1
|
Mayer MG, Fischer T. Shared Mechanisms of Blood-Brain Barrier Dysfunction and Neuroinflammation in Coronavirus Disease 2019 and Alzheimer Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00118-X. [PMID: 40254131 DOI: 10.1016/j.ajpath.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/14/2025] [Accepted: 03/21/2025] [Indexed: 04/22/2025]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has highlighted the virus's impact on the central nervous system and its potential to exacerbate neurodegenerative diseases, like Alzheimer disease (AD). Emerging evidence suggests that SARS-CoV-2 infection contributes to chronic neuroinflammation, a key driver in the etiopathogenesis of AD. Shared mechanisms, including blood-brain barrier (BBB) dysfunction, systemic inflammation, and activation of immune pathways, may link SARS-CoV-2 infection to AD onset and/or progression, particularly among vulnerable individuals, such as those of advanced age. This review explores convergent pathways involving the renin-angiotensin-aldosterone system, Wnt/β-catenin signaling, NF-κB activation, and interferon signaling, focusing on their roles in BBB integrity and neuroinflammation. SARS-CoV-2-mediated angiotensin-converting enzyme 2 depletion disrupts renin-angiotensin-aldosterone system homeostasis, favoring proinflammatory signaling that parallels vascular dysfunction in AD. Dysregulation of Wnt/β-catenin signaling exacerbates BBB permeability, whereas NF-κB and interferon pathways contribute to BBB breakdown and propagate central nervous system inflammation via endothelial and immune cell activation. These interactions may amplify prodromal AD pathology and/or initiate AD pathogenesis. By identifying mechanistic overlaps between COVID-19 and AD, this review underlines the need for therapeutic strategies targeting shared pathways of inflammation and BBB dysfunction. Understanding these connections is critical for mitigating the long-term neurologic sequelae of COVID-19 and reducing the burden of AD.
Collapse
Affiliation(s)
| | - Tracy Fischer
- Tulane National Primate Research Center, Covington, Louisiana; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
2
|
Kardol-Hoefnagel T, Luijk B, Reteig L, Haitjema S, Leavis HL, Otten HG. Clara cell 16 kDa protein: an important marker for COVID-19 severity. Front Immunol 2025; 16:1527377. [PMID: 40236699 PMCID: PMC11996771 DOI: 10.3389/fimmu.2025.1527377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
The coronavirus disease 19 (COVID-19) is a disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that invades lung epithelial cells and can lead to severe respiratory failure. In this study, we evaluated whether Clara cell 16 kDa protein (CC16), a serum marker of lung alveolar cell damage, is predictive for disease severity. Patients suspected of SARS-CoV-2 infection were included in this study. Serum levels of Clara cell 16 kDa protein (CC16), soluble Fas Ligand, cytochrome C, thymus- and activation regulated chemokine (TARC) and of oxidate stress related proteins were analyzed. Clinical patient data were extracted from the Utrecht Patient Oriented Database. COVID-19 positive patients were divided in two groups according to disease severity. The mean day difference between COVID-19 diagnosis date and sampling date was +11 days. Concentrations of TARC were lower in COVID-19 positive versus COVID-19 negative patients (unpaired t-test, p=0.002). In addition, CC16 serum levels were significantly elevated in sera taken from patients that were admitted at the intensive care unit (ICU) (p=0.0082). In a matched cohort, sera taken prior to ICU admission (-3 days) contained higher CC16 levels (paired t-test, p=0.0072). Multivariable analyses adjusted for known risk factors (age, gender, blood counts, lactate dehydrogenase, c-reactive protein, underlying disease) showed that CC16 levels were independently associated to COVID-19 severity (interquartile-range, odds ratio 1.53, p=0.0102). In conclusion, our findings highlight CC16 as a promising biomarker for early identification of severe COVID19 cases, which could improve patient management and resource allocation.
Collapse
Affiliation(s)
- Tineke Kardol-Hoefnagel
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Bart Luijk
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Leon Reteig
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Saskia Haitjema
- Utrecht Patient Oriented Database, University Medical Center Utrecht, Utrecht, Netherlands
| | - Helen L. Leavis
- Department of Rheumatology & Clinical Immunology, University Medical Center, Utrecht, Netherlands
| | - Henny G. Otten
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
3
|
Song Z, Clemens RA, Zhang Y, Chen J, Wang Y, Dinauer MC, Meng S. Investigating pulmonary neutrophil responses to inflammation in mice via flow cytometry. J Leukoc Biol 2025; 117:qiae189. [PMID: 39212489 DOI: 10.1093/jleuko/qiae189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/20/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
Neutrophils play a crucial role in maintaining lung health by defending against infections and participating in inflammation processes. Here we describe a detailed protocol for evaluating pulmonary neutrophil phenotype using a murine model of sterile inflammation induced by the fungal cell wall particle zymosan. We provide step-by-step instructions for the isolation of single cells from both lung tissues and airspaces, followed by comprehensive staining techniques for both cell surface markers and intracellular components. This protocol facilitates the sorting and detailed characterization of lung neutrophils via flow cytometry, making it suitable for downstream applications such as mRNA extraction, single-cell sequencing, and analysis of neutrophil heterogeneity. We also identify and discuss essential considerations for conducting successful neutrophil flow cytometry experiments. This work is aimed at researchers exploring the intricate functions of neutrophils in the lung under physiological and pathological conditions with the aid of flow cytometry.
Collapse
Affiliation(s)
- Zhimin Song
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, No. 195, Dongfeng West Road, Guangzhou, Guangdong 510180, China
- Department of Basic Science Research, Guangzhou National Laboratory, No. 9, Xing Dao Huan Bei Road, Guangzhou International Bio Island, Haizhu District, Guangzhou, Guangdong 510005, China
| | - Regina A Clemens
- Departments of Pediatrics, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave, PO Box 8208, St. Louis, MO 63110, United States
| | - Yun Zhang
- Department of Basic Science Research, Guangzhou National Laboratory, No. 9, Xing Dao Huan Bei Road, Guangzhou International Bio Island, Haizhu District, Guangzhou, Guangdong 510005, China
| | - Jingjing Chen
- Department of Basic Science Research, Guangzhou National Laboratory, No. 9, Xing Dao Huan Bei Road, Guangzhou International Bio Island, Haizhu District, Guangzhou, Guangdong 510005, China
| | - Yaofeng Wang
- Department of Basic Science Research, Guangzhou National Laboratory, No. 9, Xing Dao Huan Bei Road, Guangzhou International Bio Island, Haizhu District, Guangzhou, Guangdong 510005, China
| | - Mary C Dinauer
- Departments of Pediatrics, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave, PO Box 8208, St. Louis, MO 63110, United States
- Departments of Pathology and Immunology, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave, PO Box 8208, St. Louis, MO 63110, United States
| | - Shu Meng
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, No. 195, Dongfeng West Road, Guangzhou, Guangdong 510180, China
- Department of Basic Science Research, Guangzhou National Laboratory, No. 9, Xing Dao Huan Bei Road, Guangzhou International Bio Island, Haizhu District, Guangzhou, Guangdong 510005, China
| |
Collapse
|
4
|
Dogadov DI, Kyuregyan KK, Minosyan AA, Goncharenko AM, Shmat EV, Mikhailov MI. [Acute respiratory viral infections in monkeys]. Vopr Virusol 2025; 70:7-24. [PMID: 40233333 DOI: 10.36233/0507-4088-293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Indexed: 04/17/2025]
Abstract
Acute respiratory viral infections (ARVI) are one of the most significant infections affecting the breeding of monkeys, especially among imported and captive primates. Respiratory diseases are also an important cause of morbidity and mortality in wild populations, and most of these infections can affect humans. Many anthropoid species, including apes, are susceptible to ARVI. Outbreaks of spontaneous respiratory infections have been described in many zoos and primatological centers around the world. Moreover, the study of spontaneous and experimental infection in laboratory primates provides an invaluable source of information on the biology and pathogenesis of ARVI and remains an indispensable tool for testing vaccines and drugs. The aim of this literature review was to summarize and analyze published data on the circulation of ARVI causative agents (parainfluenza viruses, adenoviruses, respiratory syncytial virus, influenza viruses, rhinoviruses, coronaviruses, metapneumoviruses, bocaviruses) among wild and captive primates, as well as the results of experimental modeling these infections in monkeys.
Collapse
Affiliation(s)
- D I Dogadov
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - K K Kyuregyan
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
- Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia
| | - A A Minosyan
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - A M Goncharenko
- Kurchatov Complex of Medical Primatology of NRC «Kurchatov Institute»
| | - E V Shmat
- Sochi Institute (branch) of the Federal State Autonomous Educational Institution of Higher Education Peoples' Friendship University of Russia named after Patrice Lumumba
| | - M I Mikhailov
- Central Research Institute of Epidemiology of the Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing
- I.I. Mechnikov Research Institute of Vaccines and Sera
| |
Collapse
|
5
|
Zheng J, Dhakal H, Qing E, Shrestha R, Geller AE, Morrissey SM, Saxena D, Hu X, Li H, Li H, Wilhelmsen K, Wendt LH, Klumpp K, Hume PS, Janssen WJ, Brody R, Palmer KE, Uriarte SM, Ten Eyck P, Meyerholz DK, Merchant ML, McLeish K, Gallagher T, Huang J, Yan J, Perlman S. CXCL12 ameliorates neutrophilia and disease severity in SARS-CoV-2 infection. J Clin Invest 2025; 135:e188222. [PMID: 39773555 PMCID: PMC11827850 DOI: 10.1172/jci188222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025] Open
Abstract
Neutrophils, particularly low-density neutrophils (LDNs), are believed to contribute to acute COVID-19 severity. Here, we showed that neutrophilia can be detected acutely and even months after SARS-CoV-2 infection in patients and mice, while neutrophil depletion reduced disease severity in mice. A key factor in neutrophilia and severe disease in infected mice was traced to the chemokine CXCL12 secreted by bone marrow cells and unexpectedly, endothelial cells. CXCL12 levels were negatively correlated with LDN numbers in longitudinal analyses of patient blood samples. CXCL12 blockade in SARS-CoV-2-infected mice increased blood/lung neutrophil numbers, thereby accelerating disease progression without changing lung virus titers. The exaggerated mortality caused by CXCL12 blockade could be reversed by neutrophil depletion. In addition, blocking interactions between SARS-CoV-2 and angiotensin-converting enzyme 2 (ACE2) reduced CXCL12 levels, suggesting a signal transduction from virus-mediated ACE2 ligation to increased CXCL12 secretion. Collectively, these results demonstrate a previously unappreciated role of CXCL12 in diminishing neutrophilia, including low-density neutrophilia, and its deleterious effects in SARS-CoV-2 infections. The results also support the involvement of SARS-CoV-2-endothelial cell interactions in viral pathogenesis.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Microbiology and Immunology and
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Hima Dhakal
- Department of Microbiology and Immunology and
| | - Enya Qing
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Rejeena Shrestha
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Anne E. Geller
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Samantha M. Morrissey
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Divyasha Saxena
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Xiaoling Hu
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Hong Li
- Functional Immunomics Core, Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | - Haiyan Li
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | | | - Linder H. Wendt
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, USA
| | | | - Patrick S. Hume
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - William J. Janssen
- Department of Medicine, Division of Pulmonary and Critical Care, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
| | - Rachel Brody
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kenneth E. Palmer
- Center for Predictive Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology and
| | - Silvia M. Uriarte
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Patrick Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, Iowa, USA
| | | | | | - Kenneth McLeish
- Department of Medicine, Division of Nephrology and Hypertension and
| | - Tom Gallagher
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Jiapeng Huang
- Department of Anesthesiology and Perioperative Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jun Yan
- Department of Microbiology and Immunology and
- Division of Immunotherapy, the Hiram C. Polk, Jr., MD, Department of Surgery, Immuno-Oncology Program, Brown Cancer Center and
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
6
|
Zhao M, Zhou L, Wang S. Immune crosstalk between respiratory and intestinal mucosal tissues in respiratory infections. Mucosal Immunol 2025:S1933-0219(24)00136-3. [PMID: 39755173 DOI: 10.1016/j.mucimm.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Mucosal tissues, including those in the respiratory and gastrointestinal tracts, are critical barrier surfaces for pathogen invasion. Infections at these sites not only trigger local immune response, but also recruit immune cells from other tissues. Emerging evidence in the mouse models and human samples indicates that the immune crosstalk between the lung and gut critically impacts and determines the course of respiratory disease. Here we summarize the current knowledge of the immune crosstalk between the respiratory and gastrointestinal tracts, and discuss how immune cells are recruited and migrate between these tissues during respiratory infections. We also discuss how commensal bacteria contribute to these processes.
