1
|
Sherchan S, Ikner LA, Gerba CP. Inactivation of SARS-CoV-2 in Water by Chlorination. FOOD AND ENVIRONMENTAL VIROLOGY 2023; 15:262-264. [PMID: 37421543 DOI: 10.1007/s12560-023-09559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is present in both respiratory secretions and feces, creating its potential for transmission by swimming pools. Recreational water activity is known to be at increased risk of respiratory infections and respiratory viruses have caused been detected and have caused outbreaks in swimming pools. However, little is known regarding the chlorine inactivation of SARS-CoV-2 in water typical of swimming pools in the USA. In this study, the inactivation of SARS-CoV-2 Isolate hCoV-19/USA-WA1/2020 was observed in water by chlorination. All experiments were conducted within a BSL-3 laboratory at room temperature. Our results show that the virus was reduced by 3.5 log (> 99.9%) after 30 s of 2.05-mg/L free chlorine contact and greater than 4.17 log (limit of detection) (> 99.99%) within 2 min.
Collapse
Affiliation(s)
- Samendra Sherchan
- Department of Environmental Health Sciences, Tulane University, New Orleans, LA, 70112, USA.
- BioEnvironmental Science Program, Morgan State University, Baltimore, MD, 21251, USA.
| | - Luisa A Ikner
- Department of Environmental Science, University of Arizona, Tucson, AZ, USA
| | - Charles P Gerba
- Department of Environmental Science, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
2
|
Vemuri K, Radi SH, Sladek FM, Verzi MP. Multiple roles and regulatory mechanisms of the transcription factor HNF4 in the intestine. Front Endocrinol (Lausanne) 2023; 14:1232569. [PMID: 37635981 PMCID: PMC10450339 DOI: 10.3389/fendo.2023.1232569] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Hepatocyte nuclear factor 4-alpha (HNF4α) drives a complex array of transcriptional programs across multiple organs. Beyond its previously documented function in the liver, HNF4α has crucial roles in the kidney, intestine, and pancreas. In the intestine, a multitude of functions have been attributed to HNF4 and its accessory transcription factors, including but not limited to, intestinal maturation, differentiation, regeneration, and stem cell renewal. Functional redundancy between HNF4α and its intestine-restricted paralog HNF4γ, and co-regulation with other transcription factors drive these functions. Dysregulated expression of HNF4 results in a wide range of disease manifestations, including the development of a chronic inflammatory state in the intestine. In this review, we focus on the multiple molecular mechanisms of HNF4 in the intestine and explore translational opportunities. We aim to introduce new perspectives in understanding intestinal genetics and the complexity of gastrointestinal disorders through the lens of HNF4 transcription factors.
Collapse
Affiliation(s)
- Kiranmayi Vemuri
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Sarah H. Radi
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry, University of California, Riverside, Riverside, CA, United States
| | - Frances M. Sladek
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Michael P. Verzi
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
3
|
Bauer L, Rissmann M, Benavides FFW, Leijten L, van Run P, Begeman L, Veldhuis Kroeze EJB, Lendemeijer B, Smeenk H, de Vrij FMS, Kushner SA, Koopmans MPG, Rockx B, van Riel D. In vitro and in vivo differences in neurovirulence between D614G, Delta And Omicron BA.1 SARS-CoV-2 variants. Acta Neuropathol Commun 2022; 10:124. [PMID: 36058935 PMCID: PMC9441226 DOI: 10.1186/s40478-022-01426-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/09/2022] [Indexed: 01/16/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with various neurological complications. Although the mechanism is not fully understood, several studies have shown that neuroinflammation occurs in the acute and post-acute phase. As these studies have predominantly been performed with isolates from 2020, it is unknown if there are differences among SARS-CoV-2 variants in their ability to cause neuroinflammation. Here, we compared the neuroinvasiveness, neurotropism and neurovirulence of the SARS-CoV-2 ancestral strain D614G, the Delta (B.1.617.2) and Omicron BA.1 (B.1.1.529) variants using in vitro and in vivo models. The Omicron BA.1 variant showed reduced neurotropism and neurovirulence compared to Delta and D614G in human induced pluripotent stem cell (hiPSC)-derived cortical neurons co-cultured with astrocytes. Similar differences were obtained in Syrian hamsters inoculated with D614G, Delta and the Omicron BA.1 variant 5 days post infection. Replication in the olfactory mucosa was observed in all hamsters, but most prominently in D614G inoculated hamsters. Furthermore, neuroinvasion into the CNS via the olfactory nerve was observed in D614G, but not Delta or Omicron BA.1 inoculated hamsters. Furthermore, neuroinvasion was associated with neuroinflammation in the olfactory bulb of hamsters inoculated with D614G. Altogether, our findings suggest differences in the neuroinvasive, neurotropic and neurovirulent potential between SARS-CoV-2 variants using in vitro hiPSC-derived neural cultures and in vivo in hamsters during the acute phase of the infection.
Collapse
Affiliation(s)
- Lisa Bauer
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Melanie Rissmann
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Lonneke Leijten
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Peter van Run
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lineke Begeman
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Bas Lendemeijer
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hilde Smeenk
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Femke M S de Vrij
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Debby van Riel
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Akkız H. The Biological Functions and Clinical Significance of SARS-CoV-2 Variants of Corcern. Front Med (Lausanne) 2022; 9:849217. [PMID: 35669924 PMCID: PMC9163346 DOI: 10.3389/fmed.2022.849217] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is continuing to evolve, emerging novel variants with spike protein mutations. Although most mutations emerged in the SARS-CoV-2 genome are neutral or mildly deleterious, a small number of mutations can affect virus phenotype that confers the virus a fitness advantage. These mutations can enhance viral replication, raise the risk of reinfection and blunt the potency of neutralizing antibodies triggered by previous infection and vaccination. Since December 2020, the SARS-CoV-2 has emerged five quickly spreading strains, designated variants of concern (VOCs), including the Alpha (B.1.1.7) variant, the Beta (B.1.351) variant, the Gamma (P.1) variant, the Delta (B.1.617.2) variant and the Omicron (B.1.1.529) variant. These variants have a high number of the mutations in the spike protein that promotes viral cell entry through the angiotensin-converting enzyme -2 (ACE2). Mutations that have arisen in the receptor binding domain (RBD) of the spike protein are of great concern due to their potential to evade neutralizing antibodies triggered by previous infection and vaccines. The Alpha variant emerged in the United Kingdom in the second half of 2020 that has spread quickly globally and acquired the E484K mutation in the United Kingdom and the United States. The Beta and Gamma variants emerged in South Africa and Brazil, respectively, that have additional mutations at positions E484 and K417 in the RBD. SARS-CoV-2 variants containing the combination of N501Y, E484K, and K417N/T mutations exhibit remarkably decreased sensitivity to neutralizing antibodies mediated by vaccination or previous infection. The Gamma variant may result in more severe disease than other variants do even in convalescent individuals. The Delta variant emerged in India in December 2020 and has spread to many countries including the United States and the United Kingdom. The Delta variant has 8 mutations in the spike protein, some of which can influence immune responses to the key antigenic regions of RBD. In early November 2021, the Omicron (B.1.1.529) variant was first detected in Botswana and South Africa. The Omicron variant harbors more than 30 mutations in the spike protein, many of which are located within the RBD, which have been associated with increased transmissibility and immune evasion after previous infection and vaccination. Additionally, the Omicron variant contains 3 deletions and one insertion in the spike protein. Recently, the Omicron variant has been classified into three sublineages, including BA.1, BA.2, and BA.3, with strikingly different genetic characteristics. The Omicron BA.2 sublineage has different virological landscapes, such as transmissibility, pathogenicity and resistance to the vaccine-induced immunity compared to BA.1 and BA.3 sublineages. Mutations emerged in the RBD of the spike protein of VOCs increase viral replication, making the virus more infectious and more transmissible and enable the virus to evade vaccine-elicited neutralizing antibodies. Unfortunately, the emergence of novel SARS-CoV-2 VOCs has tempered early optimism regarding the efficacy of COVID-19 vaccines. This review addresses the biological and clinical significance of SARS-CoV-2 VOCs and their impact on neutralizing antibodies mediated by existing COVID-19 vaccines.
