIV-1 and SARS-CoV-2 infection, implicating treatment strategies to reduce these miRNAs to upregulate and normalize NRP-1 expression. However, inconsistencies in the data of the role and regulation of miRNAs in PE, HIV-1 and SARS-CoV-2 infections require clarification. This review provides a platform for early diagnosis and potential therapeutic intervention of PE, HIV-1, and SARS-CoV-2 infections independently and as comorbidities.
Collapse
Affiliation(s)
- Nitalia Naidoo
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Olive Pearl Khaliq
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
20
|
Wang Y, Hao Y, Hu M, Wang Y, Yang H. Interstitial lung disease independently associated with higher risk for COVID-19 severity and mortality: A meta-analysis of adjusted effect estimates. Int Immunopharmacol 2022; 111:109088. [PMID: 35921779 PMCID: PMC9325673 DOI: 10.1016/j.intimp.2022.109088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/13/2022] [Accepted: 07/22/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The aim of this study was to address the association between interstitial lung disease and the risk for severity and mortality among patients with coronavirus disease 2019 (COVID-19). METHODS The electronic databases of PubMed, Web of Science and EMBASE were systematically searched. The pooled effect size with 95 % confidence interval (CI) was computed by a random-effects meta-analysis model. Heterogeneity test, sensitivity analysis, subgroup analysis, meta-regression analysis, Begg's test and Egger's test were performed. RESULTS A total of sixteen eligible studies with 217,260 COVID-19 patients were enrolled in this meta-analysis. The findings based on adjusted effect estimates indicated that pre-existing interstitial lung disease was significantly associated with higher risk for COVID-19 severity (pooled effect = 1.34 [95 % CI: 1.16-1.55]) and mortality (pooled effect = 1.26 [95 % CI: 1.09-1.46]). Consistent results were observed in the subgroup analysis stratified by sample size, age, the percentage of male patients, study design, setting, the methods for adjustment and the factors for adjustment. The results of meta-regression demonstrated that sample size, age and region might be the potential sources of heterogeneity. Sensitivity analysis exhibited that our results were stable and robust. No publication bias was observed in Egger's test and Begg's test. CONCLUSION This meta-analysis on the basis of adjusted effect estimates demonstrated that pre-existing interstitial lung disease was independently associated with significantly higher risk for COVID-19 severity and mortality.
Collapse
Affiliation(s)
- Ying Wang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yuqing Hao
- International College of Zhengzhou University, Zhengzhou 450052, China
| | - Mengke Hu
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China
| | - Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou 450016, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
21
|
miR-142 Targets TIM-1 in Human Endothelial Cells: Potential Implications for Stroke, COVID-19, Zika, Ebola, Dengue, and Other Viral Infections. Int J Mol Sci 2022; 23:ijms231810242. [PMID: 36142146 PMCID: PMC9499484 DOI: 10.3390/ijms231810242] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/30/2022] Open
Abstract
T-cell immunoglobulin and mucin domain 1 (TIM-1) has been recently identified as one of the factors involved in the internalization of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in human cells, in addition to angiotensin-converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2), neuropilin-1, and others. We hypothesized that specific microRNAs could target TIM-1, with potential implications for the management of patients suffering from coronavirus disease 2019 (COVID-19). By combining bioinformatic analyses and functional assays, we identified miR-142 as a specific regulator of TIM-1 transcription. Since TIM-1 has been implicated in the regulation of endothelial function at the level of the blood-brain barrier (BBB) and its levels have been shown to be associated with stroke and cerebral ischemia-reperfusion injury, we validated miR-142 as a functional modulator of TIM-1 in human brain microvascular endothelial cells (hBMECs). Taken together, our results indicate that miR-142 targets TIM-1, representing a novel strategy against cerebrovascular disorders, as well as systemic complications of SARS-CoV-2 and other viral infections.
Collapse
|
22
|
Ketenci S, Uygar Kalaycı M, Dündar B, Duranay R, Şükrü Aynacıoğlu A. Elevated serum midkine levels in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infected patients. Int Immunopharmacol 2022; 110:108939. [PMID: 35717836 PMCID: PMC9181266 DOI: 10.1016/j.intimp.2022.108939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND The coronavirus disease-2019 (COVID-19) pandemic has caused important health, economic, social, and cultural problems worldwide. Recent findings demonstrate an excessive cytokine release during the disease development, especially in the seriously life-threatening form of COVID-19. Among other chemokines and cytokines that are released in high amounts at the infection site of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), midkine (MK), which is a potent pro-inflammatory growth factor/ cytokine, can be also overexpressed and contribute to the pathophysiological process in patients infected with SARS-CoV-2. MATERIALS AND METHOD Serum was collected from 87 intensive care unit (ICU) patients that are COVID-19 positive and 50 healthy volunteers in the control group with a negative PCR test and without disease symptoms. Circulating MK concentration was measured by enzyme-linked immunosorbent assay (ELISA). RESULTS COVID-19 patients had a significantly higher serum MK concentration compared to non-COVID-19 control subjects (1892.8 ± 1615.8 pg/mL versus 680.7 ± 907.6 pg/mL, respectively; P < 0.001). The cut-off MK concentration was 716.7 pg/ mL, with the sensitivity and specificity of 75.9 % and 76.0 %, respectively. The area under the receiver operating characteristic (ROC) curve of MK was = 0.827. Our findings showed that circulating MK levels are significantly increased in SARS-CoV-2 infected patients. CONCLUSION We suggest that MK is involved in the pathogenesis of COVID-19 and may be a part of hypercytokinaemia. Therefore, MK may serve as a supporting biomarker in the diagnosis of COVID-19, and blocking MK actions or its targets may attenuate the inflammatory process and the severity of the disease.
Collapse
Affiliation(s)
- Sema Ketenci
- Istanbul Atlas University, Faculty of Medicine, Department of Medical Pharmacology, Istanbul, Turkey
| | - M. Uygar Kalaycı
- Istanbul Atlas University, Faculty of Medicine, Department of General Surgery, Istanbul, Turkey
| | - Bağnu Dündar
- Istanbul Atlas University, Faculty of Medicine, Department of Biochemistry, Istanbul, Turkey
| | - Recep Duranay
- Istanbul Atlas University, Faculty of Engineering, Computer Engineering, Istanbul, Turkey
| | - A. Şükrü Aynacıoğlu
- Istanbul Atlas University, Faculty of Medicine, Department of Medical Pharmacology, Istanbul, Turkey,Corresponding author at: Istanbul Atlas University, Anadolu Cad. No: 40, Kağıthane, 34408 Istanbul, Turkey
| |
Collapse
|
23
|
Tang Q, Gao L, Tong Z, Li W. Hyperlipidemia, COVID-19 and acute pancreatitis: A tale of three entities. Am J Med Sci 2022; 364:257-263. [PMID: 35381217 PMCID: PMC8977370 DOI: 10.1016/j.amjms.2022.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/04/2022] [Accepted: 03/30/2022] [Indexed: 01/09/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the ongoing pandemic of coronavirus disease 2019 (COVID-19), which presented as not only respiratory symptoms, but various digestive manifestations including pancreatic injury and acute pancreatitis (AP). The underlying mechanism is still unclear. Hypertriglyceridemia has become one of the leading causes of AP in recent years and hyperlipidemia is highly reported in COVID-19 cases. The current narrative review aimed to explore the associations between AP, COVID-19 and hyperlipidemia. Substantial cases of COVID-19 patients complicated with AP were reported, while the incidence of AP in the COVID-19 population was relatively low. Hyperlipidemia was common in COVID-19 patients with a pooled incidence of 32.98%. Hyperlipidemia could be a mediating factor in the pathogenesis of AP in COVID-19 patients. Further studies are warranted to clarify the relationship among AP, lipid metabolism disorders and COVID-19.
Collapse
Affiliation(s)
- Qiuyi Tang
- Medical School, Southeast University, Nanjing, Jiangsu, China; Center of Severe Acute Pancreatitis (CSAP), Department of Intensive Care Unit, Jinling Hospital, Nanjing, Jiangsu, China
| | - Lin Gao
- Center of Severe Acute Pancreatitis (CSAP), Department of Intensive Care Unit, Jinling Hospital, Nanjing, Jiangsu, China
| | - Zhihui Tong
- Center of Severe Acute Pancreatitis (CSAP), Department of Intensive Care Unit, Jinling Hospital, Nanjing, Jiangsu, China.
| | - Weiqin Li
- Medical School, Southeast University, Nanjing, Jiangsu, China; Center of Severe Acute Pancreatitis (CSAP), Department of Intensive Care Unit, Jinling Hospital, Nanjing, Jiangsu, China.
| |
Collapse
|
24
|
Izzo R, Trimarco V, Mone P, Aloè T, Capra Marzani M, Diana A, Fazio G, Mallardo M, Maniscalco M, Marazzi G, Messina N, Mininni S, Mussi C, Pelaia G, Pennisi A, Santus P, Scarpelli F, Tursi F, Zanforlin A, Santulli G, Trimarco B. Combining L-Arginine with vitamin C improves long-COVID symptoms: The LINCOLN Survey. Pharmacol Res 2022; 183:106360. [PMID: 35868478 PMCID: PMC9295384 DOI: 10.1016/j.phrs.2022.106360] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Recent evidence suggests that oxidative stress and endothelial dysfunction play critical roles in the pathophysiology of COVID-19 and Long-COVID. We hypothesized that a supplementation combining L-Arginine (to improve endothelial function) and Vitamin C (to reduce oxidation) could have favorable effects on Long-COVID symptoms. METHODS We designed a survey (LINCOLN: L-Arginine and Vitamin C improves Long-COVID), assessing several symptoms that have been associated with Long-COVID to be administered nationwide to COVID-19 survivors; the survey also included effort perception, measured using the Borg scale. Patients receiving the survey were divided in two groups, with a 2:1 ratio: the first group included patients that received L-Arginine + Vitamin C, whereas the second group received a multivitamin combination (alternative treatment). RESULTS 1390 patients successfully completed the survey. Following a 30-day treatment in both groups, the survey revealed that patients in the L-Arginine + Vitamin C treatment arm had significantly lower scores compared to patients who had received the multivitamin combination. There were no other significant differences between the two groups. When examining effort perception, we observed a significantly lower value (p < 0.0001) in patients receiving L-Arginine + Vitamin C compared to the alternative-treatment arm. CONCLUSIONS Our survey indicates that the supplementation with L-Arginine + Vitamin C has beneficial effects in Long-COVID, in terms of attenuating its typical symptoms and improving effort perception.