Collapse
Affiliation(s)
- Min Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Zhou
- Shanghai Immune Therapy Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
7
|
Che Mohd Nassir CMN, Che Ramli MD, Jaffer U, Abdul Hamid H, Mehat MZ, Mohamad Ghazali M, Kottakal Cheriya EN. Neurological Sequelae of Post-COVID-19 Fatigue: A Narrative Review of Dipeptidyl Peptidase IV-Mediated Cerebrovascular Complications. Curr Issues Mol Biol 2024; 46:13565-13582. [PMID: 39727939 PMCID: PMC11727395 DOI: 10.3390/cimb46120811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/09/2024] [Accepted: 11/17/2024] [Indexed: 12/28/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a global pandemic affecting millions of people's lives, which has led to 'post-COVID-19 fatigue'. Alarmingly, severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) not only infects the lungs but also influences the heart and brain. Endothelial cell dysfunction and hypercoagulation, which we know occur with this infection, lead to thrombo-inflammation that can manifest as many myriad cardio-cerebrovascular disorders, such as brain fog, fatigue, cognitive dysfunction, etc. Additionally, SARS-CoV-2 has been associated with oxidative stress, protein aggregation, cytokine storm, and mitochondrial dysfunction in neurodegenerative diseases. Accordingly, the identification of molecular targets involved in these actions could provide strategies for preventing and treating this disease. In particular, the very common enzyme dipeptidyl peptidase IV (DPPIV) has recently been identified as a candidate co-receptor for the cell entry of the SARS-CoV-2 virus with its involvement in infection. In addition, DPPIV has been reported as a co-receptor for some viruses such as Middle East respiratory syndrome-coronavirus (MERS-CoV). It mediates immunologic reactions and diseases such as type 2 diabetes mellitus, obesity, and hypertension, which have been considered the prime risk factors for stroke among other types of cardio-cerebrovascular diseases. Unlike angiotensin-converting enzyme 2 (ACE2), DPPIV has been implicated in aggravating the course of infection due to its disruptive effect on inflammatory signaling networks and the neuro-glia-vascular unit. Regarding the neurological, physiological, and molecular grounds governing post-COVID-19 fatigue, this review focuses on DPPIV as one of such reasons that progressively establishes cerebrovascular grievances following SARS-CoV infection.
Collapse
Affiliation(s)
- Che Mohd Nasril Che Mohd Nassir
- Department of Anatomy and Physiology, Faculty of Medicine, School of Basic Medical Sciences, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Terengganu, Malaysia;
| | - Muhammad Danial Che Ramli
- Faculty of Health and Life Sciences, Management and Science University, Shah Alam 40150, Selangor, Malaysia
| | - Usman Jaffer
- Kulliyyah of Islamic Revealed Knowledge and Human Sciences, International Islamic University Malaysia, Kuala Lumpur 50728, Malaysia;
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (H.A.H.); (M.Z.M.)
| | - Muhammad Zulfadli Mehat
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (H.A.H.); (M.Z.M.)
| | - Mazira Mohamad Ghazali
- Department of Anatomy and Physiology, Faculty of Medicine, School of Basic Medical Sciences, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Terengganu, Malaysia;
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Kelantan, Malaysia
| | | |
Collapse
|
8
|
Liang H, Deng Q, Ye W, Jiang Z, Zhang B, Zhang J, Jiang M, Xu Y. Prone position ventilation-induced oxygenation improvement as a valuable predictor of survival in patients with acute respiratory distress syndrome: a retrospective observational study. BMC Pulm Med 2024; 24:575. [PMID: 39567964 PMCID: PMC11577716 DOI: 10.1186/s12890-024-03349-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND In patients with severe acute respiratory distress syndrome (ARDS), prolonged and inappropriate use of prone position ventilation (PPV) is a known risk factor for mortality. Hence, it is critical to monitor patients' response to PPV and accurately differentiate responders from non-responders at an early stage. The study aimed to investigate the relationship between oxygenation improvement after three rounds of PPV and survival rate in patients with pulmonary ARDS. Additionally, we sought to identify the earliest turning point for escalation from PPV to extracorporeal membrane oxygenation. METHODS We performed a retrospective observational study from 2015 to 2023. We included adult patients who received invasive mechanical ventilation, underwent at least three periods of at least 6 h of PPV after admission to the Intensive Care Unit, and meet the ARDS criteria. The study collected data on each PPV session, including changes in PaCO2, PaO2, pH, FiO2, PaO2:FiO2 ratio, and clinical outcomes. RESULTS A total of 104 patients were enrolled in the study. The change in PaCO2 from baseline to the third PPV session (P3) had the highest area under the receiver operating characteristic curve (AUC) of 0.70 (95% CI 0.60-0.80; p < 0.001) for predicting hospital mortality, with an optimal cut-off point of 3.15 (sensitivity 75.9%, specificity 56.0%). The percentage change in PaO2:FiO2 ratio from baseline to P3 also had significant AUC of 0.71 (95% CI 0.61-0.81; p < 0.001) for predicting hospital mortality, with an optimal cut-off value of 99.465 (sensitivity 79.6%, specificity 62.0%). PaCO2 responders were defined as those with an increase in PaCO2 of ≤ 3.15% from baseline to P3, while PaO2:FiO2 responders were defined as those with an increase in PaO2:FiO2 ratio of ≥ 99.465% from baseline to P3. In the multivariable Cox analysis, PaO2:FiO2 responders had a significantly lower 60-day mortality risk (hazard ratio 0.369; 95% CI 0.171-0.798; p = 0.011). CONCLUSIONS The percentage change in PaO2:FiO2 ratio from baseline to P3 was a significant predictor of outcomes. The model fit and prediction accuracy were improved by including the variable of PaCO2 responders.
Collapse
Affiliation(s)
- Hanwen Liang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
- Guangzhou Blood Center, 31 Luyuan Road, Yuexiu District, Guangzhou, Guangdong, 510095, China
| | - Qiuxue Deng
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
- Department of Critical Care Medicine, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Weiyan Ye
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
- Department of Critical Care Medicine, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Zhenjie Jiang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
- Department of Critical Care Medicine, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Baozhu Zhang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
- Department of Critical Care Medicine, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Jiesen Zhang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
- Department of Critical Care Medicine, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China
| | - Mei Jiang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China.
| | - Yuanda Xu
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China.
- Department of Critical Care Medicine, the First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Yuexiu District, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
9
|
Wesley UV, Dempsey RJ. Neuro-molecular perspectives on long COVID-19 impacted cerebrovascular diseases - a role for dipeptidyl peptidase IV. Exp Neurol 2024; 380:114890. [PMID: 39038507 DOI: 10.1016/j.expneurol.2024.114890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/01/2024] [Accepted: 07/14/2024] [Indexed: 07/24/2024]
Abstract
The coronavirus disease 2019 (COVID-19) has caused immense devastation globally with many outcomes that are now extending to its long-term sequel called long COVID. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infects not only lungs, but also the brain and heart in association with endothelial cell dysfunction, coagulation abnormalities, and thrombosis leading to cardio-cerebrovascular health issues. Fatigue, cognitive decline, and brain fog are common neurological symptoms in persisting long COVID. Neurodegenerative processes and SARS-CoV-2 infection manifest overlapping molecular mechanisms, such as cytokine dysregulation, inflammation, protein aggregation, mitochondrial dysfunction, and oxidative stress. Identifying the key molecules in these processes is of importance for prevention and treatment of this disease. In particular, Dipeptidyl peptidase IV (DPPIV), a multifunctional peptidase has recently drawn attention as a potential co-receptor for SARS-CoV-2 infection and cellular entry. DPPIV is a known co-receptor for some other COVID viruses including MERS-Co-V. DPPIV regulates the immune responses, obesity, glucose metabolism, diabetes, and hypertension that are associated with cerebrovascular manifestations including stroke. DPPIV likely worsens persisting COVID-19 by disrupting inflammatory signaling pathways and the neurovascular system. This review highlights the neurological, cellular and molecular processes concerning long COVID, and DPPIV as a potential key factor contributing to cerebrovascular dysfunctions following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Umadevi V Wesley
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA.
| | - Robert J Dempsey
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
| |
Collapse
|
10
|
Palmer CS, Perdios C, Abdel-Mohsen M, Mudd J, Datta PK, Maness NJ, Lehmicke G, Golden N, Hellmers L, Coyne C, Moore Green K, Midkiff C, Williams K, Tiburcio R, Fahlberg M, Boykin K, Kenway C, Russell-Lodrigue K, Birnbaum A, Bohm R, Blair R, Dufour JP, Fischer T, Saied AA, Rappaport J. Non-human primate model of long-COVID identifies immune associates of hyperglycemia. Nat Commun 2024; 15:6664. [PMID: 39164284 PMCID: PMC11335872 DOI: 10.1038/s41467-024-50339-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 07/08/2024] [Indexed: 08/22/2024] Open
Abstract
Hyperglycemia, and exacerbation of pre-existing deficits in glucose metabolism, are manifestations of the post-acute sequelae of SARS-CoV-2. Our understanding of metabolic decline after acute COVID-19 remains unclear due to the lack of animal models. Here, we report a non-human primate model of metabolic post-acute sequelae of SARS-CoV-2 using SARS-CoV-2 infected African green monkeys. Using this model, we identify a dysregulated blood chemokine signature during acute COVID-19 that correlates with elevated and persistent hyperglycemia four months post-infection. Hyperglycemia also correlates with liver glycogen levels, but there is no evidence of substantial long-term SARS-CoV-2 replication in the liver and pancreas. Finally, we report a favorable glycemic effect of the SARS-CoV-2 mRNA vaccine, administered on day 4 post-infection. Together, these data suggest that the African green monkey model exhibits important similarities to humans and can be utilized to assess therapeutic candidates to combat COVID-related metabolic defects.
