151
|
Teich N, Stallmach A. COVID-19 und chronisch-entzündliche Darmerkrankungen. DIE GASTROENTEROLOGIE 2023. [PMCID: PMC9969944 DOI: 10.1007/s11377-023-00679-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Die Pandemie durch die Coronavirus Disease 2019 (COVID-19) beeinflusst weiter das Leben von Patient*innen mit chronisch-entzündlichen Darmerkrankungen (CED). Umfangreiche Untersuchungen der letzten 3 Jahre ergaben, dass die allermeisten Infektionen durch „severe acute respiratory syndrome coronavirus type 2“ (SARS-CoV-2) bei CED-Patient*innen blande verlaufen. In der Regel wird die Krankheitsaktivität der CED nicht negativ beeinflusst; bei einem Teil der Patient*innen können passager gastrointestinale Symptome auftreten. Häufig eingesetzte immunmodulierende Medikamente hatten mit Ausnahme systemischer Glukokortikoide keinen Einfluss auf den Schweregrad einer COVID-19-Erkrankung und die Gesamtletalität unterschied sich nicht von der übrigen Bevölkerung. Die Impfantwort ist jedoch substanzabhängig erniedrigt. In dieser Übersichtsarbeit werden die wichtigsten Studien praxisrelevant zusammengefasst.
Collapse
Affiliation(s)
- Niels Teich
- Internistische Gemeinschaftspraxis für Verdauungs- und Stoffwechselkrankheiten Leipzig und Schkeuditz, Nordstr. 21, 04105 Leipzig, Deutschland
| | - Andreas Stallmach
- Klinik für Innere Medizin IV, Universitätsklinikum Jena, Am Klinikum 1, 07747 Jena, Deutschland
| |
Collapse
|
152
|
Staroverov V, Nersisyan S, Galatenko A, Alekseev D, Lukashevich S, Polyakov F, Anisimov N, Tonevitsky A. Development of a novel mathematical model that explains SARS-CoV-2 infection dynamics in Caco-2 cells. PeerJ 2023; 11:e14828. [PMID: 36748087 PMCID: PMC9899056 DOI: 10.7717/peerj.14828] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/09/2023] [Indexed: 02/04/2023] Open
Abstract
Mathematical modeling is widely used to study within-host viral dynamics. However, to the best of our knowledge, for the case of SARS-CoV-2 such analyses were mainly conducted with the use of viral load data and for the wild type (WT) variant of the virus. In addition, only few studies analyzed models for in vitro data, which are less noisy and more reproducible. In this work we collected multiple data types for SARS-CoV-2-infected Caco-2 cell lines, including infectious virus titers, measurements of intracellular viral RNA, cell viability data and percentage of infected cells for the WT and Delta variants. We showed that standard models cannot explain some key observations given the absence of cytopathic effect in human cell lines. We propose a novel mathematical model for in vitro SARS-CoV-2 dynamics, which included explicit modeling of intracellular events such as exhaustion of cellular resources required for virus production. The model also explicitly considers innate immune response. The proposed model accurately explained experimental data. Attenuated replication of the Delta variant in Caco-2 cells could be explained by our model on the basis of just two parameters: decreased cell entry rate and increased cytokine production rate.
Collapse
Affiliation(s)
- Vladimir Staroverov
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia,Faculty of Mechanics and Mathematics, Lomonosov Moscow State University, Moscow, Russia
| | - Stepan Nersisyan
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia,Institute of Molecular Biology, The National Academy of Sciences of the Republic of Armenia, Yerevan, Armenia,Armenian Bioinformatics Institute (ABI), Yerevan, Armenia,Current Affiliation: Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexei Galatenko
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia,Faculty of Mechanics and Mathematics, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitriy Alekseev
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia,Faculty of Mechanics and Mathematics, Lomonosov Moscow State University, Moscow, Russia
| | - Sofya Lukashevich
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia
| | - Fedor Polyakov
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Nikita Anisimov
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia,Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
153
|
Peluso MJ, Deveau TM, Munter SE, Ryder D, Buck A, Beck-Engeser G, Chan F, Lu S, Goldberg SA, Hoh R, Tai V, Torres L, Iyer NS, Deswal M, Ngo LH, Buitrago M, Rodriguez A, Chen JY, Yee BC, Chenna A, Winslow JW, Petropoulos CJ, Deitchman AN, Hellmuth J, Spinelli MA, Durstenfeld MS, Hsue PY, Kelly JD, Martin JN, Deeks SG, Hunt PW, Henrich TJ. Chronic viral coinfections differentially affect the likelihood of developing long COVID. J Clin Invest 2023; 133:e163669. [PMID: 36454631 PMCID: PMC9888380 DOI: 10.1172/jci163669] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/30/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUNDThe presence and reactivation of chronic viral infections, such as EBV, CMV, and HIV, have been proposed as potential contributors to long COVID (LC), but studies in well-characterized postacute cohorts of individuals with COVID-19 over a longer time course consistent with current case definitions of LC are limited.METHODSIn a cohort of 280 adults with prior SARS-CoV-2 infection, we assessed the presence and types of LC symptoms and prior medical history (including COVID-19 history and HIV status) and performed serological testing for EBV and CMV using a commercial laboratory. We used covariate-adjusted binary logistic regression models to identify independent associations between variables and LC symptoms.RESULTSWe observed that LC symptoms, such as fatigue and neurocognitive dysfunction, at a median of 4 months following initial diagnosis were independently associated with serological evidence suggesting recent EBV reactivation (early antigen-diffuse IgG positivity) or high nuclear antigen (EBNA) IgG levels but not with ongoing EBV viremia. Serological evidence suggesting recent EBV reactivation (early antigen-diffuse IgG positivity) was most strongly associated with fatigue (OR = 2.12). Underlying HIV infection was also independently associated with neurocognitive LC (OR = 2.5). Interestingly, participants who had serologic evidence of prior CMV infection were less likely to develop neurocognitive LC (OR = 0.52).CONCLUSIONOverall, these findings suggest differential effects of chronic viral coinfections on the likelihood of developing LC and association with distinct syndromic patterns. Further assessment during the acute phase of COVID-19 is warranted.TRIAL REGISTRATIONLong-term Impact of Infection with Novel Coronavirus; ClinicalTrials.gov NCT04362150.FUNDINGThis work was supported by NIH/National Institute of Allergy and Infectious Diseases grants (3R01AI141003-03S1, R01AI158013, and K24AI145806); the Zuckerberg San Francisco General Hospital Department of Medicine and Division of HIV, Infectious Diseases, and Global Medicine; and the UCSF-Bay Area Center for AIDS Research (P30-AI027763).
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fay Chan
- Division of Experimental Medicine, and
| | - Scott Lu
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, California, USA
| | - Sarah A. Goldberg
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, California, USA
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases, and Global Medicine
| | - Viva Tai
- Division of HIV, Infectious Diseases, and Global Medicine
| | | | | | - Monika Deswal
- Division of HIV, Infectious Diseases, and Global Medicine
| | - Lynn H. Ngo
- Division of HIV, Infectious Diseases, and Global Medicine
| | | | | | | | - Brandon C. Yee
- Monogram Biosciences Inc., South San Francisco, California, USA
| | - Ahmed Chenna
- Monogram Biosciences Inc., South San Francisco, California, USA
| | - John W. Winslow
- Monogram Biosciences Inc., South San Francisco, California, USA
| | | | | | | | | | | | | | - J. Daniel Kelly
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, California, USA
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, California, USA
| | | | | | | |
Collapse
|
154
|
Erdik B. Covid associated immune dysfunction (CAID) leading to increased pediatric hospitalizations. J Med Virol 2023; 95:e28508. [PMID: 36655737 DOI: 10.1002/jmv.28508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/19/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
|
155
|
Vojdani A, Vojdani E, Saidara E, Maes M. Persistent SARS-CoV-2 Infection, EBV, HHV-6 and Other Factors May Contribute to Inflammation and Autoimmunity in Long COVID. Viruses 2023; 15:v15020400. [PMID: 36851614 PMCID: PMC9967513 DOI: 10.3390/v15020400] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
A novel syndrome called long-haul COVID or long COVID is increasingly recognized in a significant percentage of individuals within a few months after infection with SARS-CoV-2. This disorder is characterized by a wide range of persisting, returning or even new but related symptoms that involve different tissues and organs, including respiratory, cardiac, vascular, gastrointestinal, musculo-skeletal, neurological, endocrine and systemic. Some overlapping symptomatologies exist between long COVID and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Very much like with long ME/CFS, infections with herpes family viruses, immune dysregulation, and the persistence of inflammation have been reported as the most common pattern for the development of long COVID. This review describes several factors and determinants of long COVID that have been proposed, elaborating mainly on viral persistence, reactivation of latent viruses such as Epstein-Barr virus and human herpesvirus 6 which are also associated with the pathology of ME/CFS, viral superantigen activation of the immune system, disturbance in the gut microbiome, and multiple tissue damage and autoimmunity. Based on these factors, we propose diagnostic strategies such as the measurement of IgG and IgM antibodies against SARS-CoV-2, EBV, HHV-6, viral superantigens, gut microbiota, and biomarkers of autoimmunity to better understand and manage this multi-factorial disorder that continues to affect millions of people in the world.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab, Inc., Los Angeles, CA 90035, USA
- Cyrex Laboratories, LLC, Phoenix, AZ 85034, USA
- Correspondence: ; Tel.: +1-310-657-1077
| | | | - Evan Saidara
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Road, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
156
|
Gupta T, Sharma H. COVID-19 and the liver: Are footprints still there? World J Gastroenterol 2023; 29:656-669. [PMID: 36742164 PMCID: PMC9896610 DOI: 10.3748/wjg.v29.i4.656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/13/2022] [Accepted: 11/21/2022] [Indexed: 01/20/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) hit the entire world as a global pandemic and soon became the most important concern for all patients with chronic diseases. An early trend in higher mortality in patients with acute respiratory distress attracted all researchers to closely monitor patients for the involvement of other systems. It soon became apparent that patients with chronic liver diseases are at increased risk of mortality given their cirrhosis-associated immune dysfunction. Additionally, liver function abnormalities were noted in patients with severe COVID-19. Profound cytokine storm, direct viral infection, drugs and reactivation of viral infections were causes of deranged liver functions. Here, we discuss the relation between COVID-19 and chronic liver disease, specifically cirrhosis, hepatitis B, hepatitis C, and non-alcoholic fatty liver disease (NAFLD), as well as the liver manifestations of COVID-19. The metabolic syndrome, obesity, diabetes mellitus and NAFLD were found to worsen outcome in different studies reported worldwide. Decompensated cirrhosis should be considered a risk factor for death and severe COVID-19. Recently, COVID-19 related cholangiopathy has also been reported with changes of secondary sclerosing cholangitis. The long-term persistence of viral antigens in gut epithelia raises concern regarding the future risk of autoimmune liver diseases.