Collapse
Affiliation(s)
- Raffaele Izzo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University, Naples, Italy
| | - Pasquale Mone
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | - Simone Mininni
- Associazione Scientifica Interdisciplinare Aggiornamento Medico (ASIAM), Florence, Italy
| | - Chiara Mussi
- Department of Biomedical and Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy
| | - Girolamo Pelaia
- Department of Health Science, Magna Graecia University, Catanzaro, Italy
| | | | | | | | | | | | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy; Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA.
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
25
|
Kell DB, Pretorius E. The potential role of ischaemia-reperfusion injury in chronic, relapsing diseases such as rheumatoid arthritis, Long COVID, and ME/CFS: evidence, mechanisms, and therapeutic implications. Biochem J 2022; 479:1653-1708. [PMID: 36043493 PMCID: PMC9484810 DOI: 10.1042/bcj20220154] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Ischaemia-reperfusion (I-R) injury, initiated via bursts of reactive oxygen species produced during the reoxygenation phase following hypoxia, is well known in a variety of acute circumstances. We argue here that I-R injury also underpins elements of the pathology of a variety of chronic, inflammatory diseases, including rheumatoid arthritis, ME/CFS and, our chief focus and most proximally, Long COVID. Ischaemia may be initiated via fibrin amyloid microclot blockage of capillaries, for instance as exercise is started; reperfusion is a necessary corollary when it finishes. We rehearse the mechanistic evidence for these occurrences here, in terms of their manifestation as oxidative stress, hyperinflammation, mast cell activation, the production of marker metabolites and related activities. Such microclot-based phenomena can explain both the breathlessness/fatigue and the post-exertional malaise that may be observed in these conditions, as well as many other observables. The recognition of these processes implies, mechanistically, that therapeutic benefit is potentially to be had from antioxidants, from anti-inflammatories, from iron chelators, and via suitable, safe fibrinolytics, and/or anti-clotting agents. We review the considerable existing evidence that is consistent with this, and with the biochemical mechanisms involved.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| |
Collapse
|
26
|
Spontaneous Haemothorax in a Patient with COVID-19. Case Rep Med 2022; 2022:8275326. [PMID: 35898334 PMCID: PMC9314003 DOI: 10.1155/2022/8275326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/30/2022] [Indexed: 02/07/2023] Open
Abstract
The global pandemic of COVID-19 is caused by SARS-CoV-2 virus. We continue to discover the wide spectrum of complications associated with COVID-19. Some well-known complications include pneumonia, acute respiratory distress syndrome, pneumothorax, disseminated intravascular coagulation (DIC), chronic fatigue, multiorgan dysfunction, and long COVID-19 syndrome. We report a rare case of a 51-year-old man with severe COVID-19 pneumonia who developed haemorrhagic shock secondary to spontaneous haemothorax after 17 days of hospitalisation. Clinicians should be aware of such occurrence, and hence, high clinical suspicion, prompt recognition of signs and symptoms of shock, and adequate resuscitation will improve the outcomes of patients.
Collapse
|
27
|
Zha D, Fu M, Qian Y. Vascular Endothelial Glycocalyx Damage and Potential Targeted Therapy in COVID-19. Cells 2022; 11:cells11121972. [PMID: 35741101 PMCID: PMC9221624 DOI: 10.3390/cells11121972] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/10/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19 is a highly infectious respiratory disease caused by a new coronavirus known as SARS-CoV-2. COVID-19 is characterized by progressive respiratory failure resulting from diffuse alveolar damage, inflammatory infiltrates, endotheliitis, and pulmonary and systemic coagulopathy forming obstructive microthrombi with multi-organ dysfunction, indicating that endothelial cells (ECs) play a central role in the pathogenesis of COVID-19. The glycocalyx is defined as a complex gel-like layer of glycosylated lipid–protein mixtures, which surrounds all living cells and acts as a buffer between the cell and the extracellular matrix. The endothelial glycocalyx layer (EGL) plays an important role in vascular homeostasis via regulating vascular permeability, cell adhesion, mechanosensing for hemodynamic shear stresses, and antithrombotic and anti-inflammatory functions. Here, we review the new findings that described EGL damage in ARDS, coagulopathy, and the multisystem inflammatory disease associated with COVID-19. Mechanistically, the inflammatory mediators, reactive oxygen species (ROS), matrix metalloproteases (MMPs), the glycocalyx fragments, and the viral proteins may contribute to endothelial glycocalyx damage in COVID-19. In addition, the potential therapeutic strategies targeting the EGL for the treatment of severe COVID-19 are summarized and discussed.
Collapse
Affiliation(s)
- Duoduo Zha
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China;
| | - Mingui Fu
- Shock/Trauma Research Center, Department of Biomedical Sciences, School of Medicine, University of Missouri Kansas City, Kansas City, MO 64108, USA;
| | - Yisong Qian
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China;
- Correspondence:
| |
Collapse
|
28
|
González-Ruiz FJ, Lazcano-Díaz EA, Baeza Herrera LA, Villalobos-Pedroza M, Toledo Alemán EL, Zuñiga-Salcedo MG, Cruz-Rodríguez C, López-Polanco A, Torres-Pulido A, Sierra-González de Cossio A, Cota Apodaca LA, Manzur-Sandoval D. Endotheliitis, Shunts, and Ventilation-Perfusion Mismatch in Coronavirus Disease 2019: A Literature Review of Disease Mechanisms. Ann Med Surg (Lond) 2022; 78:103820. [PMID: 35600188 PMCID: PMC9112604 DOI: 10.1016/j.amsu.2022.103820] [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: 03/27/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 10/27/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 pandemic has continued to impact global health. However, while immunity acquired by vaccines has been developed, 40% of the world's population has still not been vaccinated. Economic problems associated with acquiring novel therapies, misinformation, and differences in treatment protocols have generated catastrophic results, especially in low-resource countries. Understanding the pathophysiological aspects of coronavirus disease and the therapeutic strategies that have been validated to date is essential for successful medical care. In this review, I summarize the historical aspects of the virus, molecules involved in infecting the host, and consequences of viral interactions with and in tissues.
Collapse
Affiliation(s)
- Francisco J. González-Ruiz
- Department of Cardiovascular Critical Care, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, México
| | - Emmanuel A. Lazcano-Díaz
- Department of Cardiovascular Critical Care, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, México
| | - Luis A. Baeza Herrera
- Department of Cardiovascular Critical Care, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, México
| | | | - Enma L. Toledo Alemán
- Department of Cardiovascular Diseases, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, Mexico
| | - Miriam G. Zuñiga-Salcedo
- Department of Cardiovascular Diseases, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, Mexico
| | - Camelia Cruz-Rodríguez
- Department of Cardiovascular Critical Care, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, México
| | - Alexandra López-Polanco
- Department of Cardiovascular Critical Care, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, México
| | - Abraham Torres-Pulido
- Department of Cardiovascular Critical Care, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, México
| | | | - Luis A. Cota Apodaca
- Department of Cardiovascular Critical Care, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, México
| | - Daniel Manzur-Sandoval
- Department of Cardiovascular Critical Care, National Institute of Cardiology “Dr. Ignacio Chávez,”, Mexico City, México
| |
Collapse
|
29
|
Huntington KE, Carlsen L, So EY, Piesche M, Liang O, El-Deiry WS. Integrin/TGF-β1 Inhibitor GLPG-0187 Blocks SARS-CoV-2 Delta and Omicron Pseudovirus Infection of Airway Epithelial Cells In Vitro, Which Could Attenuate Disease Severity. Pharmaceuticals (Basel) 2022; 15:618. [PMID: 35631444 PMCID: PMC9143518 DOI: 10.3390/ph15050618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/15/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023] Open
Abstract
As COVID-19 continues to pose major risk for vulnerable populations, including the elderly, immunocompromised, patients with cancer, and those with contraindications to vaccination, novel treatment strategies are urgently needed. SARS-CoV-2 infects target cells via RGD-binding integrins, either independently or as a co-receptor with surface receptor angiotensin-converting enzyme 2 (ACE2). We used pan-integrin inhibitor GLPG-0187 to demonstrate the blockade of SARS-CoV-2 pseudovirus infection of target cells. Omicron pseudovirus infected normal human small airway epithelial (HSAE) cells significantly less than D614G or Delta variant pseudovirus, and GLPG-0187 effectively blocked SARS-CoV-2 pseudovirus infection in a dose-dependent manner across multiple viral variants. GLPG-0187 inhibited Omicron and Delta pseudovirus infection of HSAE cells more significantly than other variants. Pre-treatment of HSAE cells with MEK inhibitor (MEKi) VS-6766 enhanced the inhibition of pseudovirus infection by GLPG-0187. Because integrins activate transforming growth factor beta (TGF-β) signaling, we compared the plasma levels of active and total TGF-β in COVID-19+ patients. The plasma TGF-β1 levels correlated with age, race, and number of medications upon presentation with COVID-19, but not with sex. Total plasma TGF-β1 levels correlated with activated TGF-β1 levels. Moreover, the inhibition of integrin signaling prevents SARS-CoV-2 Delta and Omicron pseudovirus infectivity, and it may mitigate COVID-19 severity through decreased TGF-β1 activation. This therapeutic strategy may be further explored through clinical testing in vulnerable and unvaccinated populations.