Collapse
Affiliation(s)
- Clovis S Palmer
- Tulane National Primate Research Center, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| | - Chrysostomos Perdios
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Joseph Mudd
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Prasun K Datta
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Linh Hellmers
- Tulane National Primate Research Center, Covington, LA, USA
| | - Carol Coyne
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Cecily Midkiff
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Rafael Tiburcio
- Division of Experimental Medicine, Department of Medicine, University of San Francisco, CA, USA
| | | | - Kyndal Boykin
- Tulane National Primate Research Center, Covington, LA, USA
| | - Carys Kenway
- Tulane National Primate Research Center, Covington, LA, USA
| | - Kasi Russell-Lodrigue
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Rudolf Bohm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Robert Blair
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jason P Dufour
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Tracy Fischer
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ahmad A Saied
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
11
|
Edwards CT, Karunakaran KA, Garcia E, Beutler N, Gagne M, Golden N, Aoued H, Pellegrini KL, Burnett MR, Honeycutt CC, Lapp SA, Ton T, Lin MC, Metz A, Bombin A, Goff K, Scheuermann SE, Wilkes A, Wood JS, Ehnert S, Weissman S, Curran EH, Roy M, Dessasau E, Paiardini M, Upadhyay AA, Moore I, Maness NJ, Douek DC, Piantadosi A, Andrabi R, Rogers TR, Burton DR, Bosinger SE. Passive infusion of an S2-Stem broadly neutralizing antibody protects against SARS-CoV-2 infection and lower airway inflammation in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605768. [PMID: 39109178 PMCID: PMC11302620 DOI: 10.1101/2024.07.30.605768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The continued evolution of SARS-CoV-2 variants capable of subverting vaccine and infection-induced immunity suggests the advantage of a broadly protective vaccine against betacoronaviruses (β-CoVs). Recent studies have isolated monoclonal antibodies (mAbs) from SARS-CoV-2 recovered-vaccinated donors capable of neutralizing many variants of SARS-CoV-2 and other β-CoVs. Many of these mAbs target the conserved S2 stem region of the SARS-CoV-2 spike protein, rather the receptor binding domain contained within S1 primarily targeted by current SARS-CoV-2 vaccines. One of these S2-directed mAbs, CC40.8, has demonstrated protective efficacy in small animal models against SARS-CoV-2 challenge. As the next step in the pre-clinical testing of S2-directed antibodies as a strategy to protect from SARS-CoV-2 infection, we evaluated the in vivo efficacy of CC40.8 in a clinically relevant non-human primate model by conducting passive antibody transfer to rhesus macaques (RM) followed by SARS-CoV-2 challenge. CC40.8 mAb was intravenously infused at 10mg/kg, 1mg/kg, or 0.1 mg/kg into groups (n=6) of RM, alongside one group that received a control antibody (PGT121). Viral loads in the lower airway were significantly reduced in animals receiving higher doses of CC40.8. We observed a significant reduction in inflammatory cytokines and macrophages within the lower airway of animals infused with 10mg/kg and 1mg/kg doses of CC40.8. Viral genome sequencing demonstrated a lack of escape mutations in the CC40.8 epitope. Collectively, these data demonstrate the protective efficiency of broadly neutralizing S2-targeting antibodies against SARS-CoV-2 infection within the lower airway while providing critical preclinical work necessary for the development of pan-β-CoV vaccines.
Collapse
Affiliation(s)
- Christopher T. Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kirti A. Karunakaran
- Department of Pathology, Microbiology & Immunology, Vanderbilt University, Nashville, TN 37235, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, Minnesota 55356, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Matthew Gagne
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Hadj Aoued
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Kathryn L. Pellegrini
- Emory National Primate Research Center Genomics Core, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Matthew R. Burnett
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher Cole Honeycutt
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stacey A. Lapp
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Thang Ton
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mark C. Lin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Amanda Metz
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Andrei Bombin
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Amelia Wilkes
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer S. Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stephanie Ehnert
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Stacey Weissman
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Elizabeth H. Curran
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Melissa Roy
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Evan Dessasau
- Division of Histology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit A. Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Ian Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Daniel C. Douek
- Vaccine Research Center; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Anne Piantadosi
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas R. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
12
|
Melton A, Rowe LA, Penney T, Krzykwa C, Goff K, Scheuermann SE, Melton HJ, Williams K, Golden N, Green KM, Smith B, Russell-Lodrigue K, Dufour JP, Doyle-Meyers LA, Schiro F, Aye PP, Lifson JD, Beddingfield BJ, Blair RV, Bohm RP, Kolls JK, Rappaport J, Hoxie JA, Maness NJ. The Impact of SIV-Induced Immunodeficiency on SARS-CoV-2 Disease, Viral Dynamics, and Antiviral Immune Response in a Nonhuman Primate Model of Coinfection. Viruses 2024; 16:1173. [PMID: 39066335 PMCID: PMC11281476 DOI: 10.3390/v16071173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The effects of immunodeficiency associated with chronic HIV infection on COVID-19 disease and viral persistence have not been directly addressed in a controlled setting. In this pilot study, we exposed two pigtail macaques (PTMs) chronically infected with SIVmac239, exhibiting from very low to no CD4 T cells across all compartments, to SARS-CoV-2. We monitored the disease progression, viral replication, and evolution, and compared these outcomes with SIV-naïve PTMs infected with SARS-CoV-2. No overt signs of COVID-19 disease were observed in either animal, and the SARS-CoV-2 viral kinetics and evolution in the SIVmac239 PTMs were indistinguishable from those in the SIV-naïve PTMs in all sampled mucosal sites. However, the single-cell RNA sequencing of bronchoalveolar lavage cells revealed an infiltration of functionally inert monocytes after SARS-CoV-2 infection. Critically, neither of the SIV-infected PTMs mounted detectable anti-SARS-CoV-2 T-cell responses nor anti-SARS-CoV-2 binding or neutralizing antibodies. Thus, HIV-induced immunodeficiency alone may not be sufficient to drive the emergence of novel viral variants but may remove the ability of infected individuals to mount adaptive immune responses against SARS-CoV-2.
Collapse
Affiliation(s)
- Alexandra Melton
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Biomedical Science Training Program, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Lori A. Rowe
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Toni Penney
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Clara Krzykwa
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Sarah E. Scheuermann
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Hunter J. Melton
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA;
| | - Kelsey Williams
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Kristyn Moore Green
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Brandon Smith
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Kasi Russell-Lodrigue
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jason P. Dufour
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Lara A. Doyle-Meyers
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Faith Schiro
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Pyone P. Aye
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jeffery D. Lifson
- AIDS and Cancer Viruses Program, Frederick National Laboratory, Frederick, MD 21701, USA;
| | - Brandon J. Beddingfield
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Robert V. Blair
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
| | - Rudolf P. Bohm
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jay K. Kolls
- Departments of Medicine and Pediatrics, Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - James A. Hoxie
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, LA 70433, USA; (A.M.); (L.A.R.); (T.P.); (C.K.); (K.G.); (S.E.S.); (K.W.); (N.G.); (K.M.G.); (B.S.); (K.R.-L.); (J.P.D.); (L.A.D.-M.); (F.S.); (P.P.A.); (B.J.B.); (R.V.B.); (R.P.B.); (J.R.)
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
13
|
Hamlin RE, Blish CA. Challenges and opportunities in long COVID research. Immunity 2024; 57:1195-1214. [PMID: 38865966 PMCID: PMC11210969 DOI: 10.1016/j.immuni.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/19/2024] [Accepted: 05/10/2024] [Indexed: 06/14/2024]
Abstract
Long COVID (LC) is a condition in which patients do not fully recover from the initial SARS-CoV-2 infection but rather have persistent or new symptoms for months to years following the infection. Ongoing research efforts are investigating the pathophysiologic mechanisms of LC and exploring preventative and therapeutic treatment approaches for patients. As a burgeoning area of investigation, LC research can be structured to be more inclusive, innovative, and effective. In this perspective, we highlight opportunities for patient engagement and diverse research expertise, as well as the challenges of developing definitions and reproducible studies. Our intention is to provide a foundation for collaboration and progress in understanding the biomarkers and mechanisms driving LC.
Collapse
Affiliation(s)
| | - Catherine A Blish
- Department of Medicine, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
14
|
Handabile C, Ohno M, Sekiya T, Nomura N, Kawakita T, Kawahara M, Endo M, Nishimura T, Okumura M, Toba S, Sasaki M, Orba Y, Chua BY, Rowntree LC, Nguyen THO, Shingai M, Sato A, Sawa H, Ogasawara K, Kedzierska K, Kida H. Immunogenicity and protective efficacy of a co-formulated two-in-one inactivated whole virus particle COVID-19/influenza vaccine. Sci Rep 2024; 14:4204. [PMID: 38378856 PMCID: PMC10879490 DOI: 10.1038/s41598-024-54421-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/13/2024] [Indexed: 02/22/2024] Open
Abstract
Due to the synchronous circulation of seasonal influenza viruses and severe acute respiratory coronavirus 2 (SARS-CoV-2) which causes coronavirus disease 2019 (COVID-19), there is need for routine vaccination for both COVID-19 and influenza to reduce disease severity. Here, we prepared individual WPVs composed of formalin-inactivated SARS-CoV-2 WK 521 (Ancestral strain; Co WPV) or influenza virus [A/California/07/2009 (X-179A) (H1N1) pdm; Flu WPV] to produce a two-in-one Co/Flu WPV. Serum analysis from vaccinated mice revealed that a single dose of Co/Flu WPV induced antigen-specific neutralizing antibodies against both viruses, similar to those induced by either type of WPV alone. Following infection with either virus, mice vaccinated with Co/Flu WPV showed no weight loss, reduced pneumonia and viral titers in the lung, and lower gene expression of proinflammatory cytokines, as observed with individual WPV-vaccinated. Furthermore, a pentavalent vaccine (Co/qFlu WPV) comprising of Co WPV and quadrivalent influenza vaccine (qFlu WPV) was immunogenic and protected animals from severe COVID-19. These results suggest that a single dose of the two-in-one WPV provides efficient protection against SARS-CoV-2 and influenza virus infections with no evidence of vaccine interference in mice. We propose that concomitant vaccination with the two-in-one WPV can be useful for controlling both diseases.
Collapse
Affiliation(s)
- Chimuka Handabile
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Marumi Ohno
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Toshiki Sekiya
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Naoki Nomura
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Tomomi Kawakita
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Mamiko Kawahara
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | | | | | | | - Shinsuke Toba
- Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Michihito Sasaki
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yasuko Orba
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Brendon Y Chua
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Masashi Shingai
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Akihiko Sato
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hirofumi Sawa
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Kazumasa Ogasawara
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Katherine Kedzierska
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Hiroshi Kida
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan.