Collapse
Affiliation(s)
- Tarana Gupta
- Department of Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical Sciences, Rohtak 124001, Haryana, India
| | - Hemant Sharma
- Department of Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical Sciences, Rohtak 124001, Haryana, India
| |
Collapse
|
157
|
Allendes FJ, Díaz HS, Ortiz FC, Marcus NJ, Quintanilla R, Inestrosa NC, Del Rio R. Cardiovascular and autonomic dysfunction in long-COVID syndrome and the potential role of non-invasive therapeutic strategies on cardiovascular outcomes. Front Med (Lausanne) 2023; 9:1095249. [PMID: 36743679 PMCID: PMC9892856 DOI: 10.3389/fmed.2022.1095249] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/21/2022] [Indexed: 01/21/2023] Open
Abstract
A significant percentage of COVID-19 survivors develop long-lasting cardiovascular sequelae linked to autonomic nervous system dysfunction, including fatigue, arrhythmias, and hypertension. This post-COVID-19 cardiovascular syndrome is one facet of "long-COVID," generally defined as long-term health problems persisting/appearing after the typical recovery period of COVID-19. Despite the fact that this syndrome is not fully understood, it is urgent to develop strategies for diagnosing/managing long-COVID due to the immense potential for future disease burden. New diagnostic/therapeutic tools should provide health personnel with the ability to manage the consequences of long-COVID and preserve/improve patient quality of life. It has been shown that cardiovascular rehabilitation programs (CRPs) stimulate the parasympathetic nervous system, improve cardiorespiratory fitness (CRF), and reduce cardiovascular risk factors, hospitalization rates, and cognitive impairment in patients suffering from cardiovascular diseases. Given their efficacy in improving patient outcomes, CRPs may have salutary potential for the treatment of cardiovascular sequelae of long-COVID. Indeed, there are several public and private initiatives testing the potential of CRPs in treating fatigue and dysautonomia in long-COVID subjects. The application of these established rehabilitation techniques to COVID-19 cardiovascular syndrome represents a promising approach to improving functional capacity and quality of life. In this brief review, we will focus on the long-lasting cardiovascular and autonomic sequelae occurring after COVID-19 infection, as well as exploring the potential of classic and novel CRPs for managing COVID-19 cardiovascular syndrome. Finally, we expect this review will encourage health care professionals and private/public health organizations to evaluate/implement non-invasive techniques for the management of COVID-19 cardiovascular sequalae.
Collapse
Affiliation(s)
- Francisca J. Allendes
- Laboratory Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hugo S. Díaz
- Laboratory Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando C. Ortiz
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de Salud, Universidad Autónoma de Chile, Santiago, Chile,Departamento de Biología, Mechanisms of Myelin Formation and Repair Laboratory, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Noah J. Marcus
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA, United States
| | - Rodrigo Quintanilla
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Nibaldo C. Inestrosa
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA, United States
| | - Rodrigo Del Rio
- Laboratory Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile,Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile,*Correspondence: Rodrigo Del Rio,
| |
Collapse
|
158
|
Kouri A, Gupta S. Respiratory manifestations of long COVID. CANADIAN JOURNAL OF RESPIRATORY, CRITICAL CARE, AND SLEEP MEDICINE 2023. [DOI: 10.1080/24745332.2022.2156407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Andrew Kouri
- Department of Medicine, Division of Respirology, St. Michael’s Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Samir Gupta
- Department of Medicine, Division of Respirology, St. Michael’s Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| |
Collapse
|
159
|
Li H, Xia J, Bennett D, Roque F, Bam RA, Tavares ABT, Gokhale M, Ida F, Rhee JJ, Soriano Gabarro M, Song Y. Long-COVID-19 clinical and health outcomes: an umbrella review. Ther Adv Infect Dis 2023; 10:20499361231198335. [PMID: 37720384 PMCID: PMC10501084 DOI: 10.1177/20499361231198335] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/03/2023] [Indexed: 09/19/2023] Open
Abstract
Background A growing interest in long-term sequelae of COVID-19 has prompted several systematic literature reviews (SLRs) to evaluate long-COVID-19 effects. However, many of these reviews lack in-depth information on the timing, duration, and severity of these conditions. Objectives Our aim was to synthesize both qualitative and quantitative evidence on prevalence and outcomes of long-term effect of COVID-19 through an umbrella review. Design Umbrella review of relevant SLRs on long-COVID-19 in terms of prolonged symptoms and clinical conditions, and comprehensively synthesized the latest existing evidence. Data Sources and Methods We systematically identified and appraised prior systematic reviews/meta-analyses using MEDLINE, Embase, and Cochrane database of systematic review from 2020 to 2021 following the preferred reporting items for systematic reviews and meta-analyses guidance. We summarized and categorized all relevant clinical symptoms and outcomes in adults with COVID-19 using the Medical Dictionary for Regulatory Activities System Organ Class (MedDRA SOC). Results We identified 967 systematic reviews/meta-analyses; 36 were retained for final data extraction. The most prevalent SOC were social circumstances (40%), blood and lymphatic system disorders (39%), and metabolism and nutrition disorder (38%). The most frequently reported SOC outcomes within each MedDRA category were poor quality of life (59%), wheezing and dyspnea (19-49%), fatigue (30-64%), chest pain (16%), decreased or loss of appetite (14-17%), abdominal discomfort or digestive disorder (12-18%), arthralgia with or without myalgia (16-24%), paresthesia (27%) and hair loss (14-25%), and hearing loss or tinnitus (15%). Conclusion This study confirmed a high prevalence of several long COVID-19 outcomes according to the MedDRA categories and indicated that the majority of evidence was rated as moderate to low. Registration The review was registered at PROSPERO (https://www.crd.york.ac.uk/prospero/) (CRD42022303557).
Collapse
Affiliation(s)
- Hu Li
- Gilead Sciences Inc., 333 Lakeside Drive, Foster City, CA 94404, USA
| | - Jin Xia
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Dimitri Bennett
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fatima Roque
- Research Unit for Inland Development, Polytechnic of Guarda (UDI-IPG), Guarda, Portugal
| | | | | | - Mugdha Gokhale
- Pfizer Inc., New York, NY, USA
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | - Yiqing Song
- Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
160
|
Hany M, Zidan A, Gaballa M, Ibrahim M, Agayby ASS, Abouelnasr AA, Sheta E, Torensma B. Lingering SARS-CoV-2 in Gastric and Gallbladder Tissues of Patients with Previous COVID-19 Infection Undergoing Bariatric Surgery. Obes Surg 2023; 33:139-148. [PMID: 36316598 PMCID: PMC9628579 DOI: 10.1007/s11695-022-06338-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Lingering severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in gut tissue might be a source of infection during bariatric surgery. This study aimed to confirm the presence of SARS-CoV-2 nucleocapsid in gastric and gallbladder tissues removed during bariatric surgery in individuals previously infected with coronavirus disease 2019 (COVID-19) who had negative polymerase chain reaction results prior to the surgery. METHODS Gastric and gallbladder specimens from 80 patients who underwent bariatric surgery between November 2021 and May 2022 and had a history of COVID-19 infection with gastrointestinal symptoms were examined for the presence of lingering SARS-CoV-2 nucleocapsid proteins using immunohistochemistry. RESULTS Gastric specimens from 26 (32.5%) patients and 4 (100%) cholecystectomy specimens showed positive cytoplasmic staining for the anti-SARS-CoV-2 nucleocapsid protein in surface mucosal epithelial cells. The mean age was 37.8 ± 10.3 years. The average body mass index was 44.2 ± 7.0 kg/m2; most of the patients were females (71.3%). The positive staining group was significantly younger than the negative staining group (p = 0.007). The full-dose vaccination rate was 58.8%, with a median of 91 days after the last vaccine dose. A positive serological anti-spike IgG response was observed in 99% of the patients. The median time between initial COVID-19 infection and surgery was 274 and 380 days in the positive and negative staining groups, respectively (p = 0.371). CONCLUSION Gastric and gallbladder tissues can retain SARS-CoV-2 particles for a long time after COVID-19 infection, handling stomach specimens from patients during an operation must be done with care, as we usually do, but now with the knowledge that in 1/3 of patients they can be present. Performing LSG on post-COVID patients did not seem to increase perioperative morbidity.