Collapse
Affiliation(s)
- Kelsey E. Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (K.E.H.); (L.C.)
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02903, USA; (E.-Y.S.); (O.L.)
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Pathobiology Graduate Program, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Lindsey Carlsen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (K.E.H.); (L.C.)
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02903, USA; (E.-Y.S.); (O.L.)
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Pathobiology Graduate Program, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Eui-Young So
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02903, USA; (E.-Y.S.); (O.L.)
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Matthias Piesche
- Biomedical Research Laboratories, Medicine Faculty, Universidad Católica del Maule, Talca 3466706, Chile
- Oncology Center, Medicine Faculty, Universidad Católica del Maule, Talca 3466706, Chile
| | - Olin Liang
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02903, USA; (E.-Y.S.); (O.L.)
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA; (K.E.H.); (L.C.)
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02903, USA; (E.-Y.S.); (O.L.)
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Pathobiology Graduate Program, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| |
Collapse
|
30
|
Aggarwal R, Dewan A, Pandey A, Trehan N, Majid MA. Efficacy of high-dose intravenous immunoglobulin in severe and critical COVID-19: A retrospective cohort study. Int Immunopharmacol 2022; 106:108615. [PMID: 35168081 PMCID: PMC8825318 DOI: 10.1016/j.intimp.2022.108615] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/26/2022] [Accepted: 02/06/2022] [Indexed: 01/08/2023]
Abstract
Background Methods Results Conclusion
Collapse
Affiliation(s)
- Ritesh Aggarwal
- Department of Critical Care, Max Smart Super Speciality Hospital, New Delhi 110017, India.
| | - Arun Dewan
- Department of Critical Care, Max Smart Super Speciality Hospital, New Delhi 110017, India
| | - Ankita Pandey
- Department of Internal Medicine, Max Smart Super Speciality Hospital, New Delhi 110017, India
| | - Nikita Trehan
- Department of Critical Care, Max Smart Super Speciality Hospital, New Delhi 110017, India
| | - Muhammad Aamir Majid
- Department of Critical Care, Max Smart Super Speciality Hospital, New Delhi 110017, India
| |
Collapse
|
31
|
Gustafson D, Ngai M, Wu R, Hou H, Schoffel AC, Erice C, Mandla S, Billia F, Wilson MD, Radisic M, Fan E, Trahtemberg U, Baker A, McIntosh C, Fan CPS, Dos Santos CC, Kain KC, Hanneman K, Thavendiranathan P, Fish JE, Howe KL. Cardiovascular signatures of COVID-19 predict mortality and identify barrier stabilizing therapies. EBioMedicine 2022; 78:103982. [PMID: 35405523 PMCID: PMC8989492 DOI: 10.1016/j.ebiom.2022.103982] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Background Endothelial cell (EC) activation, endotheliitis, vascular permeability, and thrombosis have been observed in patients with severe coronavirus disease 2019 (COVID-19), indicating that the vasculature is affected during the acute stages of SARS-CoV-2 infection. It remains unknown whether circulating vascular markers are sufficient to predict clinical outcomes, are unique to COVID-19, and if vascular permeability can be therapeutically targeted. Methods Prospectively evaluating the prevalence of circulating inflammatory, cardiac, and EC activation markers as well as developing a microRNA atlas in 241 unvaccinated patients with suspected SARS-CoV-2 infection allowed for prognostic value assessment using a Random Forest model machine learning approach. Subsequent ex vivo experiments assessed EC permeability responses to patient plasma and were used to uncover modulated gene regulatory networks from which rational therapeutic design was inferred. Findings Multiple inflammatory and EC activation biomarkers were associated with mortality in COVID-19 patients and in severity-matched SARS-CoV-2-negative patients, while dysregulation of specific microRNAs at presentation was specific for poor COVID-19-related outcomes and revealed disease-relevant pathways. Integrating the datasets using a machine learning approach further enhanced clinical risk prediction for in-hospital mortality. Exposure of ECs to COVID-19 patient plasma resulted in severity-specific gene expression responses and EC barrier dysfunction, which was ameliorated using angiopoietin-1 mimetic or recombinant Slit2-N. Interpretation Integration of multi-omics data identified microRNA and vascular biomarkers prognostic of in-hospital mortality in COVID-19 patients and revealed that vascular stabilizing therapies should be explored as a treatment for endothelial dysfunction in COVID-19, and other severe diseases where endothelial dysfunction has a central role in pathogenesis. Funding Information This work was directly supported by grant funding from the Ted Rogers Center for Heart Research, Toronto, Ontario, Canada and the Peter Munk Cardiac Center, Toronto, Ontario, Canada.
Collapse
Affiliation(s)
- Dakota Gustafson
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Michelle Ngai
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Ruilin Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Huayun Hou
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | | | - Clara Erice
- Johns Hopkins School of Medicine, Baltimore, USA
| | - Serena Mandla
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Filio Billia
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Michael D Wilson
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Eddy Fan
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Interdepartmental Division of Critical Care and Institute of Medical Sciences, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Uriel Trahtemberg
- Keenan Research Center for Biomedical Research, Unity Health Toronto, Toronto, Canada; Critical Care Department, Galilee Medical Center, Nahariya, Israel
| | - Andrew Baker
- Interdepartmental Division of Critical Care and Institute of Medical Sciences, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Critical Care Department, Galilee Medical Center, Nahariya, Israel
| | - Chris McIntosh
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada; Techna Institute, University Health Network, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Vector Institute, University of Toronto, Toronto, Canada
| | - Chun-Po S Fan
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada
| | - Claudia C Dos Santos
- Interdepartmental Division of Critical Care and Institute of Medical Sciences, University of Toronto, Toronto, Canada; Keenan Research Center for Biomedical Research, Unity Health Toronto, Toronto, Canada
| | - Kevin C Kain
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Kate Hanneman
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada
| | - Paaladinesh Thavendiranathan
- Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Joint Department of Medical Imaging, University Health Network, University of Toronto, Toronto, Canada; Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, Toronto, Canada
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada.
| | - Kathryn L Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, Canada.
| |
Collapse
|
32
|
Anti-COVID-19 Potential of Azadirachta indica (Neem) Leaf Extract. SCIENTIFIC AFRICAN 2022; 16:e01184. [PMID: 35434432 PMCID: PMC8990437 DOI: 10.1016/j.sciaf.2022.e01184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/24/2021] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is caused by infection with the “severe acute respiratory syndrome coronavirus-2″ (i.e., SARS-CoV-2). This is an enveloped virus having a positive sense, single-stranded RNA genome; like the two earlier viruses SARS-CoV and the Middle East respiratory syndrome (MERS) virus. COVID-19 is unique in that, in the severe case, it has the propensity to affect multiple organs, leading to multiple organ distress syndrome (MODS), and causing high morbidity and mortality in the extreme case. In addition, comorbidities like age, cardiovascular disease, diabetes and its complications, obesity, are risk factors for severe COVID-19. It turns out that a most plausible, simple, single explanation for this propensity for MODS is the pivotal involvement of the vascular endothelium (VE). This is a consequence of the fact that the VE seamlessly connects all the entire vascular bed in the body, thus linking all the target organs (heart, lungs, kidney, liver, brain) and systems. Infection with SARS-CoV-2 leads to hyper-inflammation yielding uncontrolled production of a mixture of cytokines, chemokines, reactive oxygen species, nitric oxide, oxidative stress, acute phase proteins (e.g., C-reactive protein), and other pro-inflammatory substances. In the extreme case, a cytokine storm is created. Displacement of the virus bound to the VE, and/or inhibition of binding of the virus, would constitute an effective strategy for preventing COVID-19. In this regard, the acetone-water extract of the leaf of the Neem (Azadirachta indica) plant has been known to prevent the adherence of malaria parasitized red blood cells (pRBCs) to VE; prevent cytoadherence of cancer cells in metastasis; and prevent HIV from invading target T lymphocytes. We therefore hypothesize that this Neem leaf acetone-water extract will prevent the binding of SARS-CoV-2 to the VE, and therefore be an effective therapeutic formulation against COVID-19. It is therefore advocated herein that this extract be investigated through rigorous clinical trials for this purpose. It has the advantages of being (i) readily available, and renewable in favor of the populations positioned to benefit from it; (ii) simple to prepare; and (iii) devoid of any detectable toxicity.