Collapse
Affiliation(s)
- Hikmet Akkız
- Department of Gastroenterology and Hepatology, The University of Çukurova, Adana, Turkey
| |
Collapse
|
5
|
Mendonça-Gomes JM, Charlie-Silva I, Guimarães ATB, Estrela FN, Calmon MF, Miceli RN, Sanches PRS, Bittar C, Rahal P, Cilli EM, Ahmed MAI, Vogel CFA, Malafaia G. Shedding light on toxicity of SARS-CoV-2 peptides in aquatic biota: A study involving neotropical mosquito larvae (Diptera: Culicidae). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 289:117818. [PMID: 34333265 PMCID: PMC8291650 DOI: 10.1016/j.envpol.2021.117818] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/21/2021] [Accepted: 07/19/2021] [Indexed: 05/19/2023]
Abstract
Knowledge about how the COVID-19 pandemic can affect aquatic wildlife is still extremely limited, and no effect of SARS-CoV-2 or its structural constituents on invertebrate models has been reported so far. Thus, we investigated the presence of the 2019-new coronavirus in different urban wastewater samples and, later, evaluated the behavioral and biochemical effects of the exposure of Culex quinquefasciatus larvae to two SARS-CoV-2 spike protein peptides (PSPD-2002 and PSPD-2003) synthesized in our laboratory. Initially, our results show the contamination of wastewater by the new coronavirus, via RT-qPCR on the viral N1 gene. On the other hand, our study shows that short-term exposure (48 h) to a low concentration (40 μg/L) of the synthesized peptides induced changes in the locomotor and the olfactory-driven behavior of the C. quinquefascitus larvae, which were associated with increased production of ROS and AChE activity (cholinesterase effect). To our knowledge, this is the first study that reports the indirect effects of the COVID-19 pandemic on the larval phase of a freshwater invertebrate species. The results raise concerns at the ecological level where the observed biological effects may lead to drastic consequences.
Collapse
Affiliation(s)
| | - Ives Charlie-Silva
- Departamento de Farmacologia, Instituto de Ciências Biomédicas, Universidade de São Paulo; São Paulo, SP, Brazil
| | | | - Fernanda Neves Estrela
- Programa de Pós-Graduação Em Conservação de Recursos Naturais do Cerrado, Instituto Federal Goiano; Urutaí, GO, Brazil
| | - Marilia Freitas Calmon
- Instituto de Biociências, Letras e Ciências Exatas, Universidade Estadual Paulista "Júlio de Mesquita Filho"; São José do Rio Preto, SP, Brazil
| | - Rafael Nava Miceli
- SeMAE - Serviço Municipal Autonômo de Água e Esgoto, São José do Rio Preto; São Paulo, SP, Brazil
| | - Paulo R S Sanches
- Instituto de Química, Universidade Estadual Paulista; Araraquara, SP, Brazil
| | - Cíntia Bittar
- Instituto de Biociências, Letras e Ciências Exatas, Universidade Estadual Paulista "Júlio de Mesquita Filho"; São José do Rio Preto, SP, Brazil
| | - Paula Rahal
- Instituto de Biociências, Letras e Ciências Exatas, Universidade Estadual Paulista "Júlio de Mesquita Filho"; São José do Rio Preto, SP, Brazil
| | - Eduardo M Cilli
- Instituto de Química, Universidade Estadual Paulista; Araraquara, SP, Brazil
| | | | - Christoph F A Vogel
- Department of Environmental Toxicology and Center for Health and the Environment, University of California, Davis, CA, 95616, USA
| | - Guilherme Malafaia
- Programa de Pós-Graduação Em Conservação de Recursos Naturais do Cerrado, Instituto Federal Goiano; Urutaí, GO, Brazil; Programa de Pós-Graduação Em Biotecnologia e Biodiversidade, Universidade Federal de Goiás, Goiânia, GO, Brazil; Programa de Pós-Graduação Em Ecologia e Conservação de Recursos Naturais, Universidade Federal de Uberlância, Uberlândia, MG, Brazil.
| |
Collapse
|
6
|
Miloradovic D, Pavlovic D, Jankovic MG, Nikolic S, Papic M, Milivojevic N, Stojkovic M, Ljujic B. Human Embryos, Induced Pluripotent Stem Cells, and Organoids: Models to Assess the Effects of Environmental Plastic Pollution. Front Cell Dev Biol 2021; 9:709183. [PMID: 34540831 PMCID: PMC8446652 DOI: 10.3389/fcell.2021.709183] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/19/2021] [Indexed: 02/03/2023] Open
Abstract
For a long time, animal models were used to mimic human biology and diseases. However, animal models are not an ideal solution due to numerous interspecies differences between humans and animals. New technologies, such as human-induced pluripotent stem cells and three-dimensional (3D) cultures such as organoids, represent promising solutions for replacing, refining, and reducing animal models. The capacity of organoids to differentiate, self-organize, and form specific, complex, biologically suitable structures makes them excellent in vitro models of development and disease pathogenesis, as well as drug-screening platforms. Despite significant potential health advantages, further studies and considerable nuances are necessary before their clinical use. This article summarizes the definition of embryoids, gastruloids, and organoids and clarifies their appliance as models for early development, diseases, environmental pollution, drug screening, and bioinformatics.
Collapse
Affiliation(s)
- Dragana Miloradovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragica Pavlovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Marina Gazdic Jankovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Sandra Nikolic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Milos Papic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nevena Milivojevic
- Laboratory for Bioengineering, Department of Science, Institute for Information Technologies, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag Stojkovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- SPEBO Medical Fertility Hospital, Leskovac, Serbia
| | - Biljana Ljujic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
7
|
Amjad W, Haider R, Malik A, Qureshi W. Insights into the management of anorectal disease in the coronavirus 2019 disease era. Therap Adv Gastroenterol 2021; 14:17562848211028117. [PMID: 34290826 PMCID: PMC8274100 DOI: 10.1177/17562848211028117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/08/2021] [Indexed: 02/04/2023] Open
Abstract
Coronavirus 2019 disease (COVID-19) has created major impacts on public health. The virus has plagued a large population requiring hospitalization and resource utilization. Knowledge about the COVID-19 virus continues to grow. It can commonly present with gastrointestinal symptoms; initially, this was considered an atypical presentation, which led to delays in care. The pandemic has posed serious threats to the care of anorectal diseases. Urgent surgeries have been delayed, and the care of cancer patients and cancer screenings disrupted. This had added to patient discomfort and the adverse outcomes on healthcare will continue into the future. The better availability of personal protective equipment to providers and standard checklist protocols in operating rooms can help minimize healthcare-related spread of the virus. Telehealth, outpatient procedures, and biochemical tumor marker tests can help with mitigation of anorectal-disease-related problems. There is limited literature about the clinical management of anorectal diseases during the pandemic. We performed a detailed literature review to guide clinicians around management options for anorectal disease patients. We also highlighted the health challenges seen during the pandemic.