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
15
|
Hua S, Latha K, Marlin R, Benmeziane K, Bossevot L, Langlois S, Relouzat F, Dereuddre-Bosquet N, Le Grand R, Cavarelli M. Intestinal immunological events of acute and resolved SARS-CoV-2 infection in non-human primates. Mucosal Immunol 2024; 17:25-40. [PMID: 37827377 DOI: 10.1016/j.mucimm.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023]
Abstract
SARS-CoV-2 infection has been associated with intestinal mucosal barrier damage, leading to microbial and endotoxin translocation, heightened inflammatory responses, and aggravated disease outcomes. This study aimed to investigate the immunological mechanisms associated with impaired intestinal barrier function. We conducted a comprehensive analysis of gut damage and inflammation markers and phenotypic characterization of myeloid and lymphoid populations in the ileum and colon of SARS-CoV-2-exposed macaques during both the acute and resolved infection phases. Our findings revealed a significant accumulation of terminally differentiated and activated CD4+ and CD8+ T cells, along with memory B cells, within the gastrointestinal tract up to 43 days after exposure to SARS-CoV-2. This robust infection-induced immune response was accompanied by a notable depletion of plasmacytoid dendritic cells, myeloid dendritic cells, and macrophages, particularly affecting the colon during the resolved infection phase. Additionally, we identified a population of CX3CR1Low inflammatory macrophages associated with intestinal damage during active viral replication. Elevated levels of immune activation and gut damage markers, and perturbation of macrophage homeostasis, persisted even after the resolution of the infection, suggesting potential long-term clinical sequelae. These findings enhance our understanding of gastrointestinal immune pathology following SARS-CoV-2 infection and provide valuable information for developing and testing medical countermeasures.
Collapse
Affiliation(s)
- Stéphane Hua
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Krishna Latha
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Romain Marlin
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Keltouma Benmeziane
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Laetitia Bossevot
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Sébastien Langlois
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Mariangela Cavarelli
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France.
| |
Collapse
|
16
|
Chen Z, Yuan Y, Hu Q, Zhu A, Chen F, Li S, Guan X, Lv C, Tang T, He Y, Cheng J, Zheng J, Hu X, Zhao J, Zhao J, Sun J. SARS-CoV-2 immunity in animal models. Cell Mol Immunol 2024; 21:119-133. [PMID: 38238440 PMCID: PMC10806257 DOI: 10.1038/s41423-023-01122-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
The COVID-19 pandemic, which was caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a worldwide health crisis due to its transmissibility. SARS-CoV-2 infection results in severe respiratory illness and can lead to significant complications in affected individuals. These complications encompass symptoms such as coughing, respiratory distress, fever, infectious shock, acute respiratory distress syndrome (ARDS), and even multiple-organ failure. Animal models serve as crucial tools for investigating pathogenic mechanisms, immune responses, immune escape mechanisms, antiviral drug development, and vaccines against SARS-CoV-2. Currently, various animal models for SARS-CoV-2 infection, such as nonhuman primates (NHPs), ferrets, hamsters, and many different mouse models, have been developed. Each model possesses distinctive features and applications. In this review, we elucidate the immune response elicited by SARS-CoV-2 infection in patients and provide an overview of the characteristics of various animal models mainly used for SARS-CoV-2 infection, as well as the corresponding immune responses and applications of these models. A comparative analysis of transcriptomic alterations in the lungs from different animal models revealed that the K18-hACE2 and mouse-adapted virus mouse models exhibited the highest similarity with the deceased COVID-19 patients. Finally, we highlighted the current gaps in related research between animal model studies and clinical investigations, underscoring lingering scientific questions that demand further clarification.
Collapse
Affiliation(s)
- Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yaochang Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Fenghua Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Shu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xin Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Chao Lv
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Tian Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Yiyun He
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jinling Cheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jie Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Xiaoyu Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510005, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, 518005, China.
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China.
| |
Collapse
|
17
|
Zhou W, Cao X, Xu Q, Qu J, Sun Y. The double-edged role of neutrophil heterogeneity in inflammatory diseases and cancers. MedComm (Beijing) 2023; 4:e325. [PMID: 37492784 PMCID: PMC10363828 DOI: 10.1002/mco2.325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 07/27/2023] Open
Abstract
Neutrophils are important immune cells act as the body's first line of defense against infection and respond to diverse inflammatory cues. Many studies have demonstrated that neutrophils display plasticity in inflammatory diseases and cancers. Clarifying the role of neutrophil heterogeneity in inflammatory diseases and cancers will contribute to the development of novel treatment strategies. In this review, we have presented a review on the development of the understanding on neutrophil heterogeneity from the traditional perspective and a high-resolution viewpoint. A growing body of evidence has confirmed the double-edged role of neutrophils in inflammatory diseases and tumors. This may be due to a lack of precise understanding of the role of specific neutrophil subsets in the disease. Thus, elucidating specific neutrophil subsets involved in diseases would benefit the development of precision medicine. Thusly, we have summarized the relevance and actions of neutrophil heterogeneity in inflammatory diseases and cancers comprehensively. Meanwhile, we also discussed the potential intervention strategy for neutrophils. This review is intended to deepen our understanding of neutrophil heterogeneity in inflammatory diseases and cancers, while hold promise for precise treatment of neutrophil-related diseases.
Collapse
Affiliation(s)
- Wencheng Zhou
- Department of PharmacyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Xinran Cao
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Yang Sun
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| |
Collapse
|
18
|
Izadpanah A, Mudd JC, Garcia JGN, Srivastav S, Abdel-Mohsen M, Palmer C, Goldman AR, Kolls JK, Qin X, Rappaport J. SARS-CoV-2 infection dysregulates NAD metabolism. Front Immunol 2023; 14:1158455. [PMID: 37457744 PMCID: PMC10344451 DOI: 10.3389/fimmu.2023.1158455] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/19/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Severe COVID-19 results initially in pulmonary infection and inflammation. Symptoms can persist beyond the period of acute infection, and patients with Post-Acute Sequelae of COVID (PASC) often exhibit a variety of symptoms weeks or months following acute phase resolution including continued pulmonary dysfunction, fatigue, and neurocognitive abnormalities. We hypothesized that dysregulated NAD metabolism contributes to these abnormalities. Methods RNAsequencing of lungs from transgenic mice expressing human ACE2 (K18-hACE2) challenged with SARS-CoV-2 revealed upregulation of NAD biosynthetic enzymes, including NAPRT1, NMNAT1, NAMPT, and IDO1 6 days post-infection. Results Our data also demonstrate increased gene expression of NAD consuming enzymes: PARP 9,10,14 and CD38. At the same time, SIRT1, a protein deacetylase (requiring NAD as a cofactor and involved in control of inflammation) is downregulated. We confirmed our findings by mining sequencing data from lungs of patients that died from SARS-CoV-2 infection. Our validated findings demonstrating increased NAD turnover in SARS-CoV-2 infection suggested that modulating NAD pathways may alter disease progression and may offer therapeutic benefits. Specifically, we hypothesized that treating K18-hACE2 mice with nicotinamide riboside (NR), a potent NAD precursor, may mitigate lethality and improve recovery from SARS-CoV-2 infection. We also tested the therapeutic potential of an anti- monomeric NAMPT antibody using the same infection model. Treatment with high dose anti-NAMPT antibody resulted in significantly decreased body weight compared to control, which was mitigated by combining HD anti-NAMPT antibody with NR. We observed a significant increase in lipid metabolites, including eicosadienoic acid, oleic acid, and palmitoyl carnitine in the low dose antibody + NR group. We also observed significantly increased nicotinamide related metabolites in NR treated animals. Discussion Our data suggest that infection perturbs NAD pathways, identify novel mechanisms that may explain some pathophysiology of CoVID-19 and suggest novel strategies for both treatment and prevention.
Collapse
Affiliation(s)
- Amin Izadpanah
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| | - Joseph C. Mudd
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| | - Joe G. N. Garcia
- Department of Medicine, College of Medicine Tucson, University of Arizona, Tucson, AZ, United States
| | - Sudesh Srivastav
- Biostatistics and Data Science, Tulane University School of Public Health, New Orleans, LA, United States
| | | | - Clovis Palmer
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| | - Aaron R. Goldman
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, United States
- Proteomics and Metabolomics Shared Resource, The Wistar Institute, Philadelphia, PA, United States
| | - Jay K. Kolls
- Center for Translational Research in Infection and Inflammation, Tulane School of Medicine, New Orleans, Louisiana, LA, United States
| | - Xuebin Qin
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, Louisiana, LA, United States
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, LA, United States
| |
Collapse
|
19
|
Upadhyay AA, Viox EG, Hoang TN, Boddapati AK, Pino M, Lee MYH, Corry J, Strongin Z, Cowan DA, Beagle EN, Horton TR, Hamilton S, Aoued H, Harper JL, Edwards CT, Nguyen K, Pellegrini KL, Tharp GK, Piantadosi A, Levit RD, Amara RR, Barratt-Boyes SM, Ribeiro SP, Sekaly RP, Vanderford TH, Schinazi RF, Paiardini M, Bosinger SE. TREM2 + and interstitial-like macrophages orchestrate airway inflammation in SARS-CoV-2 infection in rhesus macaques. Nat Commun 2023; 14:1914. [PMID: 37024448 PMCID: PMC10078029 DOI: 10.1038/s41467-023-37425-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/16/2023] [Indexed: 04/08/2023] Open
Abstract
The immunopathological mechanisms driving the development of severe COVID-19 remain poorly defined. Here, we utilize a rhesus macaque model of acute SARS-CoV-2 infection to delineate perturbations in the innate immune system. SARS-CoV-2 initiates a rapid infiltration of plasmacytoid dendritic cells into the lower airway, commensurate with IFNA production, natural killer cell activation, and a significant increase of blood CD14-CD16+ monocytes. To dissect the contribution of lung myeloid subsets to airway inflammation, we generate a longitudinal scRNA-Seq dataset of airway cells, and map these subsets to corresponding populations in the human lung. SARS-CoV-2 infection elicits a rapid recruitment of two macrophage subsets: CD163+MRC1-, and TREM2+ populations that are the predominant source of inflammatory cytokines. Treatment with baricitinib (Olumiant®), a JAK1/2 inhibitor is effective in eliminating the influx of non-alveolar macrophages, with a reduction of inflammatory cytokines. This study delineates the major lung macrophage subsets driving airway inflammation during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Amit A Upadhyay
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Elise G Viox
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Timothy N Hoang
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Arun K Boddapati
- Emory NPRC Genomics Core Laboratory, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Maria Pino
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Michelle Y-H Lee
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Jacqueline Corry
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary Strongin
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - David A Cowan
- Emory NPRC Genomics Core Laboratory, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Elizabeth N Beagle
- Emory NPRC Genomics Core Laboratory, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Tristan R Horton
- Emory NPRC Genomics Core Laboratory, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Sydney Hamilton
- Emory NPRC Genomics Core Laboratory, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Hadj Aoued
- Emory NPRC Genomics Core Laboratory, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Justin L Harper
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Christopher T Edwards
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Kevin Nguyen
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Kathryn L Pellegrini
- Emory NPRC Genomics Core Laboratory, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Gregory K Tharp
- Emory NPRC Genomics Core Laboratory, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Anne Piantadosi
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Rebecca D Levit
- Department of Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Rama R Amara
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Simon M Barratt-Boyes
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susan P Ribeiro
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Rafick P Sekaly
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA
| | - Thomas H Vanderford
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Raymond F Schinazi
- Department of Pediatrics, School of Medicine, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA.