Collapse
|
33
|
Zanoli L, Gaudio A, Mikhailidis DP, Katsiki N, Castellino N, Lo Cicero L, Geraci G, Sessa C, Fiorito L, Marino F, Antonietta Di Rosolini M, Colaci M, Longo A, Montineri A, Malatino L, Castellino P, Aparo P, Arena A, Barchitta M, Castelletti F, Noto MD, Pino AD, Giarrusso O, Isaia I, Lentini P, Magnano San Lio P, Manuele R, Marino E, Morale W, Sciuto A, Scuto SS, Xourafa A, Zocco S. Vascular Dysfunction of COVID-19 Is Partially Reverted in the Long-Term. Circ Res 2022; 130:1276-1285. [DOI: 10.1161/circresaha.121.320460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
COVID-19 is characterized by severe inflammation during the acute phase and increased aortic stiffness in the early postacute phase. In other models, aortic stiffness is improved after the reduction of inflammation. We aimed to evaluate the mid- and long-term effects of COVID-19 on vascular and cardiac autonomic function. The primary outcome was aortic pulse wave velocity (aPWV).
Methods:
The cross-sectional Study-1 included 90 individuals with a history of COVID-19 and 180 matched controls. The longitudinal Study-2 included 41 patients with COVID-19 randomly selected from Study-1 who were followed-up for 27 weeks.
Results:
Study-1: Compared with controls, patients with COVID-19 had higher aPWV and brachial PWV 12 to 24 (but not 25–48) weeks after COVID-19 onset, and they had higher carotid Young’s elastic modulus and lower distensibility 12 to 48 weeks after COVID-19 onset. In partial least squares structural equation modeling, the higher the hs-CRP (high-sensitivity C-reactive protein) at hospitalization was, the higher the aPWV 12 to 48 weeks from COVID-19 onset (path coefficient: 0.184;
P
=0.04). Moreover, aPWV (path coefficient: −0.186;
P
=0.003) decreased with time. Study-2: mean blood pressure and carotid intima-media thickness were comparable at the end of follow-up, whereas aPWV (−9%;
P
=0.01), incremental Young’s elastic modulus (−17%;
P
=0.03), baroreflex sensitivity (+28%;
P
=0.049), heart rate variability triangular index (+15%;
P
=0.01), and subendocardial viability ratio (+12%;
P
=0.01×10
−4
) were significantly improved. There was a trend toward improvement in brachial PWV (−6%;
P
=0.14) and carotid distensibility (+18%;
P
=0.05). Finally, at the end of follow-up (48 weeks after the onset of COVID-19) aPWV (+6%;
P
=0.04) remained significantly higher in patients with COVID-19 than in control subjects.
Conclusions:
COVID-19-related arterial stiffening involves several arterial tree portions and is partially resolved in the long-term.
Collapse
Affiliation(s)
- Luca Zanoli
- Department of Clinical and Experimental Medicine, University of Catania, Italy. (L.Z., A.G., L.LC., L.F., M.C., L.M., P.C.)
| | - Agostino Gaudio
- Department of Clinical and Experimental Medicine, University of Catania, Italy. (L.Z., A.G., L.LC., L.F., M.C., L.M., P.C.)
| | - Dimitri P. Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital campus, University College London, United Kingdom (D.P.M.)
| | - Niki Katsiki
- Diabetes Center, Division of Endocrinology and Metabolism, First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki, Greece (N.K.)
| | | | - Lorenzo Lo Cicero
- Department of Clinical and Experimental Medicine, University of Catania, Italy. (L.Z., A.G., L.LC., L.F., M.C., L.M., P.C.)
| | - Giulio Geraci
- Internal Medicine, St Elia Hospital, Caltanissetta, Italy (G.G.)
| | - Concetto Sessa
- Nephrology and Dialysis Unit, Giovanni Paolo II Hospital, Ragusa, Italy (C.S.)
| | - Letizia Fiorito
- Department of Clinical and Experimental Medicine, University of Catania, Italy. (L.Z., A.G., L.LC., L.F., M.C., L.M., P.C.)
| | - Francesca Marino
- Infectious Diseases, Giovanni Paolo II Hospital, Ragusa, Italy (F.M., M.A.D.R.)
| | | | - Michele Colaci
- Department of Clinical and Experimental Medicine, University of Catania, Italy. (L.Z., A.G., L.LC., L.F., M.C., L.M., P.C.)
| | - Antonio Longo
- Eye Clinic, University of Catania, Italy. (N.C., A.L.)
| | | | - Lorenzo Malatino
- Department of Clinical and Experimental Medicine, University of Catania, Italy. (L.Z., A.G., L.LC., L.F., M.C., L.M., P.C.)
| | - Pietro Castellino
- Department of Clinical and Experimental Medicine, University of Catania, Italy. (L.Z., A.G., L.LC., L.F., M.C., L.M., P.C.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Xu C, Chen Y, Yu J. Foe and friend in the COVID-19-associated acute kidney injury: an insight on intrarenal renin-angiotensin system. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1-11. [PMID: 35130610 PMCID: PMC9828085 DOI: 10.3724/abbs.2021002] [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] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/08/2021] [Indexed: 01/08/2023] Open
Abstract
Since the first reported case in December of 2019, the coronavirus disease 2019 (COVID-19) has became an international public health emergency. So far, there are more than 228,206,384 confirmed cases including 4,687,066 deaths. Kidney with high expression of angiotensin-converting enzyme 2 (ACE2) is one of the extrapulmonary target organs affected in patients with COVID-19. Acute kidney injury (AKI) is one of the independent risk factors for the death of COVID-19 patients. The imbalance between ACE2-Ang(1-7)-MasR and ACE-Ang II-AT1R axis in the kidney may contribute to COVID-19-associated AKI. Although series of research have shown the inconsistent effects of multiple common RAS inhibitors on ACE2 expression and enzyme activity, most of the retrospective cohort studies indicated the safety and protective effects of ACEI/ARB in COVID-19 patients. This review article highlights the current knowledge on the possible involvement of intrarenal RAS in COVID-19-associated AKI with a primary focus on the opposing effects of ACE2-Ang(1-7)-MasR and ACE-Ang II-AT1R signaling in the kidney. Human recombinant soluble ACE2 or ACE2 variants with preserved ACE2-enzymatic activity may be the best options to improve COVID-19-associated AKI.
Collapse
Affiliation(s)
- Chuanming Xu
- Translational Medicine CentreJiangxi University of Chinese MedicineNanchang330002China
| | - Yanting Chen
- Institute of HypertensionSun Yat-sen University School of MedicineGuangzhou510080China
| | - Jun Yu
- Center for Metabolic Disease Research and Department of PhysiologyLewis Katz School of MedicineTemple UniversityPhiladelphiaPA19140USA
| |
Collapse
|
35
|
Geronikolou SA, Takan I, Pavlopoulou A, Mantzourani M, Chrousos GP. Thrombocytopenia in COVID‑19 and vaccine‑induced thrombotic thrombocytopenia. Int J Mol Med 2022; 49:35. [PMID: 35059730 PMCID: PMC8815408 DOI: 10.3892/ijmm.2022.5090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
The highly heterogeneous symptomatology and unpredictable progress of COVID-19 triggered unprecedented intensive biomedical research and a number of clinical research projects. Although the pathophysiology of the disease is being progressively clarified, its complexity remains vast. Moreover, some extremely infrequent cases of thrombotic thrombocytopenia following vaccination against SARS-CoV-2 infection have been observed. The present study aimed to map the signaling pathways of thrombocytopenia implicated in COVID-19, as well as in vaccine-induced thrombotic thrombocytopenia (VITT). The biomedical literature database, MEDLINE/PubMed, was thoroughly searched using artificial intelligence techniques for the semantic relations among the top 50 similar words (>0.9) implicated in COVID-19-mediated human infection or VITT. Additionally, STRING, a database of primary and predicted associations among genes and proteins (collected from diverse resources, such as documented pathway knowledge, high-throughput experimental studies, cross-species extrapolated information, automated text mining results, computationally predicted interactions, etc.), was employed, with the confidence threshold set at 0.7. In addition, two interactomes were constructed: i) A network including 119 and 56 nodes relevant to COVID-19 and thrombocytopenia, respectively; and ii) a second network containing 60 nodes relevant to VITT. Although thrombocytopenia is a dominant morbidity in both entities, three nodes were observed that corresponded to genes (AURKA, CD46 and CD19) expressed only in VITT, whilst ADAM10, CDC20, SHC1 and STXBP2 are silenced in VITT, but are commonly expressed in both COVID-19 and thrombocytopenia. The calculated average node degree was immense (11.9 in COVID-19 and 6.43 in VITT), illustrating the complexity of COVID-19 and VITT pathologies and confirming the importance of cytokines, as well as of pathways activated following hypoxic events. In addition, PYCARD, NLP3 and P2RX7 are key potential therapeutic targets for all three morbid entities, meriting further research. This interactome was based on wild-type genes, revealing the predisposition of the body to hypoxia-induced thrombosis, leading to the acute COVID-19 phenotype, the 'long-COVID syndrome', and/or VITT. Thus, common nodes appear to be key players in illness prevention, progression and treatment.
