Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
medrxiv; 2023.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2023.11.27.23298847

ABSTRACT

The orphan gene of SARS-CoV-2, ORF10, is the least studied gene in the virus responsible for the COVID-19 pandemic. Recent experimentation indicated ORF10 expression moderates innate immunity in vitro. However, whether ORF10 affects COVID-19 in humans remained unknown. We determine that the ORF10 sequence is identical to the Wuhan-Hu-1 ancestral haplotype in 95% of genomes across five variants of concern (VOC). Four ORF10 variants are associated with less virulent clinical outcomes in the human host: three of these affect ORF10 protein structure, one affects ORF10 RNA structural dynamics. RNA-Seq data from 2070 samples from diverse human cells and tissues reveals ORF10 accumulation is conditionally discordant from that of other SARS-CoV-2 transcripts. Expression of ORF10 in A549 and HEK293 cells perturbs immune-related gene expression networks, alters expression of the majority of mitochondrially-encoded genes of oxidative respiration, and leads to large shifts in levels of 14 newly-identified transcripts. We conclude ORF10 contributes to more severe COVID-19 clinical outcomes in the human host.


Subject(s)
COVID-19
2.
Schwartz, Ken, Madan, Robert, Kates, Nick, Kates, Nick, Rajji, Tarek, Rajji, Tarek, Kates, Nick, Aelick, Katelynn, Bretzlaff, Monica, Colborne, Debbie Hewitt, Judd, Teresa, McConnell, Jillian, Seguin, Jacquie, Turcotte, Kylie, Liu, Linda, Colborne, Debbie Hewitt, Fortin, Natasha, McConnell, Jillian, Lesiuk, Nancy, Glover, Terri, Koop, Jennifer, Judd, Teresa, Madan, Robert, Schwartz, Kenneth, Colman, Sarah, Tau, Michael, Stanley, Claire, Colman, Sarah, Stanley, Claire, Tau, Michael, Colman, Sarah, Seitz, Dallas, Checkland, Claire, Benjamin, Sophiya, Bruneau, Marie-Andree, Cappella, Antonia, Cassidy, Beverley, Conn, David, Grief, Cindy, Keng, Alvin, Iaboni, Andrea, Grigorovich, Alisa, Kontoa, Pia, Astell, Arlene, McMurray, Josephine, Chu, Charlene, Rodrigues, Kevin, Barned, Claudia, Dementia Isolation Toolkit, Team, Thoo, Vanessa, Giddens-Zuker, Leslie, Benjamin, Sophiya, Ho, Joanne, Carthew, Julie, Cox, Lindsay, Rofaiel, Rymon, Burhan, Amer, Guseva, Elena, Iaboni, Andrea, Herrmann, Nathan, Seitz, Dallas, Burhan, Amer M.; Lanctot, Krista, Lim, Andrew, Wilchesky, Machelle, Iaboni, Andrea, Spasojevic, Sofija, Newman, Kristine, Schindel-Martin, Lori, Ye, Bing, Soltan, Aurelia, Blair, Mervin, McGregor, Carolyn, Burhan, Amer M.; Skosireva, Anna, Gobessi, Linda, Douglass, Alan, Kirkham, Julia, Seitz, Dallas, Goodarzi, Zahra, Denis, Emily St, Malvern, Riley, Sivanthanan, Saskia, Christie, Nathan, Canfield, Amanda, Rowa, Karen, Cassidy, Beverley, Eskes, Gail, Wilson, Ryan, Cassidy, Beverley, Wilton, Steven, Zamora, Nick, Alders, Ashley, Cassidy, Beverley, Wilton, Steven, Checkland, Claire, Zamora, Nick, Alders, Ashley, Kirkham, Julia, Freeland, Alison, Wilkes, Chris, Urness, Doug, Conn, David, Rabheru, Kiran, Checkland, Claire, Cassidy, Keri-Leigh, Rabheru, Kiran, Conn, David, Checkland, Claire, Seitz, Dallas, Abdool, Petal, Mulsant, Benoit H.; Rajji, Tarek K.; Kinjal, Patel, Thitiporn, Supasitthumrong, Seitz, Dallas, Rej, Soham, Clemens, Sara, Heer, Carrie, Devitt, Audrey, Yu, Song Yang, Rostas, Aviva, Cumberbatch, Simonne, Tafler, Melissa, Iroanyah, Ngozi Faith, Sivananthan, Saskia, Apostolides, Haridos, Jaggers, Kaitlyn, Badali, Jocelyn, Guimond, Josée, Sivananthan, Saskia, Martin-Zement, Isabelle, Nadeau-Lessard, Marie-Isabelle, Davies, Kelly, Schryburt-Brown, Kim, Benjamin, Sophiya, Morrison, Adam, Kay, Kelly, Young, Kevin, Kim, Doyoung, Kiss, Alex, Bronskill, Susan E.; Lanctot, Krista L.; Herrmann, Nathan, Gallagher, Damien, Kumar, Sanjeev, Joseph, Shaylyn, Patterson, Rachel, Wang, Wei, Blumberger, Daniel, Rajji, Tarek, Nunes, Paula Villela, Haidar, Atmis Medeiros, Mancine, Livia, Neves, Beatriz Astolfi, Leite, Renata Elaine Paraizo, Pasqualucci, Carlos Augusto, Lafer, Beny, Salvini, Rogerio, Suemoto, Claudia Kimie, King, Annalee, Daniel, Geoff, Hooper, Nancy, Easson-Bruno, Sandra, Lennard, Tamara Nowak, Greco, Martina, Greco, Martina, Veri, Sabrina, Bol, Alexa, Mullaly, Laura, Ostrom, Caroline, Huynh, Dan, Kong, Alice, Thorpe, Lilian, Payne, Sarah, Saperson, Karen, Brown, Michael, Levinson, Anthony, Levinson, Anthony, Payne, Sarah, Hategan, Ana, Esliger, Mandy, Singh, Kathleen, Hickey, Catherine, Chisholm, Terry, Sokoloff, Lisa, Checkland, Claire, Guraya, Jasmeen, Conn, David, Rabheru, Kiran, Seitz, Dallas, Feldman, Sid, Ewa, Vivian, Hunter, Andrea, Conn, David, Rabheru, Kiran, Checkland, Claire, Lee-Cheong, Stephen, Amanullah, Shabbir, Jarvie, Ann, Van Berkum, Amy, Graf, Shauna, Mansour, Reham, Amanallah, Shabbir, Golas, Angela C.; Elgallab, Bishoy M.; Abdool, Petal S.; Bowie, Christopher R.; Rajji, Tarek K.; Cuperfain, Ari, Furqan, Zainab, Sinyor, Mark, Shulman, Kenneth, Zaheer, Juveria, Wathra, Rafae, Mulsant, Benoit, Reynolds, Charles, Lenze, Eric, Karp, Jordan, Daskalakis, Zafiris, Blumberger, Daniel, Gough, Amy, Cassidy, Keri-Leigh, Vallis, Michael, Robinson-Dexter, Jean, Jasrai, Ashitija, Amanullah, Shabbir, Bolshin, Lisa, Khatri, Nasreen, Ryan, Jennifer.
Canadian geriatrics journal : CGJ ; 25(1):88-109, 2022.
Article in English | EuropePMC | ID: covidwho-1749133
3.
biorxiv; 2022.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2022.01.18.476786