| | - Steven E Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
20
|
Maity S, Mayer MG, Shu Q, Linh H, Bao D, Blair RV, He Y, Lyon CJ, Hu TY, Fischer T, Fan J. Cerebrospinal Fluid Protein Markers Indicate Neuro-Damage in SARS-CoV-2-Infected Nonhuman Primates. Mol Cell Proteomics 2023; 22:100523. [PMID: 36870567 PMCID: PMC9981268 DOI: 10.1016/j.mcpro.2023.100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 02/18/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Neurologic manifestations are among the most frequently reported complications of COVID-19. However, given the paucity of tissue samples and the highly infectious nature of the etiologic agent of COVID-19, we have limited information to understand the neuropathogenesis of COVID-19. Therefore, to better understand the impact of COVID-19 on the brain, we used mass-spectrometry-based proteomics with a data-independent acquisition mode to investigate cerebrospinal fluid (CSF) proteins collected from two different nonhuman primates, Rhesus Macaque and African Green Monkeys, for the neurologic effects of the infection. These monkeys exhibited minimal to mild pulmonary pathology but moderate to severe central nervous system (CNS) pathology. Our results indicated that CSF proteome changes after infection resolution corresponded with bronchial virus abundance during early infection and revealed substantial differences between the infected nonhuman primates and their age-matched uninfected controls, suggesting these differences could reflect altered secretion of CNS factors in response to SARS-CoV-2-induced neuropathology. We also observed the infected animals exhibited highly scattered data distributions compared to their corresponding controls indicating the heterogeneity of the CSF proteome change and the host response to the viral infection. Dysregulated CSF proteins were preferentially enriched in functional pathways associated with progressive neurodegenerative disorders, hemostasis, and innate immune responses that could influence neuroinflammatory responses following COVID-19. Mapping these dysregulated proteins to the Human Brain Protein Atlas found that they tended to be enriched in brain regions that exhibit more frequent injury following COVID-19. It, therefore, appears reasonable to speculate that such CSF protein changes could serve as signatures for neurologic injury, identify important regulatory pathways in this process, and potentially reveal therapeutic targets to prevent or attenuate the development of neurologic injuries following COVID-19.
Collapse
Affiliation(s)
- Sudipa Maity
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Meredith G Mayer
- Division of Comparative Pathology, National Primate Research Center, Covington, Louisiana, USA
| | - Qingbo Shu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Hellmers Linh
- Division of Comparative Pathology, National Primate Research Center, Covington, Louisiana, USA
| | - Duran Bao
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Robert V Blair
- Division of Comparative Pathology, National Primate Research Center, Covington, Louisiana, USA
| | - Yanlin He
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Christopher J Lyon
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tony Y Hu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tracy Fischer
- Division of Comparative Pathology, National Primate Research Center, Covington, Louisiana, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jia Fan
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana, USA; Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
| |
Collapse
|
21
|
Gonzalez AA, Olsen EL, Killeen SZ, Blair RV, Seshan SV, Jaimes EA, Roy CJ, Prieto MC. Elevated soluble urokinase plasminogen activator receptor is associated with renal dysfunction in a Chlorocebus atheiops COVID-19 model. J Med Primatol 2023; 52:131-134. [PMID: 36377612 PMCID: PMC10023264 DOI: 10.1111/jmp.12626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/18/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022]
Abstract
Increases of soluble urokinase plasminogen activator receptor (suPAR) were measured in both urine and plasma of a Chlorocebus aethiops (African green monkey; AGM) mucosal infected with SARS-CoV-2. The data indicate that elevated suPAR may be associated with renal dysfunction and pathology in the context of COVID-19.
Collapse
Affiliation(s)
| | - Emily L. Olsen
- Department of Microbiology & Immunology, Tulane School of Medicine, New Orleans, LA, USA
- Tulane National Primate Research Center, Covington, LA, USA
| | | | | | - Surya V. Seshan
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Edgar A. Jaimes
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chad J Roy
- Department of Microbiology & Immunology, Tulane School of Medicine, New Orleans, LA, USA
- Tulane National Primate Research Center, Covington, LA, USA
| | - Minolfa C. Prieto
- Department of Physiology and Tulane Renal Hypertension Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
22
|
Calvert BA, Quiroz EJ, Lorenzana Z, Doan N, Kim S, Senger CN, Anders JJ, Wallace WD, Salomon MP, Henley J, Ryan AL. Neutrophilic inflammation promotes SARS-CoV-2 infectivity and augments the inflammatory responses in airway epithelial cells. Front Immunol 2023; 14:1112870. [PMID: 37006263 PMCID: PMC10061003 DOI: 10.3389/fimmu.2023.1112870] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction In response to viral infection, neutrophils release inflammatory mediators as part of the innate immune response, contributing to pathogen clearance through virus internalization and killing. Pre- existing co-morbidities correlating to incidence to severe COVID-19 are associated with chronic airway neutrophilia. Furthermore, examination of COVID-19 explanted lung tissue revealed a series of epithelial pathologies associated with the infiltration and activation of neutrophils, indicating neutrophil activity in response to SARS-CoV-2 infection. Methods To determine the impact of neutrophil-epithelial interactions on the infectivity and inflammatory responses to SARS-CoV-2 infection, we developed a co-culture model of airway neutrophilia. This model was infected with live SARS-CoV-2 virus the epithelial response to infection was evaluated. Results SARS-CoV-2 infection of airway epithelium alone does not result in a notable pro-inflammatory response from the epithelium. The addition of neutrophils induces the release of proinflammatory cytokines and stimulates a significantly augmented proinflammatory response subsequent SARS-CoV-2 infection. The resulting inflammatory responses are polarized with differential release from the apical and basolateral side of the epithelium. Additionally, the integrity of the \epithelial barrier is impaired with notable epithelial damage and infection of basal stem cells. Conclusions This study reveals a key role for neutrophil-epithelial interactions in determining inflammation and infectivity.
Collapse
Affiliation(s)
- Ben A. Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Erik J. Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zareeb Lorenzana
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ngan Doan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Seongjae Kim
- The Salk Institute of Biological Studies, La Jolla, CA, United States
| | - Christiana N. Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jeffrey J. Anders
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
| | - Wiliam D. Wallace
- Department of Pathology, University of Southern California, Los Angeles, CA, United States
| | - Matthew P. Salomon
- Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jill Henley
- Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa, IA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
23
|
Martínez-Diz S, Marín-Benesiu F, López-Torres G, Santiago O, Díaz-Cuéllar JF, Martín-Esteban S, Cortés-Valverde AI, Arenas-Rodríguez V, Cuenca-López S, Porras-Quesada P, Ruiz-Ruiz C, Abadía-Molina AC, Entrala-Bernal C, Martínez-González LJ, Álvarez-Cubero MJ. Relevance of TMPRSS2, CD163/CD206, and CD33 in clinical severity stratification of COVID-19. Front Immunol 2023; 13:1094644. [PMID: 36969980 PMCID: PMC10031647 DOI: 10.3389/fimmu.2022.1094644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/15/2022] [Indexed: 03/10/2023] Open
Abstract
BackgroundApproximately 13.8% and 6.1% of coronavirus disease 2019 (COVID-19) patients require hospitalization and sometimes intensive care unit (ICU) admission, respectively. There is no biomarker to predict which of these patients will develop an aggressive stage that we could improve their quality of life and healthcare management. Our main goal is to include new markers for the classification of COVID-19 patients.MethodsTwo tubes of peripheral blood were collected from a total of 66 (n = 34 mild and n = 32 severe) samples (mean age 52 years). Cytometry analysis was performed using a 15-parameter panel included in the Maxpar® Human Monocyte/Macrophage Phenotyping Panel Kit. Cytometry by time-of-flight mass spectrometry (CyTOF) panel was performed in combination with genetic analysis using TaqMan® probes for ACE2 (rs2285666), MX1 (rs469390), and TMPRSS2 (rs2070788) variants. GemStone™ and OMIQ software were used for cytometry analysis.ResultsThe frequency of CD163+/CD206- population of transitional monocytes (T-Mo) was decreased in the mild group compared to that of the severe one, while T-Mo CD163-/CD206- were increased in the mild group compared to that of the severe one. In addition, we also found differences in CD11b expression in CD14dim monocytes in the severe group, with decreased levels in the female group (p = 0.0412). When comparing mild and severe disease, we also found that CD45- [p = 0.014; odds ratio (OR) = 0.286, 95% CI 0.104–0.787] and CD14dim/CD33+ (p = 0.014; OR = 0.286, 95% CI 0.104–0.787) monocytes were the best options as biomarkers to discriminate between these patient groups. CD33 was also indicated as a good biomarker for patient stratification by the analysis of GemStone™ software. Among genetic markers, we found that G carriers of TMPRSS2 (rs2070788) have an increased risk (p = 0.02; OR = 3.37, 95% CI 1.18–9.60) of severe COVID-19 compared to those with A/A genotype. This strength is further increased when combined with CD45-, T-Mo CD163+/CD206-, and C14dim/CD33+.ConclusionsHere, we report the interesting role of TMPRSS2, CD45-, CD163/CD206, and CD33 in COVID-19 aggressiveness. This strength is reinforced for aggressiveness biomarkers when TMPRSS2 and CD45-, TMPRSS2 and CD163/CD206, and TMPRSS2 and CD14dim/CD33+ are combined.
Collapse
Affiliation(s)
- Silvia Martínez-Diz
- Preventive Medicine and Public Health Service, Hospital Universitario Clínico San Cecilio, Granada, Spain
| | - Fernando Marín-Benesiu
- GENYO, Center for Genomics and Oncological Research, Granada, Spain
- Department of Biochemistry, Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
| | | | - Olivia Santiago
- GENYO, Center for Genomics and Oncological Research, Granada, Spain
| | | | | | | | | | | | | | - Carmen Ruiz-Ruiz
- Department of Biochemistry, Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
- Immunology Unit, Institute of Regenerative Biomedicine (IBIMER), Center for Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| | - Ana C. Abadía-Molina
- Department of Biochemistry, Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
- Immunology Unit, Institute of Regenerative Biomedicine (IBIMER), Center for Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| | - Carmen Entrala-Bernal
- LORGEN G.P., PT, Ciencias de la Salud - Business Innovation Centre (BIC), Granada, Spain
| | - Luis J. Martínez-González
- GENYO, Center for Genomics and Oncological Research, Granada, Spain
- *Correspondence: Luis J. Martínez-González,
| | - Maria Jesus Álvarez-Cubero
- GENYO, Center for Genomics and Oncological Research, Granada, Spain
- Department of Biochemistry, Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
- Biosanitary Research Institute (ibs. GRANADA), University of Granada, Granada, Spain
| |
Collapse
|
24
|
Monitoring and immunogenicity of SARS-CoV-2 vaccination of laboratory rhesus monkeys (Macaca mulatta). Sci Rep 2023; 13:3274. [PMID: 36841887 PMCID: PMC9958316 DOI: 10.1038/s41598-023-30473-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/23/2023] [Indexed: 02/27/2023] Open
Abstract
The availability of effective vaccines and a high vaccination rate allowed the recent mitigation, or even withdrawal, of many protective measures for containing the SARS CoV-2 pandemic. At the same time, new and highly mutated variants of the virus are found to have significantly higher transmissibility and reduced vaccine efficacy, thus causing high infection rates during the third year of the pandemic. The combination of reduced measures and increased infectivity poses a particular risk for unvaccinated individuals, including animals susceptible to the virus. Among the latter, non-human primates (NHPs) are particularly vulnerable. They serve as important models in various fields of biomedical research and because of their cognitive capabilities, they receive particular attention in animal welfare regulations around the world. Yet, although they played an extraordinarily important role for developing and testing vaccines against SARS-CoV-2, the protection of captive rhesus monkeys against Covid-19 has rarely been discussed. We here report upon twofold mRNA vaccination of a cohort of 19 elderly rhesus monkeys (Macaca mulatta) against infection by SARS-CoV-2. All animals were closely monitored on possible side effects of vaccination, and were tested for neutralising antibodies against the virus. The data show that vaccination of rhesus monkeys is a safe and reliable measure to protect these animals against SARS-CoV-2.