Collapse
Affiliation(s)
- Waseem Amjad
- Internal Medicine, Albany Medical Center, Albany, NY, USA
| | - Rabbia Haider
- Internal Medicine, Nishter Medical University, Multan, Punjab, Pakistan
| | - Adnan Malik
- Internal Medicine, Loyola University School of Medicine, Chicago, IL, USA
| | - Waqas Qureshi
- Section of Cardiology in Division of Internal Medicine, University of Massachusetts School of Medicine, Worcester, MA 01655, USA
| |
Collapse
|
8
|
Hysenaj L, Little S, Kulhanek K, Gbenedio OM, Rodriguez L, Shen A, Lone JC, Lupin-Jimenez LC, Bonser LR, Serwas NK, Bahl K, Mick E, Li JZ, Ding VW, Matsumoto S, Maishan M, Simoneau C, Fragiadakis G, Jablons DM, Langelier CR, Matthay M, Ott M, Krummel M, Combes AJ, Sil A, Erle DJ, Kratz JR, Roose JP. SARS-CoV-2 infection studies in lung organoids identify TSPAN8 as novel mediator. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.06.01.446640. [PMID: 34100012 PMCID: PMC8183007 DOI: 10.1101/2021.06.01.446640] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SARS coronavirus-2 (SARS-CoV-2) is causing a global pandemic with large variation in COVID-19 disease spectrum. SARS-CoV-2 infection requires host receptor ACE2 on lung epithelium, but epithelial underpinnings of variation are largely unknown. We capitalized on comprehensive organoid assays to report remarkable variation in SARS-CoV-2 infection rates of lung organoids from different subjects. Tropism is highest for TUBA- and MUC5AC-positive organoid cells, but levels of TUBA-, MUC5A-, or ACE2- positive cells do not predict infection rate. We identify surface molecule Tetraspanin 8 (TSPAN8) as novel mediator of SARS-CoV-2 infection, which is not downregulated by this specific virus. TSPAN8 levels, prior to infection, strongly correlate with infection rate and TSPAN8-blocking antibodies diminish SARS-CoV-2 infection. We propose TSPAN8 as novel functional biomarker and potential therapeutic target for COVID-19.
Collapse
Affiliation(s)
- Lisiena Hysenaj
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Samantha Little
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kayla Kulhanek
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Oghenekevwe M. Gbenedio
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Lauren Rodriguez
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
| | - Alan Shen
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
| | - Jean-Christophe Lone
- School of Life Science, University of Essex, Wivenhoe Park,Colchester C04 3SQ, United Kingdom
| | | | - Luke R. Bonser
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Nina K. Serwas
- Department of Pathology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Kriti Bahl
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| | - Eran Mick
- Division of Infectious Diseases, University of California, San Francisco, San Francisco, California 94143, USA and Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, California 94143, USA
| | - Jack Z. Li
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California 94143, USA
| | - Vivianne W. Ding
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California 94143, USA
| | - Shotaro Matsumoto
- Gladstone Institute of Virology, Department of Medicine, University of California San Francisco, California 94143, USA
| | - Mazharul Maishan
- Gladstone Institute of Virology, Department of Medicine, University of California San Francisco, California 94143, USA
| | - Camille Simoneau
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Gabriela Fragiadakis
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
| | - David M. Jablons
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California 94143, USA
| | - Charles R. Langelier
- Division of Infectious Diseases, University of California, San Francisco, San Francisco, California 94143, USA and Department of Surgery, Division of Cardiothoracic Surgery, University of California, San Francisco, San Francisco, California 94143, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Michael Matthay
- Gladstone Institute of Virology, Department of Medicine, University of California San Francisco, California 94143, USA
| | - Melanie Ott
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Matthew Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Alexis J. Combes
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
| | - Anita Sil
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California 94143, USA
| | - David J. Erle
- UCSF CoLabs, University of California San Francisco, San Francisco, California 94143, USA
- Lung Biology Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, USA
| | - Johannes R. Kratz
- Cardiovascular Research Institute, Departments of Medicine and Anesthesia, University of California, San Francisco, San Francisco, California 94143, USA
| | - Jeroen P. Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
9
|
Akkiz H. Implications of the Novel Mutations in the SARS-CoV-2 Genome for Transmission, Disease Severity, and the Vaccine Development. Front Med (Lausanne) 2021; 8:636532. [PMID: 34026780 PMCID: PMC8137987 DOI: 10.3389/fmed.2021.636532] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/25/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative virus of the coronavirus disease 2019 (COVID-19), has been identified in China in late December 2019. SARS-CoV-2 is an enveloped, positive-sense, single-stranded RNA betacoronavirus of the Coronaviridae family. Coronaviruses have genetic proofreading mechanism that corrects copying mistakes and thus SARS-CoV-2 genetic diversity is extremely low. Despite lower mutation rate of the virus, researchers have detected a total of 12,706 mutations in the SARS-CoV-2 genome, the majority of which were single nucleotide polymorphisms. Sequencing data revealed that the SARS-CoV-2 accumulates two-single nucleotide mutations per month in its genome. Recently, an amino acid aspartate (D) to glycine (G) (D614G) mutation due to an adenine to guanine nucleotide change at position 23,403 at the 614th amino-acid position of the spike protein in the original reference genotype has been identified. The SARS-CoV-2 viruses that carry the spike protein D614G mutation have become dominant variant around the world. The D614G mutation has been found to be associated with 3 other mutations in the spike protein. Clinical and pseudovirus experimental studies have demonstrated that the spike protein D614G mutation alters the virus phenotype. However, the impact of the mutation on the rate of transmission between people, disease severity and the vaccine and therapeutic development remains unclear. Three variants of SARS-CoV-2 have recently been identified. They are B.1.1.7 (UK) variant, B.1.351 (N501Y.V2, South African) variant and B.1.1.28 (Brazilian) variant. Epidemiological data suggest that they have a higher transmissibility than the original variant. There are reports that some vaccines are less efficacious against the B.1.351 variant. This review article discusses the effects of novel mutations in the SARS-CoV-2 genome on transmission, clinical outcomes and vaccine development.
Collapse
Affiliation(s)
- Hikmet Akkiz
- Department of Gastroenterology and Hepatology, The University of Çukurova, Adana, Turkey
| |
Collapse
|
10
|
Ertas YN, Mahmoodi M, Shahabipour F, Jahed V, Diltemiz SE, Tutar R, Ashammakhi N. Role of biomaterials in the diagnosis, prevention, treatment, and study of corona virus disease 2019 (COVID-19). EMERGENT MATERIALS 2021; 4:35-55. [PMID: 33748672 PMCID: PMC7962632 DOI: 10.1007/s42247-021-00165-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/12/2021] [Indexed: 05/02/2023]
Abstract
Recently emerged novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the resulting corona virus disease 2019 (COVID-19) led to urgent search for methods to prevent and treat COVID-19. Among important disciplines that were mobilized is the biomaterials science and engineering. Biomaterials offer a range of possibilities to develop disease models, protective, diagnostic, therapeutic, monitoring measures, and vaccines. Among the most important contributions made so far from this field are tissue engineering, organoids, and organ-on-a-chip systems, which have been the important frontiers in developing tissue models for viral infection studies. Also, due to low bioavailability and limited circulation time of conventional antiviral drugs, controlled and targeted drug delivery could be applied alternatively. Fortunately, at the time of writing this paper, we have two successful vaccines and new at-home detection platforms. In this paper, we aim to review recent advances of biomaterial-based platforms for protection, diagnosis, vaccination, therapeutics, and monitoring of SARS-CoV-2 and discuss challenges and possible future research directions in this field.