Collapse
Affiliation(s)
- Styliani A Geronikolou
- Clinical, Translational and Experimental Surgery Research Centre, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | - Işil Takan
- Izmir Biomedicine and Genome Center (IBG), 35340 Izmir, Turkey
| | | | - Marina Mantzourani
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece
| | - George P Chrousos
- Clinical, Translational and Experimental Surgery Research Centre, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
36
|
Tarnawski AS, Ahluwalia A. Endothelial cells and blood vessels are major targets for COVID-19-induced tissue injury and spreading to various organs. World J Gastroenterol 2022; 28:275-289. [PMID: 35110950 PMCID: PMC8771611 DOI: 10.3748/wjg.v28.i3.275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/02/2021] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) infected so far over 250 million people and caused the death of over 5 million worldwide. Aging, diabetes, and cardiovascular diseases, conditions with preexisting impaired endothelial functions predispose to COVID-19. While respiratory epithelium is the main route of virus entry, the endothelial cells (ECs) lining pulmonary blood vessels are also an integral part of lung injury in COVID-19 patients. COVID-19 not only affects the lungs and respiratory system but also gastrointestinal (GI) tract, liver, pancreas, kidneys, heart, brain, and skin. Blood vessels are likely conduits for the virus dissemination to these distant organs. Importantly, ECs are also critical for vascular regeneration during injury/lesions healing and restoration of vascular network. The World Journal of Gastroenterology has published in last two years over 67 outstanding papers on COVID-19 infection with a focus on the GI tract, liver, pancreas, etc., however, the role of the endothelial and vascular components as major targets for COVID-19-induced tissue injury, spreading to various organs, and injury healing have not been sufficiently emphasized. In the present article, we focus on these subjects and on current treatments including the most recent oral drugs molnupiravir and paxlovid that show a dramatic, significant efficacy in controlling severe COVID-19 infection.
Collapse
Affiliation(s)
- Andrzej S Tarnawski
- Gastroenterology Research Department, University of California Irvine and the Veterans Administration Long Beach Healthcare System, Long Beach, CA 90822, United States
| | - Amrita Ahluwalia
- Research Service, Veterans Administration Long Beach Healthcare System, Long Beach, CA 90822, United States
| |
Collapse
|
37
|
Al Mahmeed W, Al-Rasadi K, Banerjee Y, Ceriello A, Cosentino F, Galia M, Goh SY, Kempler P, Lessan N, Papanas N, Rizvi AA, Santos RD, Stoian AP, Toth PP, Rizzo M. Promoting a Syndemic Approach for Cardiometabolic Disease Management During COVID-19: The CAPISCO International Expert Panel. Front Cardiovasc Med 2022; 8:787761. [PMID: 34977193 PMCID: PMC8715947 DOI: 10.3389/fcvm.2021.787761] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/22/2021] [Indexed: 12/14/2022] Open
Abstract
Efforts in the fight against COVID-19 are achieving success in many parts of the world, although progress remains slow in other regions. We believe that a syndemic approach needs to be adopted to address this pandemic given the strong apparent interplay between COVID-19, its related complications, and the socio-structural environment. We have assembled an international, multidisciplinary group of researchers and clinical practitioners to promote a novel syndemic approach to COVID-19: the CArdiometabolic Panel of International experts on Syndemic COvid-19 (CAPISCO). This geographically diverse group aims to facilitate collaborative-networking and scientific exchanges between researchers and clinicians facing a multitude of challenges on different continents during the pandemic. In the present article we present our “manifesto”, with the intent to provide evidence-based guidance to the global medical and scientific community for better management of patients both during and after the current pandemic.
Collapse
Affiliation(s)
- Wael Al Mahmeed
- Cleveland Clinic, Heart and Vascular Institute, Abu Dhabi, United Arab Emirates
| | | | - Yajnavalka Banerjee
- Department of Biochemistry, Mohamed Bin Rashid University, Dubai, United Arab Emirates
| | | | - Francesco Cosentino
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, University of Stockholm, Stockholm, Sweden
| | - Massimo Galia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (Bind), University of Palermo, Palermo, Italy
| | - Su-Yen Goh
- Department of Endocrinology, Singapore General Hospital, Singapore, Singapore
| | - Peter Kempler
- Department of Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Nader Lessan
- Imperial College London Diabetes Centre, The Research Institute, Abu Dhabi, United Arab Emirates
| | - Nikolaos Papanas
- Second Department of Internal Medicine, Diabetes Center, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ali A Rizvi
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL, United States.,Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, IN, United States
| | - Raul D Santos
- Heart Institute (InCor) University of Sáo Paulo Medical School Hospital, Sáo Paulo, Brazil.,Hospital Israelita Albert Einstein, Sáo Paulo, Brazil
| | - Anca P Stoian
- Faculty of Medicine, Diabetes, Nutrition and Metabolic Diseases, Carol Davila University, Bucharest, Romania
| | - Peter P Toth
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Manfredi Rizzo
- Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, IN, United States.,Faculty of Medicine, Diabetes, Nutrition and Metabolic Diseases, Carol Davila University, Bucharest, Romania.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | | |
Collapse
|
38
|
Shen L, Qiu L, Wang L, Huang H, Liu D, Xiao Y, Liu Y, Jin J, Liu X, Wang DW, He B, Zhou N. Statin Use and In-hospital Mortality in Patients with COVID-19 and Coronary Heart Disease. Sci Rep 2021; 11:23874. [PMID: 34903765 PMCID: PMC8668894 DOI: 10.1038/s41598-021-02534-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/30/2021] [Indexed: 02/08/2023] Open
Abstract
The worsening progress of coronavirus disease 2019 (COVID-19) is attributed to the proinflammatory state, leading to increased mortality. Statin works with its anti-inflammatory effects and may attenuate the worsening of COVID-19. COVID-19 patients were retrospectively enrolled from two academic hospitals in Wuhan, China, from 01/26/2020 to 03/26/2020. Adjusted in-hospital mortality was compared between the statin and the non-statin group by CHD status using multivariable Cox regression model after propensity score matching. Our study included 3133 COVID-19 patients (median age: 62y, female: 49.8%), and 404 (12.9%) received statin. Compared with the non-statin group, the statin group was older, more likely to have comorbidities but with a lower level of inflammatory markers. The Statin group also had a lower adjusted mortality risk (6.44% vs. 10.88%; adjusted hazard ratio [HR] 0.47; 95% CI, 0.29–0.77). Subgroup analysis of CHD patients showed a similar result. Propensity score matching showed an overall 87% (HR, 0.13; 95% CI, 0.05–0.36) lower risk of in-hospital mortality for statin users than nonusers. Such survival benefit of statin was obvious both among CHD and non-CHD patients (HR = 0.30 [0.09–0.98]; HR = 0.23 [0.1–0.49], respectively). Statin use was associated with reduced in-hospital mortality in COVID-19. The benefit of statin was both prominent among CHD and non-CHD patients. These findings may further reemphasize the continuation of statins in patients with CHD during the COVID-19 era.
Collapse
Affiliation(s)
- Lan Shen
- Department of Cardiology, Clinical Research Unit, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Lin Qiu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Li Wang
- Department of Geriatrics, School of Medicine, Shanghai Renji Hospital, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Hengye Huang
- School of Public Health, School of Medicine, Shanghai Jiaotong University, Shanghai, 200025, China
| | - Dong Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Ying Xiao
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Yi Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Jingjin Jin
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Xiulan Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China
| | - Ben He
- Department of Cardiology, Clinical Research Unit, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, 200030, China.
| | - Ning Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, China.
| |
Collapse
|
39
|
Sefotho MM. Unsettling transdisciplinary perspectives on sustainability issues: A critical discourse (metadata) cross-case analysis. JOURNAL OF PSYCHOLOGY IN AFRICA 2021. [DOI: 10.1080/14330237.2021.1982505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Maximus Monaheng Sefotho
- Department of Educational Psychology, Faculty of Education, University of Johannesburg, South Africa
| |
Collapse
|
40
|
Bargehr J, Rericha P, Petchey A, Colzani M, Moule G, Malgapo MC, Rassl D, Tarkin J, Mellor G, Sampaziotis F, Brevini T, Gambardella L, Bennett MR, Sinha S. Cardiovascular ACE2 receptor expression in patients undergoing heart transplantation. ESC Heart Fail 2021; 8:4119-4129. [PMID: 34390216 PMCID: PMC8497226 DOI: 10.1002/ehf2.13528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 06/12/2021] [Accepted: 07/05/2021] [Indexed: 01/19/2023] Open
Abstract
AIMS Membrane-bound angiotensin-converting enzyme (ACE)2 is the main cellular access point for SARS-CoV-2, but its expression and the effect of ACE inhibition have not been assessed quantitatively in patients with heart failure. The aim of this study was to characterize membrane-bound ACE2 expression in the myocardium and myocardial vasculature in patients undergoing heart transplantation and to assess the effect of pharmacological ACE inhibition. METHODS AND RESULTS Left ventricular (LV) tissue was obtained from 36 explanted human hearts from patients undergoing heart transplantation. Immunohistochemical staining with antibodies directed against ACE2 co-registered with cardiac troponin T (cTnT) and α-smooth muscle cell actin (SMA) was performed across the entire cohort. ACE2 receptor expression was quantitatively assessed throughout the myocardium and vasculature. ACE2 was consistently expressed throughout the LV myocardium (28.3% ± 22.2% of cardiomyocytes). ACE2 expression was also detected in small calibre blood vessels (range, 2-9 μm), albeit at quantitatively much lower levels (5% ± 9% of blood vessels). There was no significant difference in ACE2 expression between patients receiving ACE inhibitors prior to transplantation and ACE inhibitor-negative controls (P > 0.05). ACE2 expression did not differ significantly between the different diagnostic groups as the underlying reason for heart transplantation (ANOVA > 0.05). N-terminal pro-brain natriuretic peptide (NT-proBNP) (R2 = 0.37, P = 0.0006) and pulmonary capillary wedge pressure (PCWP) (R2 = 0.13, P = 0.043) assessed by right heart catheterization were significantly correlated with greater ACE2 expression in cardiomyocytes. CONCLUSIONS These data provide a comprehensive characterization of membrane-bound cardiac ACE2 expression in patients with heart failure with no demonstrable effect exerted by ACE inhibitors.