Collapse
|
25
|
Dillard JA, Martinez SA, Dearing JJ, Montgomery SA, Baxter AK. Animal Models for the Study of SARS-CoV-2-Induced Respiratory Disease and Pathology. Comp Med 2023; 73:72-90. [PMID: 36229170 PMCID: PMC9948904 DOI: 10.30802/aalas-cm-22-000089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Emergence of the betacoronavirus SARS-CoV-2 has resulted in a historic pandemic, with millions of deaths worldwide. An unprecedented effort has been made by the medical, scientific, and public health communities to rapidly develop and implement vaccines and therapeutics to prevent and reduce hospitalizations and deaths. Although SARS-CoV-2 infection can lead to disease in many organ systems, the respiratory system is its main target, with pneumonia and acute respiratory distress syndrome as the hallmark features of severe disease. The large number of patients who have contracted COVID-19 infections since 2019 has permitted a detailed characterization of the clinical and pathologic features of the disease in humans. However, continued progress in the development of effective preventatives and therapies requires a deeper understanding of the pathogenesis of infection. Studies using animal models are necessary to complement in vitro findings and human clinical data. Multiple animal species have been evaluated as potential models for studying the respiratory disease caused by SARSCoV-2 infection. Knowing the similarities and differences between animal and human responses to infection is critical for effective translation of animal data into human medicine. This review provides a detailed summary of the respiratory disease and associated pathology induced by SARS-CoV-2 infection in humans and compares them with the disease that develops in 3 commonly used models: NHP, hamsters, and mice. The effective use of animals to study SARS-CoV-2-induced respiratory disease will enhance our understanding of SARS-CoV-2 pathogenesis, allow the development of novel preventatives and therapeutics, and aid in the preparation for the next emerging virus with pandemic potential.
Collapse
Key Words
- ace2, angiotensin-converting enzyme 2
- agm, african green monkey
- ali, acute lung injury
- ards, acute respiratory distress syndrome
- balf, bronchoalveolar lavage fluid
- cards, covid-19-associated acute respiratory distress syndrome
- dad, diffuse alveolar damage
- dpi, days postinfection
- ggo, ground glass opacities
- s, spike glycoprotein
Collapse
Affiliation(s)
- Jacob A Dillard
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sabian A Martinez
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Justin J Dearing
- Biological and Biomedical Sciences Program, Office of Graduate Education, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephanie A Montgomery
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andvictoria K Baxter
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;,
| |
Collapse
|
26
|
Arish M, Qian W, Narasimhan H, Sun J. COVID-19 immunopathology: From acute diseases to chronic sequelae. J Med Virol 2023; 95:e28122. [PMID: 36056655 PMCID: PMC9537925 DOI: 10.1002/jmv.28122] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 01/17/2023]
Abstract
The clinical manifestation of coronavirus disease 2019 (COVID-19) mainly targets the lung as a primary affected organ, which is also a critical site of immune cell activation by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, recent reports also suggest the involvement of extrapulmonary tissues in COVID-19 pathology. The interplay of both innate and adaptive immune responses is key to COVID-19 management. As a result, a robust innate immune response provides the first line of defense, concomitantly, adaptive immunity neutralizes the infection and builds memory for long-term protection. However, dysregulated immunity, both innate and adaptive, can skew towards immunopathology both in acute and chronic cases. Here we have summarized some of the recent findings that provide critical insight into the immunopathology caused by SARS-CoV-2, in acute and post-acute cases. Finally, we further discuss some of the immunomodulatory drugs in preclinical and clinical trials for dampening the immunopathology caused by COVID-19.
Collapse
Affiliation(s)
- Mohd Arish
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Wei Qian
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Harish Narasimhan
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Jie Sun
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
27
|
Qi F, Qin C. Characteristics of animal models for COVID-19. Animal Model Exp Med 2022; 5:401-409. [PMID: 36301011 PMCID: PMC9610135 DOI: 10.1002/ame2.12278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19), the most consequential pandemic of this century, threatening human health and public safety. SARS-CoV-2 has been continuously evolving through mutation of its genome and variants of concern have emerged. The World Health Organization R&D Blueprint plan convened a range of expert groups to develop animal models for COVID-19, a core requirement for the prevention and control of SARS-CoV-2 pandemic. The animal model construction techniques developed during the SARS-CoV and MERS-CoV pandemics were rapidly deployed and applied in the establishment of COVID-19 animal models. To date, a large number of animal models for COVID-19, including mice, hamsters, minks and nonhuman primates, have been established. Infectious diseases produce unique manifestations according to the characteristics of the pathogen and modes of infection. Here we classified animal model resources around the infection route of SARS-CoV-2, and summarized the characteristics of the animal models constructed via transnasal, localized, and simulated transmission routes of infection.
Collapse
Affiliation(s)
- Feifei Qi
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina,National Center of Technology Innovation for Animal ModelBeijingChina
| | - Chuan Qin
- Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine CenterPeking Union Medical CollegeBeijingChina,National Center of Technology Innovation for Animal ModelBeijingChina
| |
Collapse
|
28
|
Wei X, Rong N, Liu J. Prospects of animal models and their application in studies on adaptive immunity to SARS-CoV-2. Front Immunol 2022; 13:993754. [PMID: 36189203 PMCID: PMC9523127 DOI: 10.3389/fimmu.2022.993754] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/05/2022] [Indexed: 01/08/2023] Open
Abstract
The adaptive immune response induced by SARS-CoV-2 plays a key role in the antiviral process and can protect the body from the threat of infection for a certain period of time. However, owing to the limitations of clinical studies, the antiviral mechanisms, protective thresholds, and persistence of the immune memory of adaptive immune responses remain unclear. This review summarizes existing research models for SARS-CoV-2 and elaborates on the advantages of animal models in simulating the clinical symptoms of COVID-19 in humans. In addition, we systematically summarize the research progress on the SARS-CoV-2 adaptive immune response and the remaining key issues, as well as the application and prospects of animal models in this field. This paper provides direction for in-depth analysis of the anti-SARS-CoV-2 mechanism of the adaptive immune response and lays the foundation for the development and application of vaccines and drugs.
Collapse
Affiliation(s)
- Xiaohui Wei
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | | | - Jiangning Liu
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
29
|
Jansen EB, Orvold SN, Swan CL, Yourkowski A, Thivierge BM, Francis ME, Ge A, Rioux M, Darbellay J, Howland JG, Kelvin AA. After the virus has cleared-Can preclinical models be employed for Long COVID research? PLoS Pathog 2022; 18:e1010741. [PMID: 36070309 PMCID: PMC9451097 DOI: 10.1371/journal.ppat.1010741] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) can cause the life-threatening acute respiratory disease called COVID-19 (Coronavirus Disease 2019) as well as debilitating multiorgan dysfunction that persists after the initial viral phase has resolved. Long COVID or Post-Acute Sequelae of COVID-19 (PASC) is manifested by a variety of symptoms, including fatigue, dyspnea, arthralgia, myalgia, heart palpitations, and memory issues sometimes affecting between 30% and 75% of recovering COVID-19 patients. However, little is known about the mechanisms causing Long COVID and there are no widely accepted treatments or therapeutics. After introducing the clinical aspects of acute COVID-19 and Long COVID in humans, we summarize the work in animals (mice, Syrian hamsters, ferrets, and nonhuman primates (NHPs)) to model human COVID-19. The virology, pathology, immune responses, and multiorgan involvement are explored. Additionally, any studies investigating time points longer than 14 days post infection (pi) are highlighted for insight into possible long-term disease characteristics. Finally, we discuss how the models can be leveraged for treatment evaluation, including pharmacological agents that are currently in human clinical trials for treating Long COVID. The establishment of a recognized Long COVID preclinical model representing the human condition would allow the identification of mechanisms causing disease as well as serve as a vehicle for evaluating potential therapeutics.
Collapse
Affiliation(s)
- Ethan B. Jansen
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Spencer N. Orvold
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Cynthia L. Swan
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anthony Yourkowski
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Brittany M. Thivierge
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Magen E. Francis
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anni Ge
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Melissa Rioux
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Joseph Darbellay
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John G. Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Alyson A. Kelvin
- Vaccine and Infectious Disease Organization VIDO, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
30
|
Brosseau LM, Escandón K, Ulrich AK, Rasmussen AL, Roy CJ, Bix GJ, Popescu SV, Moore KA, Osterholm MT. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Dose, Infection, and Disease Outcomes for Coronavirus Disease 2019 (COVID-19): A Review. Clin Infect Dis 2022; 75:e1195-e1201. [PMID: 34651164 PMCID: PMC8524637 DOI: 10.1093/cid/ciab903] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Indexed: 01/19/2023] Open
Abstract
The relationship between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) dose, infection, and coronavirus disease 2019 (COVID-19) outcomes remains poorly understood. This review summarizes the existing literature regarding this issue, identifies gaps in current knowledge, and suggests opportunities for future research. In humans, host characteristics, including age, sex, comorbidities, smoking, and pregnancy, are associated with severe COVID-19. Similarly, in animals, host factors are strong determinants of disease severity, although most animal infection models manifest clinically with mild to moderate respiratory disease. The influence of variants of concern as it relates to infectious dose, consequence of overall pathogenicity, and disease outcome in dose-response remains unknown. Epidemiologic data suggest a dose-response relationship for infection contrasting with limited and inconsistent surrogate-based evidence between dose and disease severity. Recommendations include the design of future infection studies in animal models to investigate inoculating dose on outcomes and the use of better proxies for dose in human epidemiology studies.