Collapse
Affiliation(s)
- Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Mahboobeh Mahmoodi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, CA USA
- Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Fahimeh Shahabipour
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
- Skin Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Vahid Jahed
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | | | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Avcilar, Istanbul, Turkey
| | - Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, CA USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI USA
| |
Collapse
|
11
|
Organoid As a Novel Technology for Disease Modeling. JOURNAL OF BASIC AND CLINICAL HEALTH SCIENCES 2021. [DOI: 10.30621/jbachs.868837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
12
|
Müller JA, Groß R, Conzelmann C, Krüger J, Merle U, Steinhart J, Weil T, Koepke L, Bozzo CP, Read C, Fois G, Eiseler T, Gehrmann J, van Vuuren J, Wessbecher IM, Frick M, Costa IG, Breunig M, Grüner B, Peters L, Schuster M, Liebau S, Seufferlein T, Stenger S, Stenzinger A, MacDonald PE, Kirchhoff F, Sparrer KMJ, Walther P, Lickert H, Barth TFE, Wagner M, Münch J, Heller S, Kleger A. SARS-CoV-2 infects and replicates in cells of the human endocrine and exocrine pancreas. Nat Metab 2021; 3:149-165. [PMID: 33536639 DOI: 10.1038/s42255-021-00347-1] [Citation(s) in RCA: 337] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
Abstract
Infection-related diabetes can arise as a result of virus-associated β-cell destruction. Clinical data suggest that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing the coronavirus disease 2019 (COVID-19), impairs glucose homoeostasis, but experimental evidence that SARS-CoV-2 can infect pancreatic tissue has been lacking. In the present study, we show that SARS-CoV-2 infects cells of the human exocrine and endocrine pancreas ex vivo and in vivo. We demonstrate that human β-cells express viral entry proteins, and SARS-CoV-2 infects and replicates in cultured human islets. Infection is associated with morphological, transcriptional and functional changes, including reduced numbers of insulin-secretory granules in β-cells and impaired glucose-stimulated insulin secretion. In COVID-19 full-body postmortem examinations, we detected SARS-CoV-2 nucleocapsid protein in pancreatic exocrine cells, and in cells that stain positive for the β-cell marker NKX6.1 and are in close proximity to the islets of Langerhans in all four patients investigated. Our data identify the human pancreas as a target of SARS-CoV-2 infection and suggest that β-cell infection could contribute to the metabolic dysregulation observed in patients with COVID-19.
Collapse
Affiliation(s)
- Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Carina Conzelmann
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jana Krüger
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
| | - Uta Merle
- Department of Internal Medicine 4, University of Heidelberg, Heidelberg, Germany
| | | | - Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Lennart Koepke
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Clarissa Read
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| | - Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
| | - Julia Gehrmann
- Institute for Computational Genomics, RWTH Aachen University, Aachen, Germany
| | - Joanne van Vuuren
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Isabel M Wessbecher
- Tissue Bank of the German Center for Infection Research, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen University, Aachen, Germany
| | - Markus Breunig
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
| | - Beate Grüner
- Department of Internal Medicine 3, Ulm University Hospital, Ulm, Germany
| | - Lynn Peters
- Department of Internal Medicine 3, Ulm University Hospital, Ulm, Germany
| | - Michael Schuster
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
| | - Steffen Stenger
- Institute for Microbiology and Hygiene, Ulm University Medical Center, Ulm, Germany
| | | | - Patrick E MacDonald
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | | | - Martin Wagner
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany.
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Sandra Heller
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany.
| | - Alexander Kleger
- Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany.
| |
Collapse
|
13
|
Srivastava M, Srivastava N, Mishra PK, Malhotra BD. Prospects of nanomaterials-enabled biosensors for COVID-19 detection. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 754:142363. [PMID: 33254928 PMCID: PMC7492839 DOI: 10.1016/j.scitotenv.2020.142363] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/11/2020] [Accepted: 09/11/2020] [Indexed: 05/03/2023]
Abstract
We are currently facing the COVID-19 pandemic which is the consequence of severe acute respiratory syndrome coronavirus (SARS-CoV-2). Since no specific vaccines or drugs have been developed till date for the treatment of SARS-CoV-2 infection, early diagnosis is essential to further combat this pandemic. In this context, the reliable, rapid, and low-cost technique for SARS-CoV-2 diagnosis is the foremost priority. At present reverse transcription polymerase chain reaction (RT-PCR) is the reference technique presently being used for the detection of SARS-CoV-2 infection. However, in a number of cases, false results have been noticed in COVID-19 diagnosis. To develop advanced techniques, researchers are continuously working and in the series of constant efforts, nanomaterials-enabled biosensing approaches can be a hope to offer novel techniques that may perhaps meet the current demand of fast and early diagnosis of COVID-19 cases. This paper provides an overview of the COVID-19 pandemic and nanomaterials-enabled biosensing approaches that have been recently reported for the diagnosis of SARS-CoV-2. Though limited studies on the development of nanomaterials enabled biosensing techniques for the diagnosis of SARS-CoV-2 have been reported, this review summarizes nanomaterials mediated improved biosensing strategies and the possible mechanisms that may be responsible for the diagnosis of the COVID-19 disease. It is reviewed that nanomaterials e.g. gold nanostructures, lanthanide-doped polysterene nanoparticles (NPs), graphene and iron oxide NPs can be potentially used to develop advanced techniques offered by colorimetric, amperometric, impedimetric, fluorescence, and optomagnetic based biosensing of SARS-CoV-2. Finally, critical issues that are likely to accelerate the development of nanomaterials-enabled biosensing for SARS-CoV-2 infection have been discussed in detail. This review may serve as a guide for the development of advanced techniques for nanomaterials enabled biosensing to fulfill the present demand of low-cost, rapid and early diagnosis of COVID-19 infection.
Collapse
Affiliation(s)
- Manish Srivastava
- Department of Chemical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India.
| | - Neha Srivastava
- Department of Chemical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - P K Mishra
- Department of Chemical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Bansi D Malhotra
- Nano-Bioelectronics Laboratory, Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi 110042, India.
| |
Collapse
|
14
|
Zhang M, Feng C, Zhang X, Hu S, Zhang Y, Min M, Liu B, Ying X, Liu Y. Susceptibility Factors of Stomach for SARS-CoV-2 and Treatment Implication of Mucosal Protective Agent in COVID-19. Front Med (Lausanne) 2021; 7:597967. [PMID: 33521016 PMCID: PMC7840564 DOI: 10.3389/fmed.2020.597967] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/07/2020] [Indexed: 01/14/2023] Open
Abstract
Objectives: This work aims to study the gastrointestinal (GI) symptoms in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients and the susceptibility factors of the stomach for SARS-CoV-2. Materials and Methods: We investigated the SARS-CoV-2 susceptibility by analyzing the expression distribution of viral entry-associated genes, ACE2 and TMPRSS2, in single-cell RNA sequencing data derived from 12 gastric mucosa samples. We also analyzed the epidemiological, demographic, clinical, and laboratory data of 420 cases with SARS-CoV-2-caused coronavirus disease 2019 (COVID-19). Results:ACE2 and TMPRSS2 are specifically expressed in enterocytes which are mainly from gastric mucosa samples with Helicobacter pylori (H. pylori) infection history and intestinal metaplasia (IM). A total of 420 patients were surveyed, of which 62 were with and 358 were without GI symptoms. There is a significant difference in average hospital stay (p < 0.001) and cost (p < 0.001) between the two groups. Among 23 hospitalized patients including seven with upper GI symptoms and 16 with lower GI symptoms, six (85.7%) and five (31.3%) had H. pylori infection history, respectively (p = 0.03). Of 18 hospitalized patients with initial upper GI symptoms, none of the eight patients with mucosal protective agent therapy (e.g., sucralfate suspension gel, hydrotalcite tablets) had diarrhea subsequently, whereas six out of 10 patients without mucosal protective agent therapy had diarrhea subsequently (p = 0.01). Conclusion: IM and H. pylori infection history may be susceptibility factors of SARS-CoV-2, and the mucosal protective agent may be useful for the blockade of SARS-CoV-2 transmission from the stomach to the intestine.