Collapse
Affiliation(s)
- Mohamed Hany
- grid.7155.60000 0001 2260 6941Department of Surgery, Medical Research Institute, Alexandria University, 165 Horreya Avenue, Hadara, 21561 Alexandria Egypt ,Madina Women’s Hospital, Alexandria, Egypt
| | - Ahmed Zidan
- grid.7155.60000 0001 2260 6941Department of Surgery, Medical Research Institute, Alexandria University, 165 Horreya Avenue, Hadara, 21561 Alexandria Egypt
| | - Muhammad Gaballa
- grid.7155.60000 0001 2260 6941Department of Surgery, Medical Research Institute, Alexandria University, 165 Horreya Avenue, Hadara, 21561 Alexandria Egypt
| | - Mohamed Ibrahim
- grid.7155.60000 0001 2260 6941Department of Surgery, Medical Research Institute, Alexandria University, 165 Horreya Avenue, Hadara, 21561 Alexandria Egypt
| | - Ann Samy Shafiq Agayby
- grid.7155.60000 0001 2260 6941Department of Surgery, Medical Research Institute, Alexandria University, 165 Horreya Avenue, Hadara, 21561 Alexandria Egypt
| | - Anwar Ashraf Abouelnasr
- grid.7155.60000 0001 2260 6941Department of Surgery, Medical Research Institute, Alexandria University, 165 Horreya Avenue, Hadara, 21561 Alexandria Egypt
| | - Eman Sheta
- grid.7155.60000 0001 2260 6941Pathology Department, Alexandria University, Alexandria, Egypt
| | - Bart Torensma
- grid.10419.3d0000000089452978Leiden University Medical Center (LUMC), Leiden, The Netherlands
| |
Collapse
|
161
|
Schultheiß C, Willscher E, Paschold L, Gottschick C, Klee B, Bosurgi L, Dutzmann J, Sedding D, Frese T, Girndt M, Höll JI, Gekle M, Mikolajczyk R, Binder M. Liquid biomarkers of macrophage dysregulation and circulating spike protein illustrate the biological heterogeneity in patients with post-acute sequelae of COVID-19. J Med Virol 2023; 95:e28364. [PMID: 36458566 PMCID: PMC9878213 DOI: 10.1002/jmv.28364] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/24/2022] [Accepted: 11/26/2022] [Indexed: 12/04/2022]
Abstract
Post-acute sequelae of COVID-19 (PASC) are long-term consequences of SARS-CoV-2 infection that can substantially impair the quality of life. Underlying mechanisms ranging from persistent viruses to innate and adaptive immune dysregulation have been discussed. Here, we profiled the plasma of 181 individuals from the cohort study for digital health research in Germany (DigiHero), including individuals after mild to moderate COVID-19 with or without PASC and uninfected controls. We focused on soluble factors related to monocyte/macrophage biology and on circulating SARS-CoV-2 spike (S1) protein as a potential biomarker for persistent viral reservoirs. At a median time of 8 months after infection, we found pronounced dysregulation in almost all tested soluble factors, including both pro-inflammatory and pro-fibrotic cytokines. These immunological perturbations were remarkably independent of ongoing PASC symptoms per se, but further correlation and regression analyses suggested PASC-specific patterns involving CCL2/MCP-1 and IL-8 that either correlated with sCD162, sCD206/MMR, IFN-α2, IL-17A and IL-33, or IL-18 and IL-23. None of the analyzed factors correlated with the detectability or levels of circulating S1, indicating that this represents an independent subset of patients with PASC. These data confirm prior evidence of immune dysregulation and persistence of viral protein in PASC and illustrate its biological heterogeneity that still awaits correlation with clinically defined PASC subtypes.
Collapse
Affiliation(s)
- Christoph Schultheiß
- Department of Internal Medicine IV, Oncology/HematologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Edith Willscher
- Department of Internal Medicine IV, Oncology/HematologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Lisa Paschold
- Department of Internal Medicine IV, Oncology/HematologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Cornelia Gottschick
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health SciencesMedical School of the Martin‐Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Bianca Klee
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health SciencesMedical School of the Martin‐Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Lidia Bosurgi
- I. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
| | - Jochen Dutzmann
- Department of Cardiology and Intensive Care Medicine, Mid‐German Heart Center, University HospitalMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Daniel Sedding
- Department of Cardiology and Intensive Care Medicine, Mid‐German Heart Center, University HospitalMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Thomas Frese
- Institute of General Practice and Family MedicineMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Matthias Girndt
- Department of Internal Medicine IIMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Jessica I. Höll
- Pediatric Hematology and OncologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Michael Gekle
- Julius Bernstein‐Institute of Physiology, Faculty of MedicineMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| | - Rafael Mikolajczyk
- Institute for Medical Epidemiology, Biometrics and Informatics (IMEBI), Interdisciplinary Center for Health SciencesMedical School of the Martin‐Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/HematologyMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| |
Collapse
|
162
|
Sehrawat S, Osterrieder N, Schmid DS, Rouse BT. Can the triumph of mRNA vaccines against COVID-19 be extended to other viral infections of humans and domesticated animals? Microbes Infect 2023; 25:105078. [PMID: 36435367 PMCID: PMC9682868 DOI: 10.1016/j.micinf.2022.105078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022]
Abstract
The unprecedented success of mRNA vaccines in managing the COVID-19 pandemic raises the prospect of applying the mRNA platform to other viral diseases of humans and domesticated animals, which may lead to more efficacious vaccines for some agents. We briefly discuss reasons why mRNA vaccines achieved such success against COVID-19 and indicate what other virus infections and disease conditions might also be ripe for control using mRNA vaccines. We also evaluate situations where mRNA could prove valuable to rebalance the status of immune responsiveness and achieve success as a therapeutic vaccine approach against infections that induce immunoinflammatory lesions.
Collapse
Affiliation(s)
- Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City, PO Manauli, Mohali 140306, Punjab, India.
| | - Nikolaus Osterrieder
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163 Berlin, Germany; Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 5F, Block 1B, To Yuen Building, 31 To Yuen Street, Kowloon Tong, Hong Kong.
| | - D Scott Schmid
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA.
| | - Barry T Rouse
- College of Veterinary Medicine, University of Tennessee Knoxville, TN 37996-0845, USA.
| |
Collapse
|
163
|
Zhou B, Pang X, Wu J, Liu T, Wang B, Cao H. Gut microbiota in COVID-19: new insights from inside. Gut Microbes 2023; 15:2201157. [PMID: 37078497 PMCID: PMC10120564 DOI: 10.1080/19490976.2023.2201157] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/04/2023] [Indexed: 04/21/2023] Open
Abstract
The epidemic of coronavirus disease-19 (COVID-19) has grown to be a global health threat. Gastrointestinal symptoms are thought to be common clinical manifestations apart from a series of originally found respiratory symptoms. The human gut harbors trillions of microorganisms that are indispensable for complex physiological processes and homeostasis. Growing evidence demonstrate that gut microbiota alteration is associated with COVID-19 progress and severity, and post-COVID-19 syndrome, characterized by decrease of anti-inflammatory bacteria like Bifidobacterium and Faecalibacterium and enrichment of inflammation-associated microbiota including Streptococcus and Actinomyces. Therapeutic strategies such as diet, probiotics/prebiotics, herb, and fecal microbiota transplantation have shown positive effects on relieving clinical symptoms. In this article, we provide and summarize the recent evidence about the gut microbiota and their metabolites alterations during and after COVID-19 infection and focus on potential therapeutic strategies targeting gut microbiota. Understanding the connections between intestinal microbiota and COVID-19 would provide new insights into COVID-19 management in the future.
Collapse
Affiliation(s)
- Bingqian Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xiaoqi Pang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jingyi Wu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
164
|
Li L, Cui J, Tang J, Shi J, Deng X, Zheng X, Fan Q, Liu Y, Yu H, Tang X, Hu F, Li F. High titers of neutralizing antibodies in the blood fail to eliminate SARS-CoV-2 viral RNA in the upper respiratory tract. J Med Virol 2023; 95:e28219. [PMID: 36229892 PMCID: PMC9874792 DOI: 10.1002/jmv.28219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/28/2022] [Accepted: 10/11/2022] [Indexed: 01/27/2023]
Abstract
Retest-positive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral RNA, as a unique phenomenon among discharged individuals, has been demonstrated to be safe in the community. Still, the underlying mechanism of viral lingering is less investigated. In this study, first, we find that the frequency of viral RNA-positive retesting differs among variants. Higher ratios of viral RNA-positive retest were more frequently observed among Delta (61.41%, 514 of 837 cases) and Omicron (39.53%, 119 of 301 cases) infections than among ancestral viral infection (7.27%, 21 of 289 cases). Second, the tissues where viral RNA reoccurred were altered. Delta RNA reoccurred mainly in the upper respiratory tract (90%), but ancestral virus RNA reoccurred mainly in the gastrointestinal tract (71%). Third, vaccination did not reduce the frequency of viral RNA-positive retests, despite high concentrations of viral-specific antibodies in the blood. Finally, 37 of 55 (67.27%) Delta-infected patients receiving neutralizing antibody therapy become viral RNA retest positive when high concentrations of neutralizing antibodies still patrol in the blood. Altogether, our findings suggest that the presentence of high titers of neutralizing antibodies in the blood is incompetent in clearing residual viral RNA in the upper respiratory tract.
Collapse
Affiliation(s)
- Lu Li
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Jianping Cui
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Jingyan Tang
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Jingrong Shi
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Xilong Deng
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Xiaowen Zheng
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Qinghong Fan
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Ying Liu
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Haisheng Yu
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Xiaoping Tang
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
- Guangzhou LaboratoryBio‐IslandGuangzhouChina
| | - Fengyu Hu
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
- Guangzhou LaboratoryBio‐IslandGuangzhouChina
| | - Feng Li
- Guangzhou Eighth People's HospitalGuangzhou Medical UniversityGuangzhouChina
- Guangzhou LaboratoryBio‐IslandGuangzhouChina
| |
Collapse
|
165
|
Muacevic A, Adler JR, Tanaka T, Kimura S, Shinchi Y, Yamano T. A Potential Novel Treatment for Chronic Cough in Long COVID Patients: Clearance of Epipharyngeal Residual SARS-CoV-2 Spike RNA by Epipharyngeal Abrasive Therapy. Cureus 2023; 15:e33421. [PMID: 36618501 PMCID: PMC9815934 DOI: 10.7759/cureus.33421] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2023] [Indexed: 01/07/2023] Open
Abstract
A major target of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the epipharyngeal mucosa. Epipharyngeal abrasive therapy (EAT) is a Japanese treatment for chronic epipharyngitis. EAT is a treatment for chronic epipharyngitis in Japan that involves applying zinc chloride as an anti-inflammatory agent to the epipharyngeal mucosa. Here, we present a case of a 21-year-old man with chronic coughing that persisted for four months after a diagnosis of mild coronavirus disease 2019 (COVID-19), who was treated by EAT. We diagnosed chronic epipharyngitis as the cause of the chronic cough after the SARS-CoV-2 infection. SARS-CoV-2 spike RNA had persisted in the epipharyngeal mucosa of this Long COVID patient. EAT was performed once a week for three months, which eliminated residual SARS-CoV-2 RNA and reduced epipharyngeal inflammation. Moreover, a reduction in the expression of proinflammatory cytokines was found by histopathological examination. We speculate that the virus was excreted with the drainage induced by EAT, which stopped the secretion of proinflammatory cytokines. This case study suggests that EAT is a useful treatment for chronic epipharyngitis involving long COVID.