ABSTRACT

SARS-CoV-2 has been found capable of inducing prolonged pathologies collectively referred to as Long-COVID. To better understand this biology, we compared the short- and long-term systemic responses in the golden hamster following either SARS-CoV-2 or influenza A virus (IAV) infection. While SARS-CoV-2 exceeded IAV in its capacity to cause injury to the lung and kidney, the most significant changes were observed in the olfactory bulb (OB) and olfactory epithelium (OE) where inflammation was visible beyond one month post SARS-CoV-2 infection. Despite a lack of detectable virus, OB/OE demonstrated microglial and T cell activation, proinflammatory cytokine production, and interferon responses that correlated with behavioral changes. These findings could be corroborated through sequencing of individuals who recovered from COVID-19, as sustained inflammation in OB/OE tissue remained evident months beyond disease resolution. These data highlight a molecular mechanism for persistent COVID-19 symptomology and characterize a small animal model to develop future therapeutics.


Subject(s)
COVID-19 , Seizures , Inflammation , Lung Diseases
4.
ssrn; 2021.
Preprint in English | PREPRINT-SSRN | ID: ppzbmed-10.2139.ssrn.3885245

ABSTRACT

SARS-CoV-2, the virus responsible for the COVID-19 pandemic, has been found capable of inducing long term effects commonly referred to as post-acute sequelae of SARS-CoV-2 (PASC) or long COVID. To define the molecular basis of this condition, we compared the short- and long-term responses to influenza A virus and SARSCoV-2 in the golden hamster model. These data demonstrated that SARS-CoV-2 resulted in sustained changes to lung, kidney, and brain. The most significant change in response to SARS-CoV-2 was observed in the olfactory bulb, where persistent inflammation was visible beyond one month post infection. This was characterized by microglial activation, pro-inflammatory cytokine production, and a Type I interferon (IFN-I) response in the absence of detectable virus. Given the connection between olfactory bulb injury and neurological disorders, we postulate that this prolonged inflammation is an underlying cause of long COVID.Funding Information: This work was funded by generous support from the Marc Haas Foundation, the National Institutes of Health (NCI (R01CA234614) and NIAID (2R01AI107301) and NIDDK (R01DK121072 and 1RO3DK117252) to Department of Medicine, Weill Cornell Medicine (R.E.S.)), and DARPA’s PREPARE Program (HR0011-20-2-0040). The work was further funded by NINDS (NS111251, NSO86444, NSO86444S1)(V.Z., R.A.S.).Ethics Approval Statement: The Tissue Procurement Facility operates under Institutional Review Board (IRB) approved protocol and follows guidelines set by Health Insurance Portability and Accountability Act (HIPAA). Experiments using samples from human subjects were conducted in accordance with local regulations and with the approval of the IRB at the Weill Cornell Medicine. The autopsy samples are considered human tissue research and were collected under IRB protocols 20-04021814 and 19-11021069. All autopsies have consent for research use from next of kin, and these studies were determined as exempt by IRB at Weill Cornell Medicine under those protocol numbers.