Collapse
Affiliation(s)
- Lisa M Brosseau
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kevin Escandón
- School of Medicine, Universidad del Valle, Cali, Colombia
- Grupo de Investigación en Virus Emergentes y Enfermedad (VIREM), Department of Microbiology, Universidad del Valle, Cali, Colombia
| | - Angela K Ulrich
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela L Rasmussen
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Georgetown Center for Global Health Science and Security, Washington, D.C., USA
| | - Chad J Roy
- Tulane National Primate Research Center, Division of Microbiology, Covington, Louisiana, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Gregory J Bix
- Clinical Neuroscience Research Center, Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana, USA
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USAand
| | - Saskia V Popescu
- Georgetown Center for Global Health Science and Security, Washington, D.C., USA
- Biodefense Program, Schar School of Policy and Government, George Mason University, Arlington, Virginia, USA
| | - Kristine A Moore
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael T Osterholm
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
31
|
Labarile N, Castellana F, Sila A, Pesole PL, Coletta S, Curlo M, Sardone R, Giannelli G, Mastronardi M. Effects of Different Biological Therapies on S1/S2 Antibody Response to SARS-CoV-2 Vaccination in a Cohort of Patients with Inflammatory Bowel Disease. Vaccines (Basel) 2022; 10:vaccines10071077. [PMID: 35891241 PMCID: PMC9322472 DOI: 10.3390/vaccines10071077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 11/23/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has affected the entire planet. The objectives of our study were to compare responses to the vaccine (Pfizer-Biontech COMIRNATY) in a population of patients with intestinal bowel syndrome undergoing different biological therapies or conventional therapy. The study recruited 390 patients who received the first vaccination dose during the dedicated vaccination campaign for inflammatory bowel disease (IBD) patients. The inclusion criteria were a diagnosis of CD or UC and complete vaccination with the Pfizer–BioNTech COVID-19 (Comirnaty) vaccine. The exclusion criteria were other significant diseases or important therapies under way or contraindications to vaccination according to the European drug surveillance recommendations. Linear rank models were run to assess the association between the different therapies and S1/S2 antibodies at three different times. The models showed that in patients with IBD receiving Vedolizumab a significant increase in mean IgG levels was observed, independently of other therapies and confounding factors (β: 57.45, 95% CI 19.62 to 19.00). This study confirmed the complete antibody response to vaccination against COVID-19 in patients with IBD undergoing biological therapy—particularly Vedolizumab treatment—but also a reduced immune response due to concomitant steroid therapy.
Collapse
Affiliation(s)
- Nunzia Labarile
- Unit of Gastroenterology, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy;
| | - Fabio Castellana
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (F.C.); (A.S.); (R.S.)
| | - Annamaria Sila
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (F.C.); (A.S.); (R.S.)
| | - Pasqua Letizia Pesole
- Biobank Core Facilities, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (P.L.P.); (S.C.)
| | - Sergio Coletta
- Biobank Core Facilities, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (P.L.P.); (S.C.)
| | - Margherita Curlo
- Inflammatory Bowel Disease Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (M.C.); (M.M.)
| | - Rodolfo Sardone
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (F.C.); (A.S.); (R.S.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy
- Correspondence:
| | - Mauro Mastronardi
- Inflammatory Bowel Disease Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (M.C.); (M.M.)
| |
Collapse
|
32
|
Fears AC, Beddingfield BJ, Chirichella NR, Slisarenko N, Killeen SZ, Redmann RK, Goff K, Spencer S, Picou B, Golden N, Midkiff CC, Bush DJ, Branco LM, Boisen ML, Gao H, Montefiori DC, Blair RV, Doyle-Meyers LA, Russell-Lodrigue K, Maness NJ, Roy CJ. Exposure modality influences viral kinetics but not respiratory outcome of COVID-19 in multiple nonhuman primate species. PLoS Pathog 2022; 18:e1010618. [PMID: 35789343 PMCID: PMC9286241 DOI: 10.1371/journal.ppat.1010618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/15/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
The novel coronavirus SARS-CoV-2 emerged in late 2019, rapidly reached pandemic status, and has maintained global ubiquity through the emergence of variants of concern. Efforts to develop animal models have mostly fallen short of recapitulating severe disease, diminishing their utility for research focusing on severe disease pathogenesis and life-saving medical countermeasures. We tested whether route of experimental infection substantially changes COVID-19 disease characteristics in two species of nonhuman primates (Macaca mulatta; rhesus macaques; RM, Chlorocebus atheiops; African green monkeys; AGM). Species-specific cohorts were experimentally infected with SARS-CoV-2 by either direct mucosal (intratracheal + intranasal) instillation or small particle aerosol in route-discrete subcohorts. Both species demonstrated analogous viral loads in all compartments by either exposure route although the magnitude and duration of viral loading was marginally greater in AGMs than RMs. Clinical onset was nearly immediate (+1dpi) in the mucosal exposure cohort whereas clinical signs and cytokine responses in aerosol exposure animals began +7dpi. Pathologies conserved in both species and both exposure modalities include pulmonary myeloid cell influx, development of pleuritis, and extended lack of regenerative capacity in the pulmonary compartment. Demonstration of conserved pulmonary pathology regardless of species and exposure route expands our understanding of how SARS-CoV-2 infection may lead to ARDS and/or functional lung damage and demonstrates the near clinical response of the nonhuman primate model for anti-fibrotic therapeutic evaluation studies.
Collapse
Affiliation(s)
- Alyssa C. Fears
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Biomedical Science Training Program, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | | | - Nicole R. Chirichella
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Nadia Slisarenko
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Stephanie Z. Killeen
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Rachel K. Redmann
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Kelly Goff
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Skye Spencer
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Breanna Picou
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Cecily C. Midkiff
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Duane J. Bush
- Zalgen Labs, LLC, Germantown, Maryland, United States of America
| | - Luis M. Branco
- Zalgen Labs, LLC, Germantown, Maryland, United States of America
| | | | - Hongmei Gao
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - David C. Montefiori
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Robert V. Blair
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Lara A. Doyle-Meyers
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Kasi Russell-Lodrigue
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Chad J. Roy
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
33
|
Calvert BA, Quiroz EJ, Lorenzana Z, Doan N, Kim S, Senger CN, Wallace WD, Salomon MP, Henley J, Ryan AL. Neutrophilic inflammation promotes SARS-CoV-2 infectivity and augments the inflammatory responses in airway epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.08.09.455472. [PMID: 34401877 PMCID: PMC8366793 DOI: 10.1101/2021.08.09.455472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In response to viral infection, neutrophils release inflammatory mediators as part of the innate immune response, contributing to pathogen clearance through virus internalization and killing. Pre-existing co- morbidities correlating to incidence of severe COVID-19 are associated with chronic airway neutrophilia. Furthermore, examination of COVID-19 explanted lung tissue revealed a series of epithelial pathologies associated with the infiltration and activation of neutrophils, indicating neutrophil activity in response to SARS- CoV-2 infection. To determine the impact of neutrophil-epithelial interactions on the infectivity and inflammatory responses to SARS-CoV-2 infection, we developed a co-culture model of airway neutrophilia. SARS-CoV-2 infection of the airway epithelium alone does not result in a notable pro-inflammatory response from the epithelium. The addition of neutrophils induces the release of proinflammatory cytokines and stimulates a significantly augmented pro-inflammatory response subsequent SARS-CoV-2 infection. The resulting inflammatory response is polarized with differential release from the apical and basolateral side of the epithelium. Additionally, the integrity of the epithelial barrier is impaired with notable epithelial damage and infection of basal stem cells. This study reveals a key role for neutrophil-epithelial interactions in determining inflammation and infectivity in response to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- BA Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - EJ Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Z Lorenzana
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - N Doan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - S Kim
- The Salk Institute of Biological Studies, 10010 North Torey Pines Road, La Jolla, Ca, USA
| | - CN Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - WD Wallace
- Department of Pathology, University of Southern California, Los Angeles, CA, USA
| | - MP Salomon
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - J Henley
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - AL Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
34
|
Madden PJ, Thomas Y, Blair RV, Samer S, Doyle M, Midkiff CC, Doyle-Meyers LA, Becker ME, Arif MS, McRaven MD, Simons LM, Carias AM, Martinelli E, Lorenzo-Redondo R, Hultquist JF, Villinger FJ, Veazey RS, Hope TJ. An immunoPET probe to SARS-CoV-2 reveals early infection of the male genital tract in rhesus macaques. RESEARCH SQUARE 2022:rs.3.rs-1479315. [PMID: 35411346 PMCID: PMC8996619 DOI: 10.21203/rs.3.rs-1479315/v1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The systemic nature of SARS-CoV-2 infection is highly recognized, but poorly characterized. A non-invasive and unbiased method is needed to clarify whole body spatiotemporal dynamics of SARS-CoV-2 infection after transmission. We recently developed a probe based on the anti-SARS-CoV-2 spike antibody CR3022 to study SARS-CoV-2 pathogenesis in vivo. Herein, we describe its use in immunoPET to investigate SARS-CoV-2 infection of three rhesus macaques. Using PET/CT imaging of macaques at different times post-SARS-CoV-2 inoculation, we track the 64Cu-labelled CR3022-F(ab')2 probe targeting the spike protein of SARS-CoV-2 to study the dynamics of infection within the respiratory tract and uncover novel sites of infection. Using this method, we uncovered differences in lung pathology between infection with the WA1 isolate and the delta variant, which were readily corroborated through computed tomography scans. The 64Cu-CR3022-probe also demonstrated dynamic changes occurring between 1- and 2-weeks post-infection. Remarkably, a robust signal was seen in the male genital tract (MGT) of all three animals studied. Infection of the MGT was validated by immunofluorescence imaging of infected cells in the testicular and penile tissue and severe pathology was observed in the testes of one animal at 2-weeks post-infection. The results presented here underscore the utility of using immunoPET to study the dynamics of SARS-CoV-2 infection to understand its pathogenicity and discover new anatomical sites of viral replication. We provide direct evidence for SARS-CoV-2 infection of the MGT in rhesus macaques revealing the possible pathologic outcomes of viral replication at these sites.
Collapse
Affiliation(s)
- Patrick J Madden
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yanique Thomas
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Robert V Blair
- Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Sadia Samer
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mark Doyle
- Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Cecily C Midkiff
- Tulane National Primate Research Center, Covington, Louisiana, USA
| | | | - Mark E Becker
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Muhammad S Arif
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michael D McRaven
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Lacy M Simons
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ann M Carias
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elena Martinelli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Judd F Hultquist
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francois J Villinger
- New Iberia Research Center, University of Louisiana-Lafayette, New Iberia, Louisiana, USA
| | - Ronald S Veazey
- Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Thomas J Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
35
|
To A, Wong TAS, Lieberman MM, Thompson K, Ball AH, Pessaint L, Greenhouse J, Daham N, Cook A, Narvaez B, Flinchbaugh Z, Van Ry A, Yalley-Ogunro J, Elyard HA, Lai CY, Donini O, Lehrer AT. A Recombinant Subunit Vaccine Induces a Potent, Broadly Neutralizing, and Durable Antibody Response in Macaques against the SARS-CoV-2 P.1 (Gamma) Variant. ACS Infect Dis 2022; 8:825-840. [PMID: 35263081 PMCID: PMC8938837 DOI: 10.1021/acsinfecdis.1c00600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Indexed: 12/12/2022]
Abstract
FDA-approved and emergency use-authorized vaccines using new mRNA and viral-vector technology are highly effective in preventing moderate to severe disease; however, information on their long-term efficacy and protective breadth against severe acute respiratory syndrome coronavirus 2 variants of concern (VOCs) is currently scarce. Here, we describe the durability and broad-spectrum VOC immunity of a prefusion-stabilized spike (S) protein adjuvanted with liquid or lyophilized CoVaccine HT in cynomolgus macaques. This recombinant subunit vaccine is highly immunogenic and induces robust spike-specific and broadly neutralizing antibody responses effective against circulating VOCs (B.1.351 [Beta], P.1 [Gamma], and B.1.617 [Delta]) for at least three months after the final boost. Protective efficacy and postexposure immunity were evaluated using a heterologous P.1 challenge nearly three months after the last immunization. Our results indicate that while immunization with both high and low S doses shorten and reduce viral loads in the upper and lower respiratory tract, a higher antigen dose is required to provide durable protection against disease as vaccine immunity wanes. Histologically, P.1 infection causes similar COVID-19-like lung pathology as seen with early pandemic isolates. Postchallenge IgG concentrations were restored to peak immunity levels, and vaccine-matched and cross-variant neutralizing antibodies were significantly elevated in immunized macaques indicating an efficient anamnestic response. Only low levels of P.1-specific neutralizing antibodies with limited breadth were observed in control (nonvaccinated but challenged) macaques, suggesting that natural infection may not prevent reinfection by other VOCs. Overall, these results demonstrate that a properly dosed and adjuvanted recombinant subunit vaccine can provide protective immunity against circulating VOCs for at least three months.