Collapse
|
166
|
Mochochoko BM, Pohl CH, O’Neill HG. Candida albicans-enteric viral interactions-The prostaglandin E 2 connection and host immune responses. iScience 2022; 26:105870. [PMID: 36647379 PMCID: PMC9839968 DOI: 10.1016/j.isci.2022.105870] [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] [Indexed: 12/25/2022] Open
Abstract
The human microbiome comprises trillions of microorganisms residing within different mucosal cavities and across the body surface. The gut microbiota modulates host susceptibility to viral infections in several ways, and microbial interkingdom interactions increase viral infectivity within the gut. Candida albicans, a frequently encountered fungal species in the gut, produces highly structured biofilms and eicosanoids such as prostaglandin E2 (PGE2), which aid in viral protection and replication. These biofilms encompass viruses and provide a shield from antiviral drugs or the immune system. PGE2 is a key modulator of active inflammation with the potential to regulate interferon signaling upon microbial invasion or viral infections. In this review, we raise the perspective of gut interkingdom interactions involving C. albicans and enteric viruses, with a special focus on biofilms, PGE2, and viral replication. Ultimately, we discuss the possible implications of C. albicans-enteric virus associations on host immune responses, particularly the interferon signaling pathway.
Collapse
Affiliation(s)
- Bonang M. Mochochoko
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, 9301, South Africa
| | - Carolina H. Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, 9301, South Africa,Corresponding author
| | - Hester G. O’Neill
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, 9301, South Africa,Corresponding author
| |
Collapse
|
167
|
Haque A, Pant AB. Long Covid: Untangling the Complex Syndrome and the Search for Therapeutics. Viruses 2022; 15:42. [PMID: 36680082 PMCID: PMC9864843 DOI: 10.3390/v15010042] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Long Covid can affect anyone who has previously had acute COVID-19. The root causes of this syndrome are still unknown, and no effective therapeutics are available. This complex syndrome, with a wide array of symptoms, is still evolving. Given the dire situation, it is important to identify the causes of Long Covid and the changes occurring within the immune system of affected patients to figure out how to treat it. The immune system intersects with the persistent viral fragments and blood clots that are implicated in this syndrome; understanding how these complex systems interact may help in untangling the puzzling physiopathology of Long Covid and identifying mitigation measures to provide patients some relief. In this paper, we discuss evidence-based findings and formulate hypotheses on the mechanisms underlying Long Covid's physiopathology and propose potential therapeutic options.
Collapse
Affiliation(s)
- Azizul Haque
- One Medical Center Drive, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | | |
Collapse
|
168
|
He S, Wu K, Cheng Z, He M, Hu R, Fan N, Shen L, Li Q, Fan H, Tong Y. Long COVID: The latest manifestations, mechanisms, and potential therapeutic interventions. MedComm (Beijing) 2022; 3:e196. [PMID: 36514781 PMCID: PMC9732402 DOI: 10.1002/mco2.196] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022] Open
Abstract
COVID-19 caused by SARS-CoV-2 infection affects humans not only during the acute phase of the infection, but also several weeks to 2 years after the recovery. SARS-CoV-2 infects a variety of cells in the human body, including lung cells, intestinal cells, vascular endothelial cells, olfactory epithelial cells, etc. The damages caused by the infections of these cells and enduring immune response are the basis of long COVID. Notably, the changes in gene expression caused by viral infection can also indirectly contribute to long COVID. We summarized the occurrences of both common and uncommon long COVID, including damages to lung and respiratory system, olfactory and taste deficiency, damages to myocardial, renal, muscle, and enduring inflammation. Moreover, we provided potential treatments for long COVID symptoms manifested in different organs and systems, which were based on the pathogenesis and the associations between symptoms in different organs. Importantly, we compared the differences in symptoms and frequency of long COVID caused by breakthrough infection after vaccination and infection with different variants of concern, in order to provide a comprehensive understanding of the characteristics of long COVID and propose improvement for tackling COVID-19.
Collapse
Affiliation(s)
- Shi‐ting He
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Kexin Wu
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Zixuan Cheng
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Mengjie He
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Ruolan Hu
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Ning Fan
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Lin Shen
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Qirui Li
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Huahao Fan
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| | - Yigang Tong
- College of Life Science and TechnologyBeijing University of Chemical TechnologyBeijingChina
| |
Collapse
|
169
|
Abstract
The German Society of Pneumology initiated 2021 the AWMF S1 guideline Long COVID/Post-COVID. In a broad interdisciplinary approach, this S1 guideline was designed based on the current state of knowledge.The clinical recommendations describe current Long COVID/Post-COVID symptoms, diagnostic approaches, and therapies.In addition to the general and consensus introduction, a subject-specific approach was taken to summarize the current state of knowledge.The guideline has an explicit practical claim and will be developed and adapted by the author team based on the current increase in knowledge.
Collapse
|
170
|
Rendeiro AF, Ravichandran H, Kim J, Borczuk AC, Elemento O, Schwartz RE. Persistent alveolar type 2 dysfunction and lung structural derangement in post-acute COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.11.28.22282811. [PMID: 36482970 PMCID: PMC9727772 DOI: 10.1101/2022.11.28.22282811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
SARS-CoV-2 infection can manifest as a wide range of respiratory and systemic symptoms well after the acute phase of infection in over 50% of patients. Key questions remain on the long-term effects of infection on tissue pathology in recovered COVID-19 patients. To address these questions we performed multiplexed imaging of post-mortem lung tissue from 12 individuals who died post-acute COVID-19 (PC) and compare them to lung tissue from patients who died during the acute phase of COVID-19, or patients who died with idiopathic pulmonary fibrosis (IPF), and otherwise healthy lung tissue. We find evidence of viral presence in the lung up to 359 days after the acute phase of disease, including in patients with negative nasopharyngeal swab tests. The lung of PC patients are characterized by the accumulation of senescent alveolar type 2 cells, fibrosis with hypervascularization of peribronchial areas and alveolar septa, as the most pronounced pathophysiological features. At the cellular level, lung disease of PC patients, while distinct, shares pathological features with the chronic pulmonary disease of IPF. which may help rationalize interventions for PC patients. Altogether, this study provides an important foundation for the understanding of the long-term effects of SARS-CoV-2 pulmonary infection at the microanatomical, cellular, and molecular level.
Collapse
Affiliation(s)
- André F Rendeiro
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Current address: CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT 25.3, 1090, Vienna, Austria
| | - Hiranmayi Ravichandran
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Junbum Kim
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Alain C Borczuk
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Current address: Northwell Health, Department of Pathology and Laboratory Medicine, Greenvale, NY
| | - Olivier Elemento
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
171
|
COVID-19-Antigenpersistenz im Darmtrakt – Ursache für Long-COVID? ZEITSCHRIFT FÜR GASTROENTEROLOGIE 2022. [DOI: 10.1055/a-1923-8203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
172
|
Saif-Ur-Rahman K, Kothari K, Sadlier C, Moriarty F, Movsisyan A, Whelan S, Taneri PE, Blair M, Guyatt G, Devane D. Effect of COVID-19 vaccines for the treatment of people with post-COVID-19 condition: a rapid review. HRB Open Res 2022. [DOI: 10.12688/hrbopenres.13638.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background: Vaccination for coronavirus disease 2019 (COVID-19) has demonstrated reduced risk of hospitalisation and death against more recent variants of COVID-19. Some studies suggested improvements in patients with post-COVID-19 condition (PCC) following vaccination. We systematically explored available evidence on the effect of COVID-19 vaccines for the treatment of people with PCC. Methods: We conducted a rapid review of the literature following systematic approaches. We searched Medline (OVID), EMBASE (Elsevier), ClinicalTrials.gov, and the International Clinical Trials Registry Platform (ICTRP) for randomised trials, non-randomised trials, controlled before-after studies, and interrupted time-series studies of the effect of COVID-19 vaccines for treating people with PCC. Two independent review authors screened citations. Two review authors extracted data independently. We had planned to assess the risk of bias and use the GRADE approach (Grading of Recommendations, Assessment, Development, and Evaluation) to assess the certainty of evidence if there were completed studies. Results: We identified two ongoing randomised controlled trials. Both trials examine the effectiveness of therapeutic vaccines on PCC. The anticipated completion date of the CIMAvax-EGFA trial is January 2023, and the completion date of the COVID-19 mRNA vaccine trial is not stated. Conclusions: There is currently an absence of high‐quality evidence evaluating the effectiveness of COVID-19 vaccines for treating people with post-COVID-19 condition. The absence of published studies and only two ongoing trials highlight the need for additional studies on the effectiveness of vaccines for PCC. We recommend that researchers consider PCC as per the definition provided by the World Health Organization and use the available core outcome set for PCC in deciding which outcomes to measure and report in the trials. PROPSERO registration: CRD42022330821 (20/06/2022)
Collapse
|
173
|
Inhibition of major histocompatibility complex-I antigen presentation by sarbecovirus ORF7a proteins. Proc Natl Acad Sci U S A 2022; 119:e2209042119. [PMID: 36136978 PMCID: PMC9573092 DOI: 10.1073/pnas.2209042119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Viruses employ a variety of strategies to escape or counteract immune responses, including depletion of cell surface major histocompatibility complex class I (MHC-I), that would ordinarily present viral peptides to CD8+ cytotoxic T cells. As part of a screen to elucidate biological activities associated with individual severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) viral proteins, we found that ORF7a reduced cell surface MHC-I levels by approximately fivefold. Nevertheless, in cells infected with SARS-CoV-2, surface MHC-I levels were reduced even in the absence of ORF7a, suggesting additional mechanisms of MHC-I down-regulation. ORF7a proteins from a sample of sarbecoviruses varied in their ability to induce MHC-I down-regulation and, unlike SARS-CoV-2, the ORF7a protein from SARS-CoV lacked MHC-I downregulating activity. A single amino acid at position 59 (T/F) that is variable among sarbecovirus ORF7a proteins governed the difference in MHC-I downregulating activity. SARS-CoV-2 ORF7a physically associated with the MHC-I heavy chain and inhibited the presentation of expressed antigen to CD8+ T cells. Specifically, ORF7a prevented the assembly of the MHC-I peptide loading complex and caused retention of MHC-I in the endoplasmic reticulum. The differential ability of ORF7a proteins to function in this way might affect sarbecovirus dissemination and persistence in human populations, particularly those with infection- or vaccine-elicited immunity.