Collapse
Affiliation(s)
- Min Zhang
- State Key Laboratory of Kidney Diseases, Department of Nephrology, The First Medical Center, National Clinical Research Center for Kidney Diseases, Chinese PLA Institute of Nephrology, Chinese PLA General Hospital, Beijing, China
| | - Chao Feng
- Center for Computational Biology, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Xingchen Zhang
- Department of Pharmacy, The Central Hospital of Wuhan Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shuofeng Hu
- Center for Computational Biology, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Yuan Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Min
- The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Bing Liu
- The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaomin Ying
- Center for Computational Biology, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Yan Liu
- The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Mirzaei R, Attar A, Papizadeh S, Jeda AS, Hosseini-Fard SR, Jamasbi E, Kazemi S, Amerkani S, Talei GR, Moradi P, Jalalifar S, Yousefimashouf R, Hossain MA, Keyvani H, Karampoor S. The emerging role of probiotics as a mitigation strategy against coronavirus disease 2019 (COVID-19). Arch Virol 2021; 166:1819-1840. [PMID: 33745067 PMCID: PMC7980799 DOI: 10.1007/s00705-021-05036-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
COVID-19 is an acute respiratory infection accompanied by pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has affected millions of people globally. To date, there are no highly efficient therapies for this infection. Probiotic bacteria can interact with the gut microbiome to strengthen the immune system, enhance immune responses, and induce appropriate immune signaling pathways. Several probiotics have been confirmed to reduce the duration of bacterial or viral infections. Immune fitness may be one of the approaches by which protection against viral infections can be reinforced. In general, prevention is more efficient than therapy in fighting viral infections. Thus, probiotics have emerged as suitable candidates for controlling these infections. During the COVID-19 pandemic, any approach with the capacity to induce mucosal and systemic reactions could potentially be useful. Here, we summarize findings regarding the effectiveness of various probiotics for preventing virus-induced respiratory infectious diseases, especially those that could be employed for COVID-19 patients. However, the benefits of probiotics are strain-specific, and it is necessary to identify the bacterial strains that are scientifically established to be beneficial.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Adeleh Attar
- Department of Microbiology and Virology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saher Papizadeh
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Salimi Jeda
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elaheh Jamasbi
- Department of Anatomical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saman Amerkani
- Department of Microbiology and Virology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholam Reza Talei
- Department of Virology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Lorestan, Iran
| | - Pouya Moradi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saba Jalalifar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Akhter Hossain
- The Florey University of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Hossein Keyvani
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Peng X, Ouyang J, Isnard S, Lin J, Fombuena B, Zhu B, Routy JP. Sharing CD4+ T Cell Loss: When COVID-19 and HIV Collide on Immune System. Front Immunol 2020; 11:596631. [PMID: 33384690 PMCID: PMC7770166 DOI: 10.3389/fimmu.2020.596631] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is a distinctive infection characterized by elevated inter-human transmission and presenting from absence of symptoms to severe cytokine storm that can lead to dismal prognosis. Like for HIV, lymphopenia and drastic reduction of CD4+ T cell counts in COVID-19 patients have been linked with poor clinical outcome. As CD4+ T cells play a critical role in orchestrating responses against viral infections, important lessons can be drawn by comparing T cell response in COVID-19 and in HIV infection and by studying HIV-infected patients who became infected by SARS-CoV-2. We critically reviewed host characteristics and hyper-inflammatory response in these two viral infections to have a better insight on the large difference in clinical outcome in persons being infected by SARS-CoV-2. The better understanding of mechanism of T cell dysfunction will contribute to the development of targeted therapy against severe COVID-19 and will help to rationally design vaccine involving T cell response for the long-term control of viral infection.
Collapse
Affiliation(s)
- Xiaorong Peng
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada.,Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada.,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Ouyang
- Chongqing Public Health Medical Center, Chongqing, China
| | - Stéphane Isnard
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada.,Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada.,CIHR Canadian HIV Trials Network, Vancouver, BC, Canada
| | - John Lin
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada.,Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada
| | - Brandon Fombuena
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada.,Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada
| | - Biao Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jean-Pierre Routy
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada.,Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada.,Division of Hematology, McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
17
|
Spalinger MR, Hai R, Li J, Santos AN, Nordgren TM, Tremblay ML, Eckmann L, Hanson E, Scharl M, Wu X, Boland BS, McCole DF. Identification of a Novel Susceptibility Marker for SARS-CoV-2 Infection in Human Subjects and Risk Mitigation with a Clinically Approved JAK Inhibitor in Human/Mouse Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.12.09.416586. [PMID: 33330862 PMCID: PMC7743066 DOI: 10.1101/2020.12.09.416586] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronavirus disease (COVID-19), caused by SARS-CoV-2, has affected over 65 million individuals and killed over 1.5 million persons (December 8, 2020; www.who.int)1. While fatality rates are higher among the elderly and those with underlying comorbidities2, host factors that promote susceptibility to SARS-CoV-2 infection and severe disease are poorly understood. Although individuals with certain autoimmune/inflammatory disorders show increased susceptibility to viral infections, there is incomplete knowledge of SARS-CoV-2 susceptibility in these diseases.3-7 We report that the autoimmune PTPN2 risk variant rs1893217 promotes expression of the SARS-CoV-2 receptor, ACE2, and increases cellular entry mediated by SARS-CoV-2 spike protein. Elevated ACE2 expression and viral entry were mediated by increased JAK-STAT signalling, and were reversed by the JAK inhibitor, tofacitinib. Collectively, our findings uncover a novel risk biomarker for increased expression of the SARS-CoV-2 receptor and viral entry, and identify a clinically approved therapeutic agent to mitigate this risk.
Collapse
Affiliation(s)
- Marianne R. Spalinger
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
- Department of Gastroenterology and Hepatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Rong Hai
- Department of Microbiology & Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Alina N. Santos
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Tara M. Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Michel L. Tremblay
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Lars Eckmann
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Elaine Hanson
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of City of Hope, Monrovia, California, USA
| | - Brigid S. Boland
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Declan F. McCole
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, USA
| |
Collapse
|
18
|
Kumar M, Patel AK, Shah AV, Raval J, Rajpara N, Joshi M, Joshi CG. First proof of the capability of wastewater surveillance for COVID-19 in India through detection of genetic material of SARS-CoV-2. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020. [PMID: 32768790 DOI: 10.1101/2020.06.16.20133215] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
We made the first ever successful effort in India to detect the genetic material of SARS-CoV-2 viruses to understand the capability and application of wastewater-based epidemiology (WBE) surveillance in India. Sampling was carried out on 8 and 27 May 2020 at the Old Pirana Waste Water Treatment Plant (WWTP) at Ahmedabad, Gujarat that receives effluent from Civil Hospital treating COVID-19 patients. All three, i.e. ORF1ab, N and S genes of SARS-CoV-2, were found in the influent with no genes detected in effluent collected on 8 and 27 May 2020. Increase in SARS-CoV-2 genetic loading in the wastewater between 8 and 27 May 2020 samples concurred with corresponding increase in the number of active COVID-19 patients in the city. The number of gene copies was comparable to that reported in untreated wastewaters of Australia, China and Turkey and lower than that of the USA, France and Spain. However, temporal changes in SARS-CoV-2 RNA concentrations need to be substantiated further from the perspectives of daily and short-term changes of SARS-CoV-2 in wastewater through long-term monitoring. The study results SARS-CoV-2 will assist concerned authorities and policymakers to formulate and/or upgrade COVID-19 surveillance to have a more explicit picture of the pandemic curve. While infectivity of SARS-CoV-2 through the excreted viral genetic material in the aquatic environment is still being debated, the presence and detection of genes in wastewater systems makes a strong case for the environmental surveillance of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Manish Kumar
- Discipline of Earth Science, Indian Institute of Technology Gandhinagar, Gujarat 382 355, India; Kiran C Patel Centre for Sustainable Development, Indian Institute of Technology Gandhinagar, Gujarat, India.