Collapse
Affiliation(s)
- Johannes Bargehr
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
- Division of Cardiovascular MedicineUniversity of CambridgeCambridgeUK
| | - Patrick Rericha
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
- Division of Cardiovascular MedicineUniversity of CambridgeCambridgeUK
| | - Alex Petchey
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
- Division of Cardiovascular MedicineUniversity of CambridgeCambridgeUK
| | - Maria Colzani
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
- Division of Cardiovascular MedicineUniversity of CambridgeCambridgeUK
| | - Georgia Moule
- Department of HistopathologyRoyal Papworth HospitalCambridgeUK
| | | | - Doris Rassl
- Department of HistopathologyRoyal Papworth HospitalCambridgeUK
| | - Jason Tarkin
- Division of Cardiovascular MedicineUniversity of CambridgeCambridgeUK
| | - Greg Mellor
- Cardiology DepartmentRoyal Papworth HospitalCambridgeUK
| | - Fotis Sampaziotis
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
- Department of HepatologyCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Teresa Brevini
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
| | - Laure Gambardella
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
- Division of Cardiovascular MedicineUniversity of CambridgeCambridgeUK
| | - Martin R. Bennett
- Division of Cardiovascular MedicineUniversity of CambridgeCambridgeUK
| | - Sanjay Sinha
- Wellcome – MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical CampusUniversity of CambridgeCambridgeUK
- Division of Cardiovascular MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
41
|
Ouyang L, Gong J, Yu M. Pre-existing interstitial lung disease in patients with coronavirus disease 2019: A meta-analysis. Int Immunopharmacol 2021; 100:108145. [PMID: 34547678 PMCID: PMC8450148 DOI: 10.1016/j.intimp.2021.108145] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/25/2021] [Accepted: 09/06/2021] [Indexed: 01/15/2023]
Abstract
Background The impact of pre-existing interstitial lung disease (ILD) on the severity and mortality of COVID-19 remains largely unknown. The purpose of this meta-analysis was to investigate the prevalence of ILD among patients with COVID-19 and figure out the relationship between ILD and the poor clinical outcomes of COVID-19. Methods A systematic literature search was conducted in the PubMed, EMBASE, Web of Science and MedRxiv Database from 1 January 2020 to 26 May 2021. Results 15 studies with 135,263 COVID-19 patients were included for analysis of ILD prevalence. The pooled prevalence of comorbid ILD in patients with COVID-19 was 1.4% (95% CI, 1.1%-1.8%, I2 = 91%) with significant between-study heterogeneity. Moreover, the prevalence of ILD in non-survival patients with COVID-19 was 2.728-folds higher than that in corresponding survival patients (RR = 2.728, 95% CI 1.162–6.408, I2 = 54%, p = 0.021). Additionally, 2–3 studies were included for comparison analysis of clinical outcome between COVID-19 patients with and without ILD. The results showed that the mortality of COVID-19 patients with ILD was remarkably elevated compared with patients without ILD (RR = 2.454, 95% CI 1.111–5.421, I2 = 87%, p = 0.026). Meanwhile, the pooled RR of ICU admission for ILD vs. non-ILD cases with COVID-19 was 3.064 (95% CI 1.889–4.972, I2 = 0, p < 0.0001). No significant difference in utilizing rate of mechanical ventilation was observed between COVID-19 patients with and without ILD. Conclusions There is great variability in ILD prevalence among patients with COVID-19 across the globe. Pre-existing ILD is associated with higher severity and mortality of COVID-19.
Collapse
Affiliation(s)
- Lichen Ouyang
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, China.
| | - Jie Gong
- Department of Anesthesiology, Union Hospital, Tongji Medical College, HuazhongUniversity of Science and Technology, Wuhan, China; The Clinical Skill Center, The First Clinical College, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Muqing Yu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
42
|
García de Guadiana-Romualdo L, Martínez Martínez M, Rodríguez Mulero MD, Esteban-Torrella P, Hernández Olivo M, Alcaraz García MJ, Campos-Rodríguez V, Sancho-Rodríguez N, Galindo Martínez M, Alcaraz A, Ros Braquehais MS, Báguena Perez-Crespo C, Ramos Arenas V, Tomás Jiménez C, Consuegra-Sánchez L, Conesa-Hernandez A, Piñera-Salmerón P, Bernal-Morell E. Circulating MR-proADM levels, as an indicator of endothelial dysfunction, for early risk stratification of mid-term mortality in COVID-19 patients. Int J Infect Dis 2021; 111:211-218. [PMID: 34461254 PMCID: PMC8400460 DOI: 10.1016/j.ijid.2021.08.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/23/2022] Open
Abstract
Objectives Thromboinflammation, resulting from a complex interaction between thrombocytopathy, coagulopathy, and endotheliopathy, contributes to increased mortality in COVID-19 patients. MR-proADM, as a surrogate of adrenomedullin system disruption, leading to endothelial damage, has been reported as a promising biomarker for short-term prognosis. We evaluated the role of MR-proADM in the mid-term mortality in COVID-19 patients. Methods A prospective, observational study enrolling COVID-19 patients from August to October 2020. A blood sample for laboratory test analysis was drawn on arrival in the emergency department. The primary endpoint was 90-day mortality. The area under the curve (AUC) and Cox regression analyses were used to assess discriminatory ability and association with the endpoint. Results A total of 359 patients were enrolled, and the 90-day mortality rate was 8.9%. ROC AUC for MR-proADM predicting 90-day mortality was 0.832. An optimal cutoff of 0.80 nmol/L showed a sensitivity of 96.9% and a specificity of 58.4%, with a negative predictive value of 99.5%. Circulating MR-proADM levels (inverse transformed), after adjusting by a propensity score including eleven potential confounders, were an independent predictor of 90-day mortality (HR: 0.162 [95% CI: 0.043-0.480]) Conclusions Our data confirm that MR-proADM has a role in the mid-term prognosis of COVID-19 patients and might assist physicians with risk stratification.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Antonia Alcaraz
- Infectious Disease Unit, Hospital General Universitario Reina Sofía, Murcia, Spain
| | | | | | - Verónica Ramos Arenas
- Laboratory Medicine Department, Hospital Universitario Santa Lucía, Cartagena, Spain
| | | | | | | | | | - Enrique Bernal-Morell
- Infectious Disease Unit, Hospital General Universitario Reina Sofía, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB).
| |
Collapse
|
43
|
Neutrophil Extracellular Traps (NETs) in Severe SARS-CoV-2 Lung Disease. Int J Mol Sci 2021; 22:ijms22168854. [PMID: 34445556 PMCID: PMC8396177 DOI: 10.3390/ijms22168854] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
Neutrophil extracellular traps (NETs), built from mitochondrial or nuclear DNA, proteinases, and histones, entrap and eliminate pathogens in the course of bacterial or viral infections. Neutrophils’ activation and the formation of NETs have been described as major risk factors for acute lung injury, multi-organ damage, and mortality in COVID-19 disease. NETs-related lung injury involves both epithelial and endothelial cells, as well as the alveolar-capillary barrier. The markers for NETs formation, such as circulating DNA, neutrophil elastase (NE) activity, or myeloperoxidase-DNA complexes, were found in lung specimens of COVID-19 victims, as well as in sera and tracheal aspirates obtained from COVID-19 patients. DNA threads form large conglomerates causing local obstruction of the small bronchi and together with NE are responsible for overproduction of mucin by epithelial cells. Various components of NETs are involved in the pathogenesis of cytokine storm in SARS-CoV-2 pulmonary disease. NETs are responsible for the interplay between inflammation and thrombosis in the affected lungs. The immunothrombosis, stimulated by NETs, has a poor prognostic significance. Better understanding of the role of NETs in the course of COVID-19 can help to develop novel approaches to the therapeutic interventions in this condition.
Collapse
|
44
|
Nicosia RF, Ligresti G, Caporarello N, Akilesh S, Ribatti D. COVID-19 Vasculopathy: Mounting Evidence for an Indirect Mechanism of Endothelial Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1374-1384. [PMID: 34033751 PMCID: PMC8141344 DOI: 10.1016/j.ajpath.2021.05.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
Patients with coronavirus disease 2019 (COVID-19) who are critically ill develop vascular complications characterized by thrombosis of small, medium, and large vessels. Dysfunction of the vascular endothelium due to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been implicated in the pathogenesis of the COVID-19 vasculopathy. Although initial reports suggested that endothelial injury was caused directly by the virus, recent studies indicate that endothelial cells do not express angiotensin-converting enzyme 2, the receptor that SARS-CoV-2 uses to gain entry into cells, or express it at low levels and are resistant to the infection. These new findings, together with the observation that COVID-19 triggers a cytokine storm capable of injuring the endothelium and disrupting its antithrombogenic properties, favor an indirect mechanism of endothelial injury mediated locally by an augmented inflammatory reaction to infected nonendothelial cells, such as the bronchial and alveolar epithelium, and systemically by the excessive immune response to infection. Herein we review the vascular pathology of COVID-19 and critically discuss the potential mechanisms of endothelial injury in this disease.