Collapse
|
174
|
DeWolf S, Laracy JC, Perales MA, Kamboj M, van den Brink MRM, Vardhana S. SARS-CoV-2 in immunocompromised individuals. Immunity 2022; 55:1779-1798. [PMID: 36182669 PMCID: PMC9468314 DOI: 10.1016/j.immuni.2022.09.006] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/24/2022] [Accepted: 09/08/2022] [Indexed: 12/15/2022]
Abstract
Immunocompromised individuals and particularly those with hematologic malignancies are at increased risk for SARS-CoV-2-associated morbidity and mortality due to immunologic deficits that limit prevention, treatment, and clearance of the virus. Understanding the natural history of viral infections in people with impaired immunity due to underlying conditions, immunosuppressive therapy, or a combination thereof has emerged as a critical area of investigation during the COVID-19 pandemic. Studies focused on these individuals have provided key insights into aspects of innate and adaptive immunity underlying both the antiviral immune response and excess inflammation in the setting of COVID-19. This review presents what is known about distinct states of immunologic vulnerability to SARS-CoV-2 and how this information can be harnessed to improve prevention and treatment strategies for immunologically high-risk populations.
Collapse
Affiliation(s)
- Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin C Laracy
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Mini Kamboj
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA; Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Santosha Vardhana
- Weill Cornell Medical College, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
175
|
Gregory DA, Trujillo M, Rushford C, Flury A, Kannoly S, San KM, Lyfoung DT, Wiseman RW, Bromert K, Zhou MY, Kesler E, Bivens NJ, Hoskins J, Lin CH, O’Connor DH, Wieberg C, Wenzel J, Kantor RS, Dennehy JJ, Johnson MC. Genetic diversity and evolutionary convergence of cryptic SARS- CoV-2 lineages detected via wastewater sequencing. PLoS Pathog 2022; 18:e1010636. [PMID: 36240259 PMCID: PMC9604950 DOI: 10.1371/journal.ppat.1010636] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/26/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022] Open
Abstract
Wastewater-based epidemiology (WBE) is an effective way of tracking the appearance and spread of SARS-COV-2 lineages through communities. Beginning in early 2021, we implemented a targeted approach to amplify and sequence the receptor binding domain (RBD) of SARS-COV-2 to characterize viral lineages present in sewersheds. Over the course of 2021, we reproducibly detected multiple SARS-COV-2 RBD lineages that have never been observed in patient samples in 9 sewersheds located in 3 states in the USA. These cryptic lineages contained between 4 to 24 amino acid substitutions in the RBD and were observed intermittently in the sewersheds in which they were found for as long as 14 months. Many of the amino acid substitutions in these lineages occurred at residues also mutated in the Omicron variant of concern (VOC), often with the same substitutions. One of the sewersheds contained a lineage that appeared to be derived from the Alpha VOC, but the majority of the lineages appeared to be derived from pre-VOC SARS-COV-2 lineages. Specifically, several of the cryptic lineages from New York City appeared to be derived from a common ancestor that most likely diverged in early 2020. While the source of these cryptic lineages has not been resolved, it seems increasingly likely that they were derived from long-term patient infections or animal reservoirs. Our findings demonstrate that SARS-COV-2 genetic diversity is greater than what is commonly observed through routine SARS-CoV-2 surveillance. Wastewater sampling may more fully capture SARS-CoV-2 genetic diversity than patient sampling and could reveal new VOCs before they emerge in the wider human population.
Collapse
Affiliation(s)
- Devon A. Gregory
- Department of Molecular Microbiology and Immunology, University of Missouri-School of Medicine, Columbia, Missouri, United States of America
| | - Monica Trujillo
- Department of Biological Sciences and Geology, Queensborough Community College of The City University of New York, New York City, New York, United States of America
| | - Clayton Rushford
- Department of Molecular Microbiology and Immunology, University of Missouri-School of Medicine, Columbia, Missouri, United States of America
| | - Anna Flury
- Biology Doctoral Program, The Graduate Center of The City University of New York, New York City, New York, United States of America
| | - Sherin Kannoly
- Biology Department, Queens College of The City University of New York, New York City, New York, United States of America
| | - Kaung Myat San
- Biology Department, Queens College of The City University of New York, New York City, New York, United States of America
| | - Dustin T. Lyfoung
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Roger W. Wiseman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Karen Bromert
- Genomics Technology Core, University of Missouri, Columbia, Missouri, United States of America
| | - Ming-Yi Zhou
- Genomics Technology Core, University of Missouri, Columbia, Missouri, United States of America
| | - Ellen Kesler
- Genomics Technology Core, University of Missouri, Columbia, Missouri, United States of America
| | - Nathan J. Bivens
- Genomics Technology Core, University of Missouri, Columbia, Missouri, United States of America
| | - Jay Hoskins
- Environmental Compliance Division, Engineering Department, Metropolitan St. Louis Sewer District, St. Louis, Missouri, United States of America
| | - Chung-Ho Lin
- Center of Agroforestry, School of Natural Resources, University of Missouri, Columbia, Missouri, United States of America
| | - David H. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Chris Wieberg
- Water Protection Program, Missouri Department of Natural Resources, Jefferson City, Missouri, United States of America
| | - Jeff Wenzel
- Bureau of Environmental Epidemiology, Division of Community and Public Health, Missouri Department of Health and Senior Services, Jefferson City, Missouri, United States of America
| | - Rose S. Kantor
- Department of Civil and Environmental Engineering, University of California, Berkeley, California, United States of America
| | - John J. Dennehy
- Biology Doctoral Program, The Graduate Center of The City University of New York, New York City, New York, United States of America
- Biology Department, Queens College of The City University of New York, New York City, New York, United States of America
| | - Marc C. Johnson
- Department of Molecular Microbiology and Immunology, University of Missouri-School of Medicine, Columbia, Missouri, United States of America
| |
Collapse
|
176
|
Herman JD, Atyeo C, Zur Y, Cook CE, Patel NJ, Vanni KM, Kowalski EN, Qian G, Shadick NA, Laffenburger D, Wallace ZS, Sparks JA, Alter G. Impact of cross-coronavirus immunity in post-acute sequelae of COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.09.25.22280335. [PMID: 36203557 PMCID: PMC9536039 DOI: 10.1101/2022.09.25.22280335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Beyond the unpredictable acute illness caused by SARS-CoV-2, one-fifth of infections unpredictably result in long-term persistence of symptoms despite the apparent clearance of infection. Insights into the mechanisms that underlie post-acute sequelae of COVID-19 (PASC) will be critical for the prevention and clinical management of long-term complications of COVID-19. Several hypotheses have been proposed that may account for the development of PASC, including persistence of virus or the dysregulation of immunity. Among the immunological changes noted in PASC, alterations in humoral immunity have been observed in some patient subsets. To begin to determine whether SARS-CoV-2 or other pathogen specific humoral immune responses evolve uniquely in PASC, we performed comprehensive antibody profiling against SARS-CoV-2 and a panel of endemic pathogens or routine vaccine antigens using Systems Serology in a cohort of patients with pre-existing rheumatic disease who either developed or did not develop PASC. A distinct humoral immune response was observed in individuals with PASC. Specifically, individuals with PASC harbored less inflamed and weaker Fcγ receptor binding anti-SARS-CoV-2 antibodies and a significantly expanded and more inflamed antibody response against endemic Coronavirus OC43. Individuals with PASC, further, generated more avid IgM responses and developed an expanded inflammatory OC43 S2-specific Fc-receptor binding response, linked to cross reactivity across SARS-CoV-2 and common coronaviruses. These findings implicate previous common Coronavirus imprinting as a marker for the development of PASC.
Collapse
Affiliation(s)
- Jonathan D Herman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Division of Infectious Disease, Brigham and Women's Hospital, Boston, MA, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Yonatan Zur
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Claire E Cook
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA
| | - Naomi J Patel
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen M Vanni
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Emily N Kowalski
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Grace Qian
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy A Shadick
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Douglas Laffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zachary S Wallace
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, MA, USA
| | - Jeffrey A Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
177
|
Clerbaux LA, Mayasich SA, Muñoz A, Soares H, Petrillo M, Albertini MC, Lanthier N, Grenga L, Amorim MJ. Gut as an Alternative Entry Route for SARS-CoV-2: Current Evidence and Uncertainties of Productive Enteric Infection in COVID-19. J Clin Med 2022; 11:5691. [PMID: 36233559 PMCID: PMC9573230 DOI: 10.3390/jcm11195691] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/17/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022] Open
Abstract
The gut has been proposed as a potential alternative entry route for SARS-CoV-2. This was mainly based on the high levels of SARS-CoV-2 receptor expressed in the gastrointestinal (GI) tract, the observations of GI disorders (such as diarrhea) in some COVID-19 patients and the detection of SARS-CoV-2 RNA in feces. However, the underlying mechanisms remain poorly understood. It has been proposed that SARS-CoV-2 can productively infect enterocytes, damaging the intestinal barrier and contributing to inflammatory response, which might lead to GI manifestations, including diarrhea. Here, we report a methodological approach to assess the evidence supporting the sequence of events driving SARS-CoV-2 enteric infection up to gut adverse outcomes. Exploring evidence permits to highlight knowledge gaps and current inconsistencies in the literature and to guide further research. Based on the current insights on SARS-CoV-2 intestinal infection and transmission, we then discuss the potential implication on clinical practice, including on long COVID. A better understanding of the GI implication in COVID-19 is still needed to improve disease management and could help identify innovative therapies or preventive actions targeting the GI tract.