| | - Arbind Kumar Patel
- Discipline of Earth Science, Indian Institute of Technology Gandhinagar, Gujarat 382 355, India
| | - Anil V Shah
- Gujarat Pollution Control Board (GPCB), Paryavaran Bhavan, Gandhinagar, Gujarat 382 010, India
| | - Janvi Raval
- Gujarat Biotechnology Research Centre (GBRC), Sector- 11, Gandhinagar, Gujarat 382 011, India
| | - Neha Rajpara
- Gujarat Biotechnology Research Centre (GBRC), Sector- 11, Gandhinagar, Gujarat 382 011, India
| | - Madhvi Joshi
- Gujarat Biotechnology Research Centre (GBRC), Sector- 11, Gandhinagar, Gujarat 382 011, India
| | - Chaitanya G Joshi
- Gujarat Biotechnology Research Centre (GBRC), Sector- 11, Gandhinagar, Gujarat 382 011, India
| |
Collapse
|
19
|
Livanos AE, Jha D, Cossarini F, Gonzalez-Reiche AS, Tokuyama M, Aydillo T, Parigi TL, Ramos I, Dunleavy K, Lee B, Dixon R, Chen ST, Martinez-Delgado G, Nagula S, Ko HM, Glicksberg BS, Nadkarni G, Pujadas E, Reidy J, Naymagon S, Grinspan A, Ahmad J, Tankelevich M, Gordon R, Sharma K, Houldsworth J, Britton GJ, Chen-Liaw A, Spindler MP, Plitt T, Wang P, Cerutti A, Faith JJ, Colombel JF, Kenigsberg E, Argmann C, Merad M, Gnjatic S, Harpaz N, Danese S, Cordon-Cardo C, Rahman A, Kumta NA, Aghemo A, Petralia F, van Bakel H, Garcia-Sastre A, Mehandru S. Gastrointestinal involvement attenuates COVID-19 severity and mortality. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.09.07.20187666. [PMID: 32935117 PMCID: PMC7491532 DOI: 10.1101/2020.09.07.20187666] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Given that gastrointestinal (GI) symptoms are a prominent extrapulmonary manifestation of coronavirus disease 2019 (COVID-19), we investigated intestinal infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its effect on disease pathogenesis. SARS-CoV-2 was detected in small intestinal enterocytes by immunofluorescence staining or electron microscopy, in 13 of 15 patients studied. High dimensional analyses of GI tissues revealed low levels of inflammation in general, including active downregulation of key inflammatory genes such as IFNG, CXCL8, CXCL2 and IL1B and reduced frequencies of proinflammatory dendritic cell subsets. To evaluate the clinical significance of these findings, examination of two large, independent cohorts of hospitalized patients in the United States and Europe revealed a significant reduction in disease severity and mortality that was independent of gender, age, and examined co-morbid illnesses. The observed mortality reduction in COVID-19 patients with GI symptoms was associated with reduced levels of key inflammatory proteins including IL-6, CXCL8, IL-17A and CCL28 in circulation but was not associated with significant differences in nasopharyngeal viral loads. These data draw attention to organ-level heterogeneity in disease pathogenesis and highlight the role of the GI tract in attenuating SARS-CoV-2-associated inflammation with related mortality benefit. ONE SENTENCE SUMMARY Intestinal infection with SARS-CoV-2 is associated with a mild inflammatory response and improved clinical outcomes.
Collapse
|
20
|
Uzzan M, Corcos O, Martin JC, Treton X, Bouhnik Y. Why is SARS-CoV-2 infection more severe in obese men? The gut lymphatics - Lung axis hypothesis. Med Hypotheses 2020; 144:110023. [PMID: 32593832 PMCID: PMC7308746 DOI: 10.1016/j.mehy.2020.110023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023]
Abstract
Consistent observations report increased severity of SARS-CoV-2 infection in overweight men with cardiovascular factors. As the visceral fat possesses an intense immune activity, is involved in metabolic syndrome and is at the crossroad between the intestines, the systemic circulation and the lung, we hypothesized that it plays a major role in severe forms of SARS-CoV-2 infection. SARS-CoV2 presents the ability to infect epithelial cells of the respiratory tract as well as the intestinal tract. Several factors may increase intestinal permeability including direct enterocyte damage by SARS-CoV2, systemic inflammatory response syndrome (SIRS) and epithelial ischemia secondary to SARS-CoV2- associated endothelial dysfunction. This increase permeability further leads to translocation of microbial components such as MAMPs (microbial-associated molecular pattern), triggering an inflammatory immune response by TLR-expressing cells of the mesentery fat (mostly macrophages and adipocytes). The pro-inflammatory cytokines produced by the mesentery fat mediates systemic inflammation and aggravate acute respiratory distress syndrome (ARDS) through the mesenteric lymph drainage.
Collapse
Affiliation(s)
- Mathieu Uzzan
- Department of Gastroenterology, IBD and Nutritional Support, CHU Paris Nord-Val de Seine, Beaujon Hospital, Clichy, France.
| | - Olivier Corcos
- Department of Gastroenterology, IBD and Nutritional Support, CHU Paris Nord-Val de Seine, Beaujon Hospital, Clichy, France
| | - Jerome C Martin
- Université de Nantes, Inserm, CHU Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, F-44000 Nantes, France; CHU Nantes, Laboratoire d'Immunologie, Center for Immuno Monitoring Nantes-Atlantique (CIMNA), F-44000 Nantes, France
| | - Xavier Treton
- Department of Gastroenterology, IBD and Nutritional Support, CHU Paris Nord-Val de Seine, Beaujon Hospital, Clichy, France
| | - Yoram Bouhnik
- Department of Gastroenterology, IBD and Nutritional Support, CHU Paris Nord-Val de Seine, Beaujon Hospital, Clichy, France
| |
Collapse
|
21
|
Thompson JR, Nancharaiah YV, Gu X, Lee WL, Rajal VB, Haines MB, Girones R, Ng LC, Alm EJ, Wuertz S. Making waves: Wastewater surveillance of SARS-CoV-2 for population-based health management. WATER RESEARCH 2020; 184:116181. [PMID: 32707307 PMCID: PMC7357518 DOI: 10.1016/j.watres.2020.116181] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 05/18/2023]
Abstract
Worldwide, clinical data remain the gold standard for disease surveillance and tracking. However, such data are limited due to factors such as reporting bias and inability to track asymptomatic disease carriers. Disease agents are excreted in the urine and feces of infected individuals regardless of disease symptom severity. Wastewater surveillance - that is, monitoring disease via human effluent - represents a valuable complement to clinical approaches. Because wastewater is relatively inexpensive and easy to collect and can be monitored at different levels of population aggregation as needed, wastewater surveillance can offer a real-time, cost-effective view of a community's health that is independent of biases associated with case-reporting. For SARS-CoV-2 and other disease-causing agents we envision an aggregate wastewater-monitoring system at the level of a wastewater treatment plant and exploratory or confirmatory monitoring of the sewerage system at the neighborhood scale to identify or confirm clusters of infection or assess impact of control measures where transmission has been established. Implementation will require constructing a framework with collaborating government agencies, public or private utilities, and civil society organizations for appropriate use of data collected from wastewater, identification of an appropriate scale of sample collection and aggregation to balance privacy concerns and risk of stigmatization with public health preservation, and consideration of the social implications of wastewater surveillance.