Collapse
Affiliation(s)
- Roberto F Nicosia
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington.
| | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neuroscienze e Organi di Senso (SMBNOS), Universita' degli Studi Aldo Moro, Policlinico, Bari, Italy
| |
Collapse
|
45
|
Kaur S, Hussain S, Kolhe K, Kumar G, Tripathi DM, Tomar A, Kale P, Narayanan A, Bihari C, Bajpai M, Maiwall R, Gupta E, Sarin SK. Elevated plasma ICAM1 levels predict 28-day mortality in cirrhotic patients with COVID-19 or bacterial sepsis. JHEP Rep 2021; 3:100303. [PMID: 33997748 PMCID: PMC8106200 DOI: 10.1016/j.jhepr.2021.100303] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/01/2021] [Accepted: 04/24/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND & AIMS Endothelial injury and dysfunction play a detrimental role in the pathogenesis of infections. Endothelium-related molecules have been reported as potential diagnostic and/or prognostic biomarkers of infection. The prognostic value of these biomarkers in patients with cirrhosis and infections remains elusive. METHODS In this study, we investigated the performance of key soluble endothelial injury biomarkers, including intercellular adhesion molecule 1 (ICAM1), von Willebrand factor (vWF), vascular endothelial growth factor receptor 1 (VEGFR1), and angiopoietin 1 and 2 (Ang1, 2) as mortality predictors in patients with cirrhosis and severe COVID-19 or bacterial sepsis. RESULTS A total of 66 hospitalized patients (admitted to the COVID-19 ward or liver intensive care unit [ICU]) were included. Twenty-two patients had COVID-19 alone, while 20 patients had cirrhosis plus COVID-19. Twenty-four patients had cirrhosis plus bacterial sepsis. Among patients with cirrhosis, the most common aetiology of liver disease was alcohol. ICAM1 was increased (p = 0.003) while VEGFR1 (p <0.0001) and Ang1 (p <0.0001) were reduced in patients with COVID-19 and cirrhosis, compared to patients with COVID-19 alone. Endothelial biomarker levels did not differ significantly between patients with cirrhosis and severe COVID-19 or bacterial sepsis in the ICU. In these patients, ICAM1 levels significantly and independently predicted mortality (hazard ratio 3.24; 95% CI 1.19-8.86) along with model for end-stage liver disease (MELD) score, renal and coagulation failures. The AUC for ICAM1 was 0.74, MELD was 0.60 and combined ICAM1 and MELD was 0.70. ICAM1 also positively correlated with the composite organ failure scores recorded 3-5 days post ICU admission (CLIF-OF and SOFA) in this subgroup of patients. CONCLUSION The study indicates that in patients with cirrhosis, elevated plasma ICAM1 serves as an independent predictor of severe COVID-19- or sepsis-associated 28-day mortality. LAY SUMMARY Bacterial sepsis and COVID-19 lead to increased mortality in patients with cirrhosis. In this study, we demonstrate that high plasma levels of ICAM1, an endothelial injury biomarker, is one of the important factors predicting mortality in critically ill cirrhotic patients with severe COVID-19 or bacterial sepsis.
Collapse
Key Words
- ACLF, acute-on-chronic liver failure
- AST, aspartate aminotransferase
- Ang1, angiopoietin 1
- Ang2, angiopoietin 2
- Biomarkers
- CCI, Charlson comorbidity index
- COVID-19
- Endothelial Injury
- HR, hazard ratio
- ICAM1, intercellular adhesion molecule 1
- ICU, intensive care unit
- LDH, lactate dehydrogenase
- Liver Cirrhosis
- MELD, model for end-stage liver disease
- NLR, neutrophil to lymphocyte ratio
- PCT, procalcitonin
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SOFA, sequential organ failure assessment
- Sepsis
- VEGFR1, vascular endothelial growth factor receptor 1
- vWF, von Willebrand factor
Collapse
Affiliation(s)
- Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of liver and biliary Sciences, New Delhi, India
| | - Sadam Hussain
- Department of Molecular and Cellular Medicine, Institute of liver and biliary Sciences, New Delhi, India
| | - Kailash Kolhe
- Department of Molecular and Cellular Medicine, Institute of liver and biliary Sciences, New Delhi, India
- Department of Gastroenterology, Lokmanya Tilak Municipal Medical College and General Hospital, Sion, Mumbai, India
| | - Guresh Kumar
- Department of Molecular and Cellular Medicine, Institute of liver and biliary Sciences, New Delhi, India
- Department of Research and Biostatistics, Institute of liver and biliary Sciences, New Delhi, India
| | - Dinesh M. Tripathi
- Department of Molecular and Cellular Medicine, Institute of liver and biliary Sciences, New Delhi, India
| | - Arvind Tomar
- Department of Pulmonary Medicine, Institute of liver and biliary Sciences, New Delhi, India
| | - Pratibha Kale
- Department of Microbiology, Institute of liver and biliary Sciences, New Delhi, India
| | - Ashad Narayanan
- Department of Emergency, Institute of liver and biliary Sciences, New Delhi, India
| | - Chaggan Bihari
- Department of Pathology, Institute of liver and biliary Sciences, New Delhi, India
| | - Meenu Bajpai
- Department of Transfusion Medicine, Institute of liver and biliary Sciences, New Delhi, India
| | - Rakhi Maiwall
- Department of Hepatology, Institute of liver and biliary Sciences, New Delhi, India
| | - Ekta Gupta
- Department of Virology, Institute of liver and biliary Sciences, New Delhi, India
| | - Shiv K. Sarin
- Department of Hepatology, Institute of liver and biliary Sciences, New Delhi, India
| |
Collapse
|
46
|
Brogna B, Romano A, Tibullo L, Montuori M, Nunziata M, Russo G, Musto LA. Rare findings of spontaneous hemothorax and small subpleural lung hematoma in a COVID-19 patient: A case report. Acta Radiol Open 2021; 10:20584601211028149. [PMID: 34367668 PMCID: PMC8326360 DOI: 10.1177/20584601211028149] [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] [Received: 02/08/2021] [Accepted: 06/07/2021] [Indexed: 01/27/2023] Open
Abstract
Hemothorax (HT) and pulmonary hematoma represent rare complications of anticoagulant therapy. We present a rare case of a 53-year-old man with COVID-19 pneumonia who showed, in a follow-up computed tomography (CT) scan 13 days after hospitalization, a left HT and a small hyperdense area in a subpleural location and compatible with a small subpleural hematoma. This patient was being treated with a subcutaneous administration of low-molecular-weight heparin (100UI/kg/BID). No vascular malformations were visualized on the CT pulmonary angiography. Herein, we report the first case of both a spontaneous HT and a lung subpleural hematoma in a COVID-19 patient, probably caused by anticoagulant therapy.
Collapse
Affiliation(s)
- Barbara Brogna
- Department of Radiology, San Giuseppe Moscati Hospital Avellino, Avellino, Italy
- Barbara Brogna, Department of Radiology San Giuseppe Moscati Hospital Avellino, National Speciality Hospital San Giuseppe Moscati Neurology and Stroke Unit, Contrada Amoretta, Avellino 83100, Italy.
| | - Annamaria Romano
- Department of Pneumology, San Giuseppe Moscati Hospital Avellino, Avellino, Italy
| | - Loredana Tibullo
- Department of General Medicine, San Giuseppe Moscati Hospital Avellino, Avellino, Italy
| | - Mariagrazia Montuori
- Department of Pneumology, San Giuseppe Moscati Hospital Avellino, Avellino, Italy
| | - Mariagrazia Nunziata
- Department of General Medicine, San Giuseppe Moscati Hospital Avellino, Avellino, Italy
| | - Giuseppe Russo
- Department of Pneumology, San Giuseppe Moscati Hospital Avellino, Avellino, Italy
| | - Lanfranco A Musto
- Department of Radiology, San Giuseppe Moscati Hospital Avellino, Avellino, Italy
| |
Collapse
|
47
|
Emerging Approaches to Understanding Microvascular Endothelial Heterogeneity: A Roadmap for Developing Anti-Inflammatory Therapeutics. Int J Mol Sci 2021; 22:ijms22157770. [PMID: 34360536 PMCID: PMC8346165 DOI: 10.3390/ijms22157770] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022] Open
Abstract
The endothelium is the inner layer of all blood vessels and it regulates hemostasis. It also plays an active role in the regulation of the systemic inflammatory response. Systemic inflammatory disease often results in alterations in vascular endothelium barrier function, increased permeability, excessive leukocyte trafficking, and reactive oxygen species production, leading to organ damage. Therapeutics targeting endothelium inflammation are urgently needed, but strong concerns regarding the level of phenotypic heterogeneity of microvascular endothelial cells between different organs and species have been expressed. Microvascular endothelial cell heterogeneity in different organs and organ-specific variations in endothelial cell structure and function are regulated by intrinsic signals that are differentially expressed across organs and species; a result of this is that neutrophil recruitment to discrete organs may be regulated differently. In this review, we will discuss the morphological and functional variations in differently originated microvascular endothelia and discuss how these variances affect systemic function in response to inflammation. We will review emerging in vivo and in vitro models and techniques, including microphysiological devices, proteomics, and RNA sequencing used to study the cellular and molecular heterogeneity of endothelia from different organs. A better understanding of microvascular endothelial cell heterogeneity will provide a roadmap for developing novel therapeutics to target the endothelium.