Collapse
Affiliation(s)
| | - Sally A. Mayasich
- University of Wisconsin-Madison Aquatic Sciences Center at US EPA, Duluth, MN 55804, USA
| | - Amalia Muñoz
- European Commission, Joint Research Centre (JRC), 2440 Geel, Belgium
| | - Helena Soares
- Laboratory of Human Immunobiology and Pathogenesis, iNOVA4Health, Faculdade de Ciências Médicas—Nova Medical School, Universidade Nova de Lisboa, 1099-085 Lisbon, Portugal
| | | | | | - Nicolas Lanthier
- Laboratory of Hepatogastroenterology, Service d’Hépato-Gastroentérologie, Cliniques Universitaires Saint-Luc, UCLouvain, 1200 Brussels, Belgium
| | - Lucia Grenga
- Département Médicaments et Technologies pour la Santé, Commissariat à l’Énergie Atomique et aux Énergies Alternatives (CEA), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Université Paris-Saclay, 91190 Paris, France
| | - Maria-Joao Amorim
- Instituto Gulbenkian de Ciência, 2780-156 Lisbon, Portugal
- Católica Biomedical Research Centre, Católica Medical School, Universidade Católica Portuguesa, 1649-023 Lisbon, Portugal
| |
Collapse
|
178
|
Morello R, De Rose C, Cardinali S, Valentini P, Buonsenso D. Lactoferrin as Possible Treatment for Chronic Gastrointestinal Symptoms in Children with Long COVID: Case Series and Literature Review. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1446. [PMID: 36291381 PMCID: PMC9600357 DOI: 10.3390/children9101446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022]
Abstract
Long COVID is an emergent, heterogeneous, and multisystemic condition with an increasingly important impact also on the pediatric population. Among long COVID symptoms, patients can experience chronic gastrointestinal symptoms such as abdominal pain, constipation, diarrhea, vomiting, nausea, and dysphagia. Although there is no standard, agreed, and optimal diagnostic approach or treatment of long COVID in children, recently compounds containing multiple micronutrients and lactoferrin have been proposed as a possible treatment strategy, due to the long-standing experience gained from other gastrointestinal conditions. In particular, lactoferrin is a pleiotropic glycoprotein with antioxidant, anti-inflammatory, antithrombotic, and immunomodulatory activities. Moreover, it seems to have several physiological functions to protect the gastrointestinal tract. In this regard, we described the resolution of symptoms after the start of therapy with high doses of oral lactoferrin in two patients referred to our post-COVID pediatric unit due to chronic gastrointestinal symptoms after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Rosa Morello
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, Università Cattolica Sacro Cuore, 00168 Rome, Italy
| | - Cristina De Rose
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, Università Cattolica Sacro Cuore, 00168 Rome, Italy
| | - Sara Cardinali
- Department of Laboratory and Infectious Sciences, Università del Sacro Cuore, 20123 Rome, Italy
| | - Piero Valentini
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, Università Cattolica Sacro Cuore, 00168 Rome, Italy
| | - Danilo Buonsenso
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, Università Cattolica Sacro Cuore, 00168 Rome, Italy
- Department of Laboratory and Infectious Sciences, Università del Sacro Cuore, 20123 Rome, Italy
| |
Collapse
|
179
|
Goldman M. Long Covid, a great imitator of the 21th century. Front Med (Lausanne) 2022; 9:1026425. [PMID: 36186771 PMCID: PMC9519984 DOI: 10.3389/fmed.2022.1026425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
|
180
|
Madariaga-Mazón A, Naveja JJ, Becerra A, Alberto Campillo-Balderas J, Hernández-Morales R, Jácome R, Lazcano A, Martinez-Mayorga K. Subtle structural differences of nucleotide analogs may impact SARS-CoV-2 RNA-dependent RNA polymerase and exoribonuclease activity. Comput Struct Biotechnol J 2022; 20:5181-5192. [PMID: 36097553 PMCID: PMC9452397 DOI: 10.1016/j.csbj.2022.08.056] [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: 03/22/2022] [Revised: 08/05/2022] [Accepted: 08/27/2022] [Indexed: 11/17/2022] Open
Abstract
The rapid spread and public health impact of the novel SARS-CoV-2 variants that cause COVID-19 continue to produce major global impacts and social distress. Several vaccines were developed in record time to prevent and limit the spread of the infection, thus playing a pivotal role in controlling the pandemic. Although the repurposing of available drugs attempts to provide therapies of immediate access against COVID-19, there is still a need for developing specific treatments for this disease. Remdesivir, molnupiravir and Paxlovid remain the only evidence-supported antiviral drugs to treat COVID-19 patients, and only in severe cases. To contribute on the search of potential Covid-19 therapeutic agents, we targeted the viral RNA-dependent RNA polymerase (RdRp) and the exoribonuclease (ExoN) following two strategies. First, we modeled and analyzed nucleoside analogs sofosbuvir, remdesivir, favipiravir, ribavirin, and molnupiravir at three key binding sites on the RdRp-ExoN complex. Second, we curated and virtually screened a database containing 517 nucleotide analogs in the same binding sites. Finally, we characterized key interactions and pharmacophoric features presumably involved in viral replication halting at multiple sites. Our results highlight structural modifications that might lead to more potent SARS-CoV-2 inhibitors against an expansive range of variants and provide a collection of nucleotide analogs useful for screening campaigns.
Collapse
Affiliation(s)
- Abraham Madariaga-Mazón
- Instituto de Química Unidad Mérida, Universidad Nacional Autónoma de México, Carretera Mérida-Tetiz Km. 4.5, Ucú, Yucatán, Mexico.,Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas Unidad Mérida, Universidad Nacional Autónoma de México, Sierra Papacál Mérida, Yucatán 97302, Mexico
| | - José J Naveja
- Instituto de Química Unidad Mérida, Universidad Nacional Autónoma de México, Carretera Mérida-Tetiz Km. 4.5, Ucú, Yucatán, Mexico.,Institute for Molecular Biology and University Cancer Center (UCT) Mainz, Germany
| | - Arturo Becerra
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | - Rodrigo Jácome
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Antonio Lazcano
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico.,El Colegio Nacional, Mexico City, Mexico
| | - Karina Martinez-Mayorga
- Instituto de Química Unidad Mérida, Universidad Nacional Autónoma de México, Carretera Mérida-Tetiz Km. 4.5, Ucú, Yucatán, Mexico.,Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas Unidad Mérida, Universidad Nacional Autónoma de México, Sierra Papacál Mérida, Yucatán 97302, Mexico
| |
Collapse
|
181
|
Kumari M, Lu RM, Li MC, Huang JL, Hsu FF, Ko SH, Ke FY, Su SC, Liang KH, Yuan JPY, Chiang HL, Sun CP, Lee IJ, Li WS, Hsieh HP, Tao MH, Wu HC. A critical overview of current progress for COVID-19: development of vaccines, antiviral drugs, and therapeutic antibodies. J Biomed Sci 2022; 29:68. [PMID: 36096815 PMCID: PMC9465653 DOI: 10.1186/s12929-022-00852-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/01/2022] [Indexed: 12/12/2022] Open
Abstract
The novel coronavirus disease (COVID-19) pandemic remains a global public health crisis, presenting a broad range of challenges. To help address some of the main problems, the scientific community has designed vaccines, diagnostic tools and therapeutics for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The rapid pace of technology development, especially with regard to vaccines, represents a stunning and historic scientific achievement. Nevertheless, many challenges remain to be overcome, such as improving vaccine and drug treatment efficacies for emergent mutant strains of SARS-CoV-2. Outbreaks of more infectious variants continue to diminish the utility of available vaccines and drugs. Thus, the effectiveness of vaccines and drugs against the most current variants is a primary consideration in the continual analyses of clinical data that supports updated regulatory decisions. The first two vaccines granted Emergency Use Authorizations (EUAs), BNT162b2 and mRNA-1273, still show more than 60% protection efficacy against the most widespread current SARS-CoV-2 variant, Omicron. This variant carries more than 30 mutations in the spike protein, which has largely abrogated the neutralizing effects of therapeutic antibodies. Fortunately, some neutralizing antibodies and antiviral COVID-19 drugs treatments have shown continued clinical benefits. In this review, we provide a framework for understanding the ongoing development efforts for different types of vaccines and therapeutics, including small molecule and antibody drugs. The ripple effects of newly emergent variants, including updates to vaccines and drug repurposing efforts, are summarized. In addition, we summarize the clinical trials supporting the development and distribution of vaccines, small molecule drugs, and therapeutic antibodies with broad-spectrum activity against SARS-CoV-2 strains.
Collapse
Affiliation(s)
- Monika Kumari
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529, Taiwan
| | - Ruei-Min Lu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Mu-Chun Li
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Jhih-Liang Huang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Fu-Fei Hsu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Shih-Han Ko
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Feng-Yi Ke
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529, Taiwan
| | - Shih-Chieh Su
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529, Taiwan
| | - Kang-Hao Liang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Joyce Pei-Yi Yuan
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Hsiao-Ling Chiang
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
| | - Cheng-Pu Sun
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - I-Jung Lee
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Wen-Shan Li
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
- Institute of Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Hsing-Pang Hsieh
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Mi-Hua Tao
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Han-Chung Wu
- Biomedical Translation Research Center (BioTReC), Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Nankang District, Taipei, 11529, Taiwan.
| |
Collapse
|
182
|
Goh D, Lim JCT, Fernaíndez SB, Joseph CR, Edwards SG, Neo ZW, Lee JN, Caballero SG, Lau MC, Yeong JPS. Case report: Persistence of residual antigen and RNA of the SARS-CoV-2 virus in tissues of two patients with long COVID. Front Immunol 2022; 13:939989. [PMID: 36131932 PMCID: PMC9483160 DOI: 10.3389/fimmu.2022.939989] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
The World Health Organization has defined long COVID-19 (LC) as a condition that occurs in individuals with a history of SARS-CoV-2 infection who exhibit persistent symptoms after its acute phase that last for at least two months and cannot be explained by an alternative diagnosis. Since we had previously reported residual viral antigens in tissues of convalescent patients, we aimed to assess the presence of such antigens in long COVID tissues. Here, we established the presence of the residual virus in the appendix, skin, and breast tissues of 2 patients who exhibited LC symptoms 163 and 426 days after symptom onset. With multiplex immunohistochemistry, we detected viral nucleocapsid protein in all three tissues. The nucleocapsid protein was further observed to colocalize with macrophage marker CD68, suggesting that immune cells were direct targets of SARS-CoV-2. Additionally, using RNAscope, the presence of viral RNA was also detected. Our positive finding in the breast tissue is corroborated by the recent reports of immunocompromised patients experiencing LC symptoms and persistent viral replication. Overall, our findings and emerging LC studies raise the possibility that the gastrointestinal tract may function as a reservoir for SARS-CoV-2.