Collapse
Affiliation(s)
- Janelle R Thompson
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University (NTU), Singapore; Asian School of the Environment, NTU, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore.
| | - Yarlagadda V Nancharaiah
- Biofouling and Biofilm Processes, Water and Steam Chemistry Division, Chemistry Group, Bhabha Atomic Research Centre, Kalpakkam 603102, India; Homi Bhabha National Institute, BARC Training School Complex, Anushakti Nagar, Trombay, Mumbai 400 094, India
| | - Xiaoqiong Gu
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Singapore-MIT Alliance for Research and Technology, National University of Singapore, Singapore
| | - Wei Lin Lee
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Singapore-MIT Alliance for Research and Technology, National University of Singapore, Singapore
| | - Verónica B Rajal
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University (NTU), Singapore; Instituto de Investigaciones para la Industria Química (INIQUI), CONICET, Universidad Nacional de Salta (UNSa), Av. Bolivia 5150, Salta 4400, Argentina; Facultad de Ingeniería, UNSa, Salta, Argentina
| | - Monamie B Haines
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; School of Social Sciences, Sociology Division, NTU, Singapore
| | - Rosina Girones
- Section of Microbiology, Virology and Biotechnology, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Diagonal, 643, 08028-Barcelona, Spain
| | - Lee Ching Ng
- Environmental Health Institute, National Environment Agency, Singapore
| | - Eric J Alm
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; Singapore-MIT Alliance for Research and Technology, National University of Singapore, Singapore; Center for Microbiome Informatics and Therapeutics, Departments of Biological Engineering and Civil and Environmental Engineering, Massachusetts Institute of Technology, United States; Biobot Analytics, Cambridge MA, United States
| | - Stefan Wuertz
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University (NTU), Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore; School of Civil and Environmental Engineering, NTU, Singapore.
| |
Collapse
|
22
|
Gao C, Zeng J, Jia N, Stavenhagen K, Matsumoto Y, Zhang H, Li J, Hume AJ, Mühlberger E, van Die I, Kwan J, Tantisira K, Emili A, Cummings RD. SARS-CoV-2 Spike Protein Interacts with Multiple Innate Immune Receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.29.227462. [PMID: 32766577 PMCID: PMC7402034 DOI: 10.1101/2020.07.29.227462] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The spike (S) glycoprotein in the envelope of SARS-CoV-2 is densely glycosylated but the functions of its glycosylation are unknown. Here we demonstrate that S is recognized in a glycan-dependent manner by multiple innate immune receptors including the mannose receptor MR/CD206, DC-SIGN/CD209, L-SIGN/CD209L, and MGL/CLEC10A/CD301. Single-cell RNA sequencing analyses indicate that such receptors are highly expressed in innate immune cells in tissues susceptible to SARS-CoV-2 infection. Binding of the above receptors to S is characterized by affinities in the picomolar range and consistent with S glycosylation analysis demonstrating a variety of N- and O-glycans as receptor ligands. These results indicate multiple routes for SARS-CoV-2 to interact with human cells and suggest alternative strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Chao Gao
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Junwei Zeng
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nan Jia
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kathrin Stavenhagen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yasuyuki Matsumoto
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hua Zhang
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Jiang Li
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Adam J. Hume
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Elke Mühlberger
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Irma van Die
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Julian Kwan
- Center for Network Systems Biology, Departments of Biochemistry and Biology, Boston University, Boston, MA, 02118 USA
| | - Kelan Tantisira
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Emili
- Center for Network Systems Biology, Departments of Biochemistry and Biology, Boston University, Boston, MA, 02118 USA
| | - Richard D. Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Salahudeen AA, Choi SS, Rustagi A, Zhu J, de la O SM, Flynn RA, Margalef-Català M, Santos AJM, Ju J, Batish A, van Unen V, Usui T, Zheng GXY, Edwards CE, Wagar LE, Luca V, Anchang B, Nagendran M, Nguyen K, Hart DJ, Terry JM, Belgrader P, Ziraldo SB, Mikkelsen TS, Harbury PB, Glenn JS, Garcia KC, Davis MM, Baric RS, Sabatti C, Amieva MR, Blish CA, Desai TJ, Kuo CJ. Progenitor identification and SARS-CoV-2 infection in long-term human distal lung organoid cultures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.27.212076. [PMID: 32743583 PMCID: PMC7386503 DOI: 10.1101/2020.07.27.212076] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The distal lung contains terminal bronchioles and alveoli that facilitate gas exchange and is affected by disorders including interstitial lung disease, cancer, and SARS-CoV-2-associated COVID-19 pneumonia. Investigations of these localized pathologies have been hindered by a lack of 3D in vitro human distal lung culture systems. Further, human distal lung stem cell identification has been impaired by quiescence, anatomic divergence from mouse and lack of lineage tracing and clonogenic culture. Here, we developed robust feeder-free, chemically-defined culture of distal human lung progenitors as organoids derived clonally from single adult human alveolar epithelial type II (AT2) or KRT5 + basal cells. AT2 organoids exhibited AT1 transdifferentiation potential, while basal cell organoids progressively developed lumens lined by differentiated club and ciliated cells. Organoids consisting solely of club cells were not observed. Upon single cell RNA-sequencing (scRNA-seq), alveolar organoids were composed of proliferative AT2 cells; however, basal organoid KRT5 + cells contained a distinct ITGA6 + ITGB4 + mitotic population whose proliferation segregated to a TNFRSF12A hi subfraction. Clonogenic organoid growth was markedly enriched within the TNFRSF12A hi subset of FACS-purified ITGA6 + ITGB4 + basal cells from human lung or derivative organoids. In vivo, TNFRSF12A + cells comprised ~10% of KRT5 + basal cells and resided in clusters within terminal bronchioles. To model COVID-19 distal lung disease, we everted the polarity of basal and alveolar organoids to rapidly relocate differentiated club and ciliated cells from the organoid lumen to the exterior surface, thus displaying the SARS-CoV-2 receptor ACE2 on the outwardly-facing apical aspect. Accordingly, basal and AT2 apical-out organoids were infected by SARS-CoV-2, identifying club cells as a novel target population. This long-term, feeder-free organoid culture of human distal lung alveolar and basal stem cells, coupled with single cell analysis, identifies unsuspected basal cell functional heterogeneity and exemplifies progenitor identification within a slowly proliferating human tissue. Further, our studies establish a facile in vitro organoid model for human distal lung infectious diseases including COVID-19-associated pneumonia.
Collapse
|
24
|
Mulay A, Konda B, Garcia G, Yao C, Beil S, Sen C, Purkayastha A, Kolls JK, Pociask DA, Pessina P, de Aja JS, Garcia-de-Alba C, Kim CF, Gomperts B, Arumugaswami V, Stripp B. SARS-CoV-2 infection of primary human lung epithelium for COVID-19 modeling and drug discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.06.29.174623. [PMID: 32637946 PMCID: PMC7337376 DOI: 10.1101/2020.06.29.174623] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is the latest respiratory pandemic resulting from zoonotic transmission of severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2). Severe symptoms include viral pneumonia secondary to infection and inflammation of the lower respiratory tract, in some cases causing death. We developed primary human lung epithelial infection models to understand responses of proximal and distal lung epithelium to SARS-CoV-2 infection. Differentiated air-liquid interface cultures of proximal airway epithelium and 3D organoid cultures of alveolar epithelium were readily infected by SARS-CoV-2 leading to an epithelial cell-autonomous proinflammatory response. We validated the efficacy of selected candidate COVID-19 drugs confirming that Remdesivir strongly suppressed viral infection/replication. We provide a relevant platform for studying COVID-19 pathobiology and for rapid drug screening against SARS-CoV-2 and future emergent respiratory pathogens. ONE SENTENCE SUMMARY A novel infection model of the adult human lung epithelium serves as a platform for COVID-19 studies and drug discovery.