Collapse
|
48
|
Villalba N, Baby S, Yuan SY. The Endothelial Glycocalyx as a Double-Edged Sword in Microvascular Homeostasis and Pathogenesis. Front Cell Dev Biol 2021; 9:711003. [PMID: 34336864 PMCID: PMC8316827 DOI: 10.3389/fcell.2021.711003] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/22/2021] [Indexed: 12/27/2022] Open
Abstract
Expressed on the endothelial cell (EC) surface of blood vessels, the glycocalyx (GCX), a mixture of carbohydrates attached to proteins, regulates the access of cells and molecules in the blood to the endothelium. Besides protecting endothelial barrier integrity, the dynamic microstructure of the GCX confers remarkable functions including mechanotransduction and control of vascular tone. Recently, a novel perspective has emerged supporting the pleiotropic roles of the endothelial GCX (eGCX) in cardiovascular health and disease. Because eGCX degradation occurs in certain pathological states, the circulating levels of eGCX degradation products have been recognized to have diagnostic or prognostic values. Beyond their biomarker roles, certain eGCX fragments serve as pathogenic factors in disease progression. Pharmacological interventions that attenuate eGCX degradation or restore its integrity have been sought. This review provides our current understanding of eGCX structure and function across the microvasculature in different organs. We also discuss disease or injury states, such as infection, sepsis and trauma, where eGCX dysfunction contributes to severe inflammatory vasculopathy.
Collapse
Affiliation(s)
- Nuria Villalba
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sheon Baby
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
49
|
Zanza C, Tassi MF, Romenskaya T, Piccolella F, Abenavoli L, Franceschi F, Piccioni A, Ojetti V, Saviano A, Canonico B, Montanari M, Zamai L, Artico M, Robba C, Racca F, Longhitano Y. Lock, Stock and Barrel: Role of Renin-Angiotensin-Aldosterone System in Coronavirus Disease 2019. Cells 2021; 10:1752. [PMID: 34359922 PMCID: PMC8306543 DOI: 10.3390/cells10071752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/21/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Since the end of 2019, the medical-scientific community has been facing a terrible pandemic caused by a new airborne viral agent known as SARS-CoV2. Already in the early stages of the pandemic, following the discovery that the virus uses the ACE2 cell receptor as a molecular target to infect the cells of our body, it was hypothesized that the renin-angiotensin-aldosterone system was involved in the pathogenesis of the disease. Since then, numerous studies have been published on the subject, but the exact role of the renin-angiotensin-aldosterone system in the pathogenesis of COVID-19 is still a matter of debate. RAAS represents an important protagonist in the pathogenesis of COVID-19, providing the virus with the receptor of entry into host cells and determining its organotropism. Furthermore, following infection, the virus is able to cause an increase in plasma ACE2 activity, compromising the normal function of the RAAS. This dysfunction could contribute to the establishment of the thrombo-inflammatory state characteristic of severe forms of COVID-19. Drugs targeting RAAS represent promising therapeutic options for COVID-19 sufferers.
Collapse
Affiliation(s)
- Christian Zanza
- Department of Emergency Medicine, Foundation of Policlinico Agostino Gemelli-IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (F.F.); (A.P.); (V.O.); (A.S.)
- Department of Anesthesia and Critical Care, AON SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy; (T.R.); (F.P.); (F.R.); (Y.L.)
- Foundation Ospedale Alba-Bra and Department of Anesthesia, Critical Care and Emergency Medicine, Pietro and Michele Ferrero Hospital, 12051 Verduno, Italy
| | - Michele Fidel Tassi
- Department of Emergency Medicine, AON SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy;
| | - Tatsiana Romenskaya
- Department of Anesthesia and Critical Care, AON SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy; (T.R.); (F.P.); (F.R.); (Y.L.)
| | - Fabio Piccolella
- Department of Anesthesia and Critical Care, AON SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy; (T.R.); (F.P.); (F.R.); (Y.L.)
| | - Ludovico Abenavoli
- Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy;
| | - Francesco Franceschi
- Department of Emergency Medicine, Foundation of Policlinico Agostino Gemelli-IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (F.F.); (A.P.); (V.O.); (A.S.)
| | - Andrea Piccioni
- Department of Emergency Medicine, Foundation of Policlinico Agostino Gemelli-IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (F.F.); (A.P.); (V.O.); (A.S.)
| | - Veronica Ojetti
- Department of Emergency Medicine, Foundation of Policlinico Agostino Gemelli-IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (F.F.); (A.P.); (V.O.); (A.S.)
| | - Angela Saviano
- Department of Emergency Medicine, Foundation of Policlinico Agostino Gemelli-IRCCS, Catholic University of Sacred Heart, 00168 Rome, Italy; (F.F.); (A.P.); (V.O.); (A.S.)
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (B.C.); (M.M.); (L.Z.)
| | - Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (B.C.); (M.M.); (L.Z.)
| | - Loris Zamai
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (B.C.); (M.M.); (L.Z.)
- National Institute for Nuclear Physics (INFN)-Gran Sasso National Laboratory (LNGS), 67100 Assergi L’Aquila, Italy
| | - Marco Artico
- Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy;
| | - Chiara Robba
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, Italy;
| | - Fabrizio Racca
- Department of Anesthesia and Critical Care, AON SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy; (T.R.); (F.P.); (F.R.); (Y.L.)
| | - Yaroslava Longhitano
- Department of Anesthesia and Critical Care, AON SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy; (T.R.); (F.P.); (F.R.); (Y.L.)
- Foundation Ospedale Alba-Bra and Department of Anesthesia, Critical Care and Emergency Medicine, Pietro and Michele Ferrero Hospital, 12051 Verduno, Italy
| |
Collapse
|
50
|
Qin Z, Liu F, Blair R, Wang C, Yang H, Mudd J, Currey JM, Iwanaga N, He J, Mi R, Han K, Midkiff CC, Alam MA, Aktas BH, Heide RSV, Veazey R, Piedimonte G, Maness NJ, Ergün S, Mauvais-Jarvis F, Rappaport J, Kolls JK, Qin X. Endothelial cell infection and dysfunction, immune activation in severe COVID-19. Theranostics 2021; 11:8076-8091. [PMID: 34335981 PMCID: PMC8315069 DOI: 10.7150/thno.61810] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023] Open
Abstract
Rationale: Pulmonary vascular endotheliitis, perivascular inflammation, and immune activation are observed in COVID-19 patients. While the initial SARS-CoV-2 infection mainly infects lung epithelial cells, whether it also infects endothelial cells (ECs) and to what extent SARS-CoV-2-mediated pulmonary vascular endotheliitis is associated with immune activation remain to be determined. Methods: To address these questions, we studied SARS-CoV-2-infected K18-hACE2 (K18) mice, a severe COVID-19 mouse model, as well as lung samples from SARS-CoV-2-infected nonhuman primates (NHP) and patient deceased from COVID-19. We used immunostaining, RNAscope, and electron microscopy to analyze the organs collected from animals and patient. We conducted bulk and single cell (sc) RNA-seq analyses, and cytokine profiling of lungs or serum of the severe COVID-19 mice. Results: We show that SARS-CoV-2-infected K18 mice develop severe COVID-19, including progressive body weight loss and fatality at 7 days, severe lung interstitial inflammation, edema, hemorrhage, perivascular inflammation, systemic lymphocytopenia, and eosinopenia. Body weight loss in K18 mice correlated with the severity of pneumonia, but not with brain infection. We also observed endothelial activation and dysfunction in pulmonary vessels evidenced by the up-regulation of VCAM1 and ICAM1 and the downregulation of VE-cadherin. We detected SARS-CoV-2 in capillary ECs, activation and adhesion of platelets and immune cells to the vascular wall of the alveolar septa, and increased complement deposition in the lungs, in both COVID-19-murine and NHP models. We also revealed that pathways of coagulation, complement, K-ras signaling, and genes of ICAM1 and VCAM1 related to EC dysfunction and injury were upregulated, and were associated with massive immune activation in the lung and circulation. Conclusion: Together, our results indicate that SARS-CoV-2 causes endotheliitis via both infection and infection-mediated immune activation, which may contribute to the pathogenesis of severe COVID-19 disease.
Collapse
Affiliation(s)
- Zhongnan Qin
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Fengming Liu
- Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Robert Blair
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Chenxiao Wang
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Haoran Yang
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Joseph Mudd
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Joshua M Currey
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Naoki Iwanaga
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jibao He
- Coordinated Instrumentation Facility, Tulane University, New Orleans LA 70118, USA
| | - Ren Mi
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Kun Han
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | | | | | - Bertal H Aktas
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Ronald Veazey
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Giovanni Piedimonte
- Departments of Pediatrics, Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-Universität Würzburg, Koellikerstrasse 6, 97070 Würzburg, Germany
| | - Franck Mauvais-Jarvis
- Department of Medicine, Section of Endocrinology and Metabolism, Tulane University Health Sciences Center, School of Medicine, New Orleans, LA 70112, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
- Tulane Center of Excellence in Sex-Based Biology & Medicine, LA 70112, USA
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jay K. Kolls
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Xuebin Qin
- Tulane National Primate Research Center, Covington, LA 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|