Collapse
Affiliation(s)
- Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | | | - Craig Ryan Joseph
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | | | - Zhen Wei Neo
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Justina Nadia Lee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | | | - Mai Chan Lau
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Joe Poh Sheng Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- *Correspondence: Joe Poh Sheng Yeong,
| |
Collapse
|
183
|
Rohrhofer J, Graninger M, Lettenmaier L, Schweighardt J, Gentile SA, Koidl L, Ret D, Stingl M, Puchhammer‐Stöckl E, Untersmayr E. Association between Epstein-Barr-Virus reactivation and development of Long-COVID fatigue. Allergy 2022; 78:297-299. [PMID: 35950630 PMCID: PMC9538037 DOI: 10.1111/all.15471] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 12/30/2022]
Affiliation(s)
- Johanna Rohrhofer
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | | | - Lisa Lettenmaier
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Johannes Schweighardt
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Salvatore Alessio Gentile
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Larissa Koidl
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Davide Ret
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria,Division of Macromolecular Chemistry, Institute of Applied Synthetic ChemistryVienna University of TechnologyViennaAustria
| | | | | | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| |
Collapse
|
184
|
Leppkes M, Neurath MF. Rear Window-What Can the Gut Tell Us About Long-COVID? Gastroenterology 2022; 163:376-378. [PMID: 35660031 PMCID: PMC9175081 DOI: 10.1053/j.gastro.2022.05.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 01/01/2023]
Affiliation(s)
- Moritz Leppkes
- Department of Medicine 1, University Clinic Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany, and, Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany.
| | | |
Collapse
|
185
|
Hydroxytyrosol Recovers SARS-CoV-2-PLpro-Dependent Impairment of Interferon Related Genes in Polarized Human Airway, Intestinal and Liver Epithelial Cells. Antioxidants (Basel) 2022; 11:antiox11081466. [PMID: 36009185 PMCID: PMC9404978 DOI: 10.3390/antiox11081466] [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: 06/20/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/10/2022] Open
Abstract
The SARS-CoV-2 pandemic has caused approximately 6.3 million deaths, mainly due to the acute respiratory distress syndrome or multi-organ failure that characterizes COVID-19 acute disease. Post-acute COVID-19 syndrome, also known as long-COVID, is a condition characterized by a complex of symptoms that affects 10–20% of the individuals who have recovered from the infection. Scientific and clinical evidence demonstrates that long-COVID can develop in both adults and children. It has been hypothesized that multi-organ effects of long-COVID could be associated with the persistence of virus RNA/proteins in host cells, but the real mechanism remains to be elucidated. Therefore, we sought to determine the effects of the exogenous expression of the papain-like protease (PLpro) domain of the non-structural protein (NSP3) of SARS-CoV-2 in polarized human airway (Calu-3), intestinal (Caco-2), and liver (HepG2) epithelial cells, and to evaluate the ability of the natural antioxidant hydroxytyrosol (HXT) in neutralizing these effects. Our results demonstrated that PLpro was able to induce a cascade of inflammatory genes and proteins (mainly associated with the interferon pathway) and increase the apoptotic rate and expression of several oxidative stress markers in all evaluated epithelial cells. Noteably, the treatment with 10 μM HXT reverted PL-pro-dependent effects almost completely. This study provides the first evidence that SARS-CoV-2 PLpro remaining in host cells after viral clearance may contribute to the pathogenetic mechanisms of long-COVID. These effects may be counteracted by natural antioxidants. Further clinical and experimental studies are necessary to confirm this hypothesis.
Collapse
|
186
|
Peluso MJ, Deveau TM, Munter SE, Ryder D, Buck A, Beck-Engeser G, Chan F, Lu S, Goldberg SA, Hoh R, Tai V, Torres L, Iyer NS, Deswal M, Ngo LH, Buitrago M, Rodriguez A, Chen JY, Yee BC, Chenna A, Winslow JW, Petropoulos CJ, Deitchman AN, Hellmuth J, Spinelli MA, Durstenfeld MS, Hsue PY, Kelly JD, Martin JN, Deeks SG, Hunt PW, Henrich TJ. Impact of Pre-Existing Chronic Viral Infection and Reactivation on the Development of Long COVID. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.06.21.22276660. [PMID: 35898346 PMCID: PMC9327632 DOI: 10.1101/2022.06.21.22276660] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The presence and reactivation of chronic viral infections such as Epstein-Barr virus (EBV), cytomegalovirus (CMV) and human immunodeficiency virus (HIV) have been proposed as potential contributors to Long COVID (LC), but studies in well-characterized post-acute cohorts of individuals with COVID-19 over a longer time course consistent with current case definitions of LC are limited. In a cohort of 280 adults with prior SARS-CoV-2 infection, we observed that LC symptoms such as fatigue and neurocognitive dysfunction at a median of 4 months following initial diagnosis were independently associated with serological evidence of recent EBV reactivation (early antigen-D [EA-D] IgG positivity) or high nuclear antigen IgG levels, but not with ongoing EBV viremia. Evidence of EBV reactivation (EA-D IgG) was most strongly associated with fatigue (OR 2.12). Underlying HIV infection was also independently associated with neurocognitive LC (OR 2.5). Interestingly, participants who had serologic evidence of prior CMV infection were less likely to develop neurocognitive LC (OR 0.52) and tended to have less severe (>5 symptoms reported) LC (OR 0.44). Overall, these findings suggest differential effects of chronic viral co-infections on the likelihood of developing LC and predicted distinct syndromic patterns. Further assessment during the acute phase of COVID-19 is warranted. SUMMARY The authors found that Long COVID symptoms in a post-acute cohort were associated with serological evidence of recent EBV reactivation and pre-existing HIV infection when adjusted for participant factors, sample timing, comorbid conditions and prior hospitalization, whereas underlying CMV infection was associated with a decreased risk of Long COVID.
Collapse
Affiliation(s)
- Michael J. Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Tyler-Marie Deveau
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Sadie E. Munter
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Dylan Ryder
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Amanda Buck
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | | | - Fay Chan
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Scott Lu
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Sarah A. Goldberg
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Viva Tai
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Leonel Torres
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Nikita S. Iyer
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Monika Deswal
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Lynn H. Ngo
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Melissa Buitrago
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Antonio Rodriguez
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Jessica Y. Chen
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | | | - Ahmed Chenna
- Monogram Biosciences Inc., South San Francisco, CA, USA
| | | | | | | | - Joanna Hellmuth
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew A. Spinelli
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | | | - Priscilla Y. Hsue
- Division of Cardiology, University of California, San Francisco, CA, USA
| | - J. Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - Peter W. Hunt
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Timothy J. Henrich
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
187
|
Noval Rivas M, Porritt RA, Cheng MH, Bahar I, Arditi M. Multisystem Inflammatory Syndrome in Children and Long COVID: The SARS-CoV-2 Viral Superantigen Hypothesis. Front Immunol 2022; 13:941009. [PMID: 35874696 PMCID: PMC9300823 DOI: 10.3389/fimmu.2022.941009] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a febrile pediatric inflammatory disease that may develop weeks after initial SARS-CoV-2 infection or exposure. MIS-C involves systemic hyperinflammation and multiorgan involvement, including severe cardiovascular, gastrointestinal (GI) and neurological symptoms. Some clinical attributes of MIS-C-such as persistent fever, rashes, conjunctivitis and oral mucosa changes (red fissured lips and strawberry tongue)-overlap with features of Kawasaki disease (KD). In addition, MIS-C shares striking clinical similarities with toxic shock syndrome (TSS), which is triggered by bacterial superantigens (SAgs). The remarkable similarities between MIS-C and TSS prompted a search for SAg-like structures in the SARS-CoV-2 virus and the discovery of a unique SAg-like motif highly similar to a Staphylococcal enterotoxin B (SEB) fragment in the SARS-CoV-2 spike 1 (S1) glycoprotein. Computational studies suggest that the SAg-like motif has a high affinity for binding T-cell receptors (TCRs) and MHC Class II proteins. Immunosequencing of peripheral blood samples from MIS-C patients revealed a profound expansion of TCR β variable gene 11-2 (TRBV11-2), which correlates with MIS-C severity and serum cytokine levels, consistent with a SAg-triggered immune response. Computational sequence analysis of SARS-CoV-2 spike further identified conserved neurotoxin-like motifs which may alter neuronal cell function and contribute to neurological symptoms in COVID-19 and MIS-C patients. Additionally, autoantibodies are detected during MIS-C, which may indicate development of post-SARS-CoV-2 autoreactive and autoimmune responses. Finally, prolonged persistence of SARS-CoV-2 RNA in the gut, increased gut permeability and elevated levels of circulating S1 have been observed in children with MIS-C. Accordingly, we hypothesize that continuous and prolonged exposure to the viral SAg-like and neurotoxin-like motifs in SARS-CoV-2 spike may promote autoimmunity leading to the development of post-acute COVID-19 syndromes, including MIS-C and long COVID, as well as the neurological complications resulting from SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rebecca A Porritt
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Mary Hongying Cheng
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
188
|
Neuropsychological Predictors of Fatigue in Post-COVID Syndrome. J Clin Med 2022; 11:jcm11133886. [PMID: 35807173 PMCID: PMC9267301 DOI: 10.3390/jcm11133886] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 02/01/2023] Open
Abstract
Fatigue is one of the most disabling symptoms in several neurological disorders and has an important cognitive component. However, the relationship between self-reported cognitive fatigue and objective cognitive assessment results remains elusive. Patients with post-COVID syndrome often report fatigue and cognitive issues several months after the acute infection. We aimed to develop predictive models of fatigue using neuropsychological assessments to evaluate the relationship between cognitive fatigue and objective neuropsychological assessment results. We conducted a cross-sectional study of 113 patients with post-COVID syndrome, assessing them with the Modified Fatigue Impact Scale (MFIS) and a comprehensive neuropsychological battery including standardized and computerized cognitive tests. Several machine learning algorithms were developed to predict MFIS scores (total score and cognitive fatigue score) based on neuropsychological test scores. MFIS showed moderate correlations only with the Stroop Color–Word Interference Test. Classification models obtained modest F1-scores for classification between fatigue and non-fatigued or between 3 or 4 degrees of fatigue severity. Regression models to estimate the MFIS score did not achieve adequate R2 metrics. Our study did not find reliable neuropsychological predictors of cognitive fatigue in the post-COVID syndrome. This has important implications for the interpretation of fatigue and cognitive assessment. Specifically, MFIS cognitive domain could not properly capture actual cognitive fatigue. In addition, our findings suggest different pathophysiological mechanisms of fatigue and cognitive dysfunction in post-COVID syndrome.