Collapse
Affiliation(s)
- A. Mulay
- Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - B. Konda
- Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - G. Garcia
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - C. Yao
- Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - S. Beil
- Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - C. Sen
- UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - A. Purkayastha
- UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - J. K. Kolls
- Tulane School of Medicine, New Orleans, 70112
| | | | - P. Pessina
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary & Respiratory Diseases, Boston Children’s Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - J. Sainz de Aja
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary & Respiratory Diseases, Boston Children’s Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - C. Garcia-de-Alba
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary & Respiratory Diseases, Boston Children’s Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - C. F. Kim
- Stem Cell Program and Divisions of Hematology/Oncology and Pulmonary & Respiratory Diseases, Boston Children’s Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - B. Gomperts
- UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital UCLA, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, 90095, USA
- li and Edythe Broad, Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA
| | - V. Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- li and Edythe Broad, Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA
| | - B.R. Stripp
- Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
25
|
Rees CJ, East JE, Oppong K, Veitch A, McAlindon M, Anderson J, Hayee B, Edwards C, McKinlay A, Penman I. Restarting gastrointestinal endoscopy in the deceleration and early recovery phases of COVID-19 pandemic: Guidance from the British Society of Gastroenterology. Clin Med (Lond) 2020; 20:352-358. [PMID: 32518104 DOI: 10.7861/clinmed.2020-0296] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Many non-emergency clinical services were suspended during COVID-19 pandemic peak. It is essential to develop a plan for restarting services following the peak. It is equally important to protect patients and staff and to use resources and personal protective equipment (PPE) efficiently. The British Society of Gastroenterology Endoscopy Committee and Quality Improvement Programme has produced guidance on how a restart can be safely delivered. Key recommendations include the following: all patients should have need for endoscopy assessed by senior clinicians and prioritised according to criteria we have outlined; once the need for endoscopy is confirmed, patients should undergo telephone screening for symptoms using systematic questionnaires; all outpatients should undergo RT-PCR testing for COVID-19 virus 1-3 days prior to endoscopy; and PPE should be determined by patient risk stratification, the nature of the procedure and the results of testing. While this guidance is tailored to endoscopy services, it could be adapted for any interventional medical discipline.
Collapse
Affiliation(s)
- Colin J Rees
- Newcastle University, Newcastle Upon Tyne, UK and consultant gastroenterologist, South Tyneside and Sunderland NHS Foundation Trust, South Shields, UK
| | - James E East
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Kofi Oppong
- Newcastle upon Tyne Hospitals NHS Foundation Trust
| | - Andrew Veitch
- The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | | | | | - Bu Hayee
- King's College Hospital NHS Foundation Trust, London, UK
| | - Cathryn Edwards
- British Society of Gastroenterology (BSG) and consultant gastroenterologist, Torbay and South Devon NHS Foundation Trust, Torbay, UK
| | - Alastair McKinlay
- BSG and consultant gastroenterologist, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Ian Penman
- BSG and consultant gastroenterologist, Royal Infirmary of Edinburgh, UK
| |
Collapse
|
26
|
Abstract
AIM In addition to respiratory symptoms, COVID-19 can present with gastrointestinal complaints suggesting possible faeco-oral transmission. The primary aim of this review was to establish the incidence and timing of positive faecal samples for SARS-CoV-2 in patients with COVID-19. METHODS A systematic literature review identified studies describing COVID-19 patients tested for faecal virus. Search terms for MEDLINE included 'clinical', 'faeces', 'gastrointestinal secretions', 'stool', 'COVID-19', 'SARS-CoV-2' and '2019-nCoV'. Additional searches were done in the American Journal of Gastroenterology, Gastroenterology, Gut, Lancet Gastroenterology and Hepatology, the World Health Organization Database, the Centre for Evidence-Based Medicine, New England Journal of Medicine, social media and the National Institute for Health and Care Excellence, bioRxiv and medRxiv preprints. Data were extracted concerning the type of test, number and timing of positive samples, incidence of positive faecal tests after negative nasopharyngeal swabs and evidence of viable faecal virus or faeco-oral transmission of the virus. RESULTS Twenty-six relevant articles were identified. Combining study results demonstrated that 53.9% of those tested for faecal RNA were positive. The duration of faecal viral shedding ranged from 1 to 33 days after a negative nasopharyngeal swab with one result remaining positive 47 days after onset of symptoms. There is insufficient evidence to suggest that COVID-19 is transmitted via faecally shed virus. CONCLUSION There is a high rate of positive polymerase chain reaction tests with persistence of SARS-CoV-2 in faecal samples of patients with COVID-19. Further research is needed to confirm if this virus is viable and the degree of transmission through the faeco-oral route. This may have important implications on isolation, recommended precautions and protective equipment for interventional procedures involving the gastrointestinal tract.
Collapse
Affiliation(s)
- S Gupta
- Division of Population Medicine, Cardiff University School of Medicine, Cardiff, UK
| | - J Parker
- Division of Population Medicine, Cardiff University School of Medicine, Cardiff, UK
| | - S Smits
- Division of Population Medicine, Cardiff University School of Medicine, Cardiff, UK
| | - J Underwood
- Division of Infection and Immunity, Department of Infectious Diseases, Cardiff and Vale University Health Board, Cardiff University, Cardiff, UK
| | - S Dolwani
- Division of Population Medicine, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
27
|
Gupta S, Parker J, Smits S, Underwood J, Dolwani S. Persistent viral shedding of SARS-CoV-2 in faeces - a rapid review. Colorectal Dis 2020; 22:611-620. [PMID: 32418307 PMCID: PMC7276890 DOI: 10.1111/codi.15138] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/02/2020] [Accepted: 05/09/2020] [Indexed: 12/16/2022]
Abstract
AIM In addition to respiratory symptoms, COVID-19 can present with gastrointestinal complaints suggesting possible faeco-oral transmission. The primary aim of this review was to establish the incidence and timing of positive faecal samples for SARS-CoV-2 in patients with COVID-19. METHODS A systematic literature review identified studies describing COVID-19 patients tested for faecal virus. Search terms for MEDLINE included 'clinical', 'faeces', 'gastrointestinal secretions', 'stool', 'COVID-19', 'SARS-CoV-2' and '2019-nCoV'. Additional searches were done in the American Journal of Gastroenterology, Gastroenterology, Gut, Lancet Gastroenterology and Hepatology, the World Health Organization Database, the Centre for Evidence-Based Medicine, New England Journal of Medicine, social media and the National Institute for Health and Care Excellence, bioRxiv and medRxiv preprints. Data were extracted concerning the type of test, number and timing of positive samples, incidence of positive faecal tests after negative nasopharyngeal swabs and evidence of viable faecal virus or faeco-oral transmission of the virus. RESULTS Twenty-six relevant articles were identified. Combining study results demonstrated that 53.9% of those tested for faecal RNA were positive. The duration of faecal viral shedding ranged from 1 to 33 days after a negative nasopharyngeal swab with one result remaining positive 47 days after onset of symptoms. There is insufficient evidence to suggest that COVID-19 is transmitted via faecally shed virus. CONCLUSION There is a high rate of positive polymerase chain reaction tests with persistence of SARS-CoV-2 in faecal samples of patients with COVID-19. Further research is needed to confirm if this virus is viable and the degree of transmission through the faeco-oral route. This may have important implications on isolation, recommended precautions and protective equipment for interventional procedures involving the gastrointestinal tract.
Collapse
Affiliation(s)
- S. Gupta
- Division of Population MedicineCardiff University School of MedicineCardiffUK
| | - J. Parker
- Division of Population MedicineCardiff University School of MedicineCardiffUK
| | - S. Smits
- Division of Population MedicineCardiff University School of MedicineCardiffUK
| | - J. Underwood
- Division of Infection and ImmunityDepartment of Infectious DiseasesCardiff and Vale University Health BoardCardiff UniversityCardiffUK
| | - S. Dolwani
- Division of Population MedicineCardiff University School of MedicineCardiffUK
| |
Collapse
|