Subject(s)
COVID-19 , Influenza, Human , Nervous System Diseases , Wounds and Injuries
5.
biorxiv; 2021.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2021.06.01.446676

ABSTRACT

SARS-CoV-2 infection during pregnancy leads to an increased risk of adverse pregnancy outcomes. Although the placenta itself can be a target of virus infection, most neonates are virus free and are born healthy or recover quickly. Here, we investigated the impact of SARS-CoV-2 infection on the placenta from a cohort of women who were infected late during pregnancy and had tested nasal swab positive for SARS-CoV-2 by qRT-PCR at delivery. SARS-CoV-2 genomic and subgenomic RNA was detected in 23 out of 54 placentas. Two placentas with high virus content were obtained from mothers who presented with severe COVID-19 and whose pregnancies resulted in adverse outcomes for the fetuses, including intrauterine fetal demise and a preterm delivered baby still in newborn intensive care. Examination of the placental samples with high virus content showed efficient SARS-CoV-2 infection, using RNA in situ hybridization to detect genomic and replicating viral RNA, and immunohistochemistry to detect SARS-CoV-2 nucleocapsid protein. Infection was restricted to syncytiotrophoblast cells that envelope the fetal chorionic villi and are in direct contact with maternal blood. The infected placentas displayed massive infiltration of maternal immune cells including macrophages into intervillous spaces, potentially contributing to inflammation of the tissue. Ex vivo infection of placental cultures with SARS-CoV-2 or with SARS-CoV-2 spike (S) protein pseudotyped lentivirus targeted mostly syncytiotrophoblast and in rare events endothelial cells. Infection was reduced by using blocking antibodies against ACE2 and against Neuropilin 1, suggesting that SARS-CoV-2 may utilize alternative receptors for entry into placental cells.


Subject(s)
COVID-19
6.
researchsquare; 2021.
Preprint in English | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-513461.v1

ABSTRACT

COVID-19 patients commonly present with neurological signs of central nervous system (CNS) and/or peripheral nervous system dysfunction. However, which neural cells are permissive to infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been controversial. Here, we show that midbrain dopamine (DA) neurons derived from human pluripotent stem cells (hPSCs) are selectively permissive to SARS-CoV-2 infection both in vitro and upon transplantation in vivo, and that SARS-CoV-2 infection triggers a DA neuron inflammatory and cellular senescence response. A high-throughput screen in hPSC-derived DA neurons identified several FDA approved drugs, including riluzole, metformin, and imatinib, that can rescue the cellular senescence phenotype and prevent SARS-CoV-2 infection. RNA-seq analysis of human ventral midbrain tissue from COVID-19 patients, using formalin-fixed paraffin-embedded autopsy samples, confirmed the induction of an inflammatory and cellular senescence signature and identified low levels of SARS-CoV-2 transcripts. Our findings demonstrate that hPSC-derived DA neurons can serve as a disease model to study neuronal susceptibility to SARS-CoV-2 and to identify candidate neuroprotective drugs for COVID-19 patients. The susceptibility of hPSC-derived DA neurons to SARS-CoV-2 and the observed inflammatory and senescence transcriptional responses suggest the need for careful, long-term monitoring of neurological problems in COVID-19 patients.


Subject(s)
COVID-19
7.
researchsquare; 2020.
Preprint in English | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-94634.v1

ABSTRACT

Heart injury has been reported in up to 20% of COVID-19 patients, yet the cause of myocardial histopathology remains unknown. In order to study the cause of myocardial pathology in COVID-19 patients, we used a hamster model to determine whether following infection SARS-CoV-2, the causative agent of COVID-19, can be detected in heart tissues. Here, we clearly demonstrate that viral RNA and nucleocapsid protein is present in cardiomyocytes in the hearts of infected hamsters. Interestingly, functional cardiomyocyte associated gene expression was decreased in infected hamster hearts, corresponding to an increase in reactive oxygen species (ROS). This data using an animal model was further validated using autopsy heart samples of COVID-19 patients. Moreover, we show that both human pluripotent stem cell-derived cardiomyocytes (hPSC-derived CMs) and adult cardiomyocytes (CMs) can be infected by SARS-CoV-2 and that CCL2 is secreted upon SARS-CoV-2 infection, leading to monocyte recruitment. Increased CCL2 expression and macrophage infiltration was also observed in the hearts of infected hamsters. Using single cell RNA-seq, we also show that macrophages are able to decrease SARS-CoV-2 infection of CMs. Overall, our study provides direct evidence that SARS-CoV-2 infects CMs in vivo and proposes a mechanism of immune-cell infiltration and pathology in heart tissue of COVID-19 patients.


Subject(s)
Severe Acute Respiratory Syndrome , COVID-19 , Heart Diseases
SELECTION OF CITATIONS
SEARCH DETAIL