Collapse
|
189
|
Multicompartmental Mathematical Model of SARS-CoV-2 Distribution in Human Organs and Their Treatment. MATHEMATICS 2022. [DOI: 10.3390/math10111925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patients with COVID-19 can develop pneumonia, severe symptoms of acute respiratory distress syndrome, and multiple organ failure. Nevertheless, the variety of forms of this disease requires further research on the pathogenesis of this disease. Based on the analysis of published data and original experiments on the concentrations of SARS-CoV-2 in biological fluids of the nasopharynx, lungs, and intestines and using a developed modular model of the virus distribution in human tissue and organs, an assessment of the SARS-CoV-2 reproduction in various compartments of the body is presented. Most of the viral particles can transport to the esophagus from the nasopharynx. The viral particles entering the gastrointestinal tract will obviously be accompanied by the infection of the intestinal epithelium and accumulation of the virus in the intestinal lumen in an amount proportional to their secretory and protein-synthetic activities. The relatively low concentration of SARS-CoV-2 in tissues implies an essential role of transport processes and redistribution of the virus from the nasopharynx and intestines to the lungs. The model simulations also suppose that sanitation of the nasopharynx mucosa at the initial stage of the infectious process has prospects for the use in medical practice.
Collapse
|
190
|
Gregory DA, Trujillo M, Rushford C, Flury A, Kannoly S, San KM, Lyfoung D, Wiseman RW, Bromert K, Zhou MY, Kesler E, Bivens N, Hoskins J, Lin CH, O'Connor DH, Wieberg C, Wenzel J, Kantor RS, Dennehy JJ, Johnson MC. Genetic Diversity and Evolutionary Convergence of Cryptic SARS-CoV-2 Lineages Detected Via Wastewater Sequencing. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.06.03.22275961. [PMID: 35677072 PMCID: PMC9176656 DOI: 10.1101/2022.06.03.22275961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Wastewater-based epidemiology (WBE) is an effective way of tracking the appearance and spread of SARS-COV-2 lineages through communities. Beginning in early 2021, we implemented a targeted approach to amplify and sequence the receptor binding domain (RBD) of SARS-COV-2 to characterize viral lineages present in sewersheds. Over the course of 2021, we reproducibly detected multiple SARS-COV-2 RBD lineages that have never been observed in patient samples in 9 sewersheds located in 3 states in the USA. These cryptic lineages contained between 4 to 24 amino acid substitutions in the RBD and were observed intermittently in the sewersheds in which they were found for as long as 14 months. Many of the amino acid substitutions in these lineages occurred at residues also mutated in the Omicron variant of concern (VOC), often with the same substitution. One of the sewersheds contained a lineage that appeared to be derived from the Alpha VOC, but the majority of the lineages appeared to be derived from pre-VOC SARS-COV-2 lineages. Specifically, several of the cryptic lineages from New York City appeared to be derived from a common ancestor that most likely diverged in early 2020. While the source of these cryptic lineages has not been resolved, it seems increasingly likely that they were derived from immunocompromised patients or animal reservoirs. Our findings demonstrate that SARS-COV-2 genetic diversity is greater than what is commonly observed through routine SARS-CoV-2 surveillance. Wastewater sampling may more fully capture SARS-CoV-2 genetic diversity than patient sampling and could reveal new VOCs before they emerge in the wider human population. Author Summary During the COVID-19 pandemic, wastewater-based epidemiology has become an effective public health tool. Because many infected individuals shed SARS-CoV-2 in feces, wastewater has been monitored to reveal infection trends in the sewersheds from which the samples were derived. Here we report novel SARS-CoV-2 lineages in wastewater samples obtained from 3 different states in the USA. These lineages appeared in specific sewersheds intermittently over periods of up to 14 months, but generally have not been detected beyond the sewersheds in which they were initially found. Many of these lineages may have diverged in early 2020. Although these lineages share considerable overlap with each other, they have never been observed in patients anywhere in the world. While the wastewater lineages have similarities with lineages observed in long-term infections of immunocompromised patients, animal reservoirs cannot be ruled out as a potential source.
Collapse
Affiliation(s)
- Devon A Gregory
- Department of Molecular Microbiology and Immunology, University of Missouri-School of Medicine, Columbia, MO, USA
| | - Monica Trujillo
- Department of Biological Sciences and Geology, Queensborough Community College of The City University of New York, Queens, NY, USA
| | - Clayton Rushford
- Department of Molecular Microbiology and Immunology, University of Missouri-School of Medicine, Columbia, MO, USA
| | - Anna Flury
- Biology Doctoral Program, The Graduate Center of The City University of New York, NYC, NY, USA
| | - Sherin Kannoly
- Biology Department, Queens College of The City University of New York, Queens, NY, USA 11367
| | - Kaung Myat San
- Biology Department, Queens College of The City University of New York, Queens, NY, USA 11367
| | - Dustin Lyfoung
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA 53706
| | - Roger W Wiseman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA 53706
| | - Karen Bromert
- Genomics Technology Core, University of Missouri, Columbia, MO, USA
| | - Ming-Yi Zhou
- Genomics Technology Core, University of Missouri, Columbia, MO, USA
| | - Ellen Kesler
- Genomics Technology Core, University of Missouri, Columbia, MO, USA
| | - Nathan Bivens
- Genomics Technology Core, University of Missouri, Columbia, MO, USA
| | - Jay Hoskins
- Environmental Compliance Division, Engineering Department, Metropolitan St. Louis Sewer District, St. Louis, MO, USA 63103
| | - Chung-Ho Lin
- Center of Agroforestry, School of Natural Resources, University of Missouri, Columbia, MO, USA
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA 53706
| | - Chris Wieberg
- Water Protection Program, Missouri Department of Natural Resources, Jefferson City, MO, USA
| | - Jeff Wenzel
- Bureau of Environmental Epidemiology, Division of Community and Public Health, Missouri Department of Health and Senior Services, Jefferson City, MO, USA
| | - Rose S Kantor
- Department of Civil and Environmental Engineering, University of California, Berkeley, 663 Davis Hall, Berkeley, CA, USA 94720
| | - John J Dennehy
- Biology Doctoral Program, The Graduate Center of The City University of New York, NYC, NY, USA
- Biology Department, Queens College of The City University of New York, Queens, NY, USA 11367
| | - Marc C Johnson
- Department of Molecular Microbiology and Immunology, University of Missouri-School of Medicine, Columbia, MO, USA
| |
Collapse
|
191
|
Zhang F, Zang T, Stevenson EM, Lei X, Copertino DC, Mota TM, Boucau J, Garcia-Beltran WF, Jones RB, Bieniasz PD. Inhibition of major histocompatibility complex-I antigen presentation by sarbecovirus ORF7a proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022. [PMID: 35665005 PMCID: PMC9164438 DOI: 10.1101/2022.05.25.493467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Viruses employ a variety of strategies to escape or counteract immune responses, including depletion of cell surface major histocompatibility complex class I (MHC-I), that would ordinarily present viral peptides to CD8+ cytotoxic T cells. As part of a screen to elucidate biological activities associated with individual SARS-CoV-2 viral proteins, we found that ORF7a reduced cell surface MHC-I levels by approximately 5-fold. Nevertheless, in cells infected with SARS-CoV-2, surface MHC-I levels were reduced even in the absence of ORF7a, suggesting additional mechanisms of MHC-I downregulation. ORF7a proteins from a sample of sarbecoviruses varied in their ability to induce MHC-I downregulation and, unlike SARS-CoV-2, the ORF7a protein from SARS-CoV lacked MHC-I downregulating activity. A single-amino acid at position 59 (T/F) that is variable among sarbecovirus ORF7a proteins governed the difference in MHC-I downregulating activity. SARS-CoV-2 ORF7a physically associated with the MHC-I heavy chain and inhibited the presentation of expressed antigen to CD8+ T-cells. Speficially, ORF7a prevented the assembly of the MHC-I peptide loading complex and causing retention of MHC-I in the endoplasmic reticulum. The differential ability of ORF7a proteins to function in this way might affect sarbecovirus dissemination and persistence in human populations, particularly those with infection- or vaccine-elicited immunity.
Collapse
|
192
|
|
193
|
Nersisyan SA. Induction of Hypoxic Response in Caco-2 Cells Promote the Expression of Genes Involved in SARS-CoV-2 Endocytosis and Transcytosis. DOKL BIOCHEM BIOPHYS 2022; 506:206-209. [PMID: 36303053 PMCID: PMC9612616 DOI: 10.1134/s1607672922050118] [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: 06/08/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/07/2022]
Abstract
In the present manuscript we analyzed the influence of hypoxic response in Caco-2 cells on the expression of genes and miRNAs involved in the mechanisms of intracellular transport of SARS-CoV-2 viral particles, especially endocytosis and transcytosis. With the use of RNA sequencing of Caco-2 cells treated with hypoxia-inducing oxyquinoline derivative, we showed two-fold increase in the expression of the main SARS-CoV-2 receptor ACE2. Expression of the non-canonical receptor TFRC was also elevated. We also observed a significant increase in the expression levels of genes from the low-density lipoprotein (LDL) receptor family, which play a crucial role in the transcytosis: LDLR, LRP1, LRP4, and LRP5. Upregulation of LDLR was coupled with the downregulation of hsa-miR-148a-3p, which can directly bind to LDLR mRNA. Thus, the hypoxic response in Caco-2 cells includes upregulation of genes involved in the mechanisms of endocytosis and transcytosis of SARS-CoV-2 viral particles.
Collapse
Affiliation(s)
- S. A. Nersisyan
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia ,Institute of Molecular Biology (IMB), National Academy of Sciences of the Republic of Armenia, Yerevan, Armenia
| |
Collapse
|