Collapse
Affiliation(s)
- Albert To
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Teri Ann S. Wong
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Michael M. Lieberman
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Karen Thompson
- Department of Pathology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Aquena H. Ball
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | | | - Jack Greenhouse
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | | | - Anthony Cook
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Brandon Narvaez
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Zack Flinchbaugh
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Alex Van Ry
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Jake Yalley-Ogunro
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Hanne Andersen Elyard
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | - Chih-Yun Lai
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| | | | - Axel T. Lehrer
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI, 96813, USA
| |
Collapse
|
36
|
Madden PJ, Thomas Y, Blair RV, Samer S, Doyle M, Midkiff CC, Doyle-Meyers LA, Becker ME, Arif MS, McRaven MD, Simons LM, Carias AM, Martinelli E, Lorenzo-Redondo R, Hultquist JF, Villinger FJ, Veazey RS, Hope TJ. An immunoPET probe to SARS-CoV-2 reveals early infection of the male genital tract in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.02.25.481974. [PMID: 35262081 PMCID: PMC8902882 DOI: 10.1101/2022.02.25.481974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The systemic nature of SARS-CoV-2 infection is highly recognized, but poorly characterized. A non-invasive and unbiased method is needed to clarify whole body spatiotemporal dynamics of SARS-CoV-2 infection after transmission. We recently developed a probe based on the anti-SARS-CoV-2 spike antibody CR3022 to study SARS-CoV-2 pathogenesis in vivo. Herein, we describe its use in immunoPET to investigate SARS-CoV-2 infection of three rhesus macaques. Using PET/CT imaging of macaques at different times post-SARS-CoV-2 inoculation, we track the 64Cu-labelled CR3022-F(ab')2 probe targeting the spike protein of SARS-CoV-2 to study the dynamics of infection within the respiratory tract and uncover novel sites of infection. Using this method, we uncovered differences in lung pathology between infection with the WA1 isolate and the delta variant, which were readily corroborated through computed tomography scans. The 64Cu-CR3022-probe also demonstrated dynamic changes occurring between 1- and 2-weeks post-infection. Remarkably, a robust signal was seen in the male genital tract (MGT) of all three animals studied. Infection of the MGT was validated by immunofluorescence imaging of infected cells in the testicular and penile tissue and severe pathology was observed in the testes of one animal at 2-weeks post-infection. The results presented here underscore the utility of using immunoPET to study the dynamics of SARS-CoV-2 infection to understand its pathogenicity and discover new anatomical sites of viral replication. We provide direct evidence for SARS-CoV-2 infection of the MGT in rhesus macaques revealing the possible pathologic outcomes of viral replication at these sites.
Collapse
Affiliation(s)
- Patrick J Madden
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yanique Thomas
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Robert V Blair
- Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Sadia Samer
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mark Doyle
- Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Cecily C Midkiff
- Tulane National Primate Research Center, Covington, Louisiana, USA
| | | | - Mark E Becker
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Muhammad S Arif
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michael D McRaven
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Lacy M Simons
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ann M Carias
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elena Martinelli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Judd F Hultquist
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francois J Villinger
- New Iberia Research Center, University of Louisiana-Lafayette, New Iberia, Louisiana, USA
| | - Ronald S Veazey
- Tulane National Primate Research Center, Covington, Louisiana, USA
| | - Thomas J Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
37
|
Birindelli S, Tarkowski MS, Gallucci M, Schiuma M, Covizzi A, Lewkowicz P, Aloisio E, Falvella FS, Dolci A, Riva A, Galli M, Panteghini M. Definition of the Immune Parameters Related to COVID-19 Severity. Front Immunol 2022; 13:850846. [PMID: 35371011 PMCID: PMC8971756 DOI: 10.3389/fimmu.2022.850846] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/23/2022] [Indexed: 01/08/2023] Open
Abstract
A relevant portion of patients with disease caused by the severe acute respiratory syndrome coronavirus 2 (COVID-19) experience negative outcome, and several laboratory tests have been proposed to predict disease severity. Among others, dramatic changes in peripheral blood cells have been described. We developed and validated a laboratory score solely based on blood cell parameters to predict survival in hospitalized COVID-19 patients. We retrospectively analyzed 1,619 blood cell count from 226 consecutively hospitalized COVID-19 patients to select parameters for inclusion in a laboratory score predicting severity of disease and survival. The score was derived from lymphocyte- and granulocyte-associated parameters and validated on a separate cohort of 140 consecutive COVID-19 patients. Using ROC curve analysis, a best cutoff for score of 30.6 was derived, which was associated to an overall 82.0% sensitivity (95% CI: 78–84) and 82.5% specificity (95% CI: 80–84) for detecting outcome. The scoring trend effectively separated survivor and non-survivor groups, starting 2 weeks before the end of the hospitalization period. Patients’ score time points were also classified into mild, moderate, severe, and critical according to the symptomatic oxygen therapy administered. Fluctuations of the score should be recorded to highlight a favorable or unfortunate trend of the disease. The predictive score was found to reflect and anticipate the disease gravity, defined by the type of the oxygen support used, giving a proof of its clinical relevance. It offers a fast and reliable tool for supporting clinical decisions and, most important, triage in terms of not only prioritization but also allocation of limited medical resources, especially in the period when therapies are still symptomatic and many are under development. In fact, a prolonged and progressive increase of the score can suggest impaired chances of survival and/or an urgent need for intensive care unit admission.
Collapse
Affiliation(s)
- Sarah Birindelli
- Clinical Pathology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
- *Correspondence: Sarah Birindelli,
| | - Maciej S. Tarkowski
- Department of Biomedical and Clinical Sciences, “Luigi Sacco”, University of Milan, Milan, Italy
| | - Marcello Gallucci
- Department of Psychology, University of Milano Bicocca, Milan, Italy
| | - Marco Schiuma
- Department of Infectious Diseases, Division III, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Alice Covizzi
- Department of Infectious Diseases, Division III, ASST Fatebenefratelli-Sacco, Milan, Italy
| | | | - Elena Aloisio
- Clinical Pathology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | | | - Alberto Dolci
- Clinical Pathology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
- Department of Biomedical and Clinical Sciences, “Luigi Sacco”, University of Milan, Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, “Luigi Sacco”, University of Milan, Milan, Italy
- Department of Infectious Diseases, Division III, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Massimo Galli
- Department of Biomedical and Clinical Sciences, “Luigi Sacco”, University of Milan, Milan, Italy
- Department of Infectious Diseases, Division III, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Mauro Panteghini
- Clinical Pathology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
- Department of Biomedical and Clinical Sciences, “Luigi Sacco”, University of Milan, Milan, Italy
| |
Collapse
|
38
|
Nelson CE, Namasivayam S, Foreman TW, Kauffman KD, Sakai S, Dorosky DE, Lora NE, Brooks K, Potter EL, Garza NL, Lafont BAP, Johnson RF, Roederer M, Sher A, Weiskopf D, Sette A, de Wit E, Hickman HD, Brenchley JM, Via LE, Barber DL. Mild SARS-CoV-2 infection in rhesus macaques is associated with viral control prior to antigen-specific T cell responses in tissues. Sci Immunol 2022; 7:eabo0535. [PMID: 35271298 PMCID: PMC8995035 DOI: 10.1126/sciimmunol.abo0535] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/04/2022] [Indexed: 12/24/2022]
Abstract
SARS-CoV-2 primarily replicates in mucosal sites, and more information is needed about immune responses in infected tissues. Here, we used rhesus macaques to model protective primary immune responses in tissues during mild COVID-19. Viral RNA levels were highest on days 1-2 post-infection and fell precipitously thereafter. 18F-fluorodeoxyglucose (FDG)-avid lung abnormalities and interferon (IFN)-activated monocytes and macrophages in the bronchoalveolar lavage (BAL) were found on days 3-4 post-infection. Virus-specific effector CD8+ and CD4+ T cells became detectable in the BAL and lung tissue on days 7-10, after viral RNA, radiologic evidence of lung inflammation, and IFN-activated myeloid cells had substantially declined. Notably, SARS-CoV-2-specific T cells were not detectable in the nasal turbinates, salivary glands, and tonsils on day 10 post-infection. Thus, SARS-CoV-2 replication wanes in the lungs of rhesus macaques prior to T cell responses, and in the nasal and oral mucosa despite the apparent lack of antigen-specific T cells, suggesting that innate immunity efficiently restricts viral replication during mild COVID-19.
Collapse
Affiliation(s)
- Christine E. Nelson
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Sivaranjani Namasivayam
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Taylor W. Foreman
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Keith D. Kauffman
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Shunsuke Sakai
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Danielle E. Dorosky
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Nickiana E. Lora
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - NIAID/DIR Tuberculosis Imaging Program3†
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- Laboratory of Virology, Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, USA
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Kelsie Brooks
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - E. Lake Potter
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Nicole L. Garza
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Bernard A. P. Lafont
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Reed F. Johnson
- SARS-CoV-2 Virology Core, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Hamilton, MT, USA
| | - Heather D. Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Jason M. Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Laura E. Via
- Tuberculosis Research Section, Laboratory of Clinical Infectious Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Institute of Infectious Disease & Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Observatory, South Africa
| | - Daniel L. Barber
- T lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
39
|
Rabbani MY, Rappaport J, Gupta MK. Activation of Immune System May Cause Pathophysiological Changes in the Myocardium of SARS-CoV-2 Infected Monkey Model. Cells 2022; 11:611. [PMID: 35203260 PMCID: PMC8869860 DOI: 10.3390/cells11040611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/03/2022] [Accepted: 02/06/2022] [Indexed: 02/01/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is an extremely contagious disease whereby the virus damages the host's respiratory tract via entering through the ACE2 receptor. Cardiovascular disorder is being recognized in the majority of COVID-19 patients; yet, the relationship between SARS-CoV-2 and heart failure has not been established. In the present study, SARS-CoV-2 infection was induced in the monkey model. Thereafter, heart tissue samples were collected, and pathological changes were analyzed in the left ventricular tissue by hematoxylin and eosin, trichrome, and immunohistochemical staining specific to T lymphocytes and macrophages. The findings revealed that SARS-CoV-2 infection induces several pathological changes in the heart, which cause cardiomyocyte disarray, mononuclear infiltrates of inflammatory cells, and hypertrophy. Furthermore, collagen-specific staining showed the development of cardiac fibrosis in the interstitial and perivascular regions in the hearts of infected primates. Moreover, the myocardial tissue samples displayed multiple foci of inflammatory cells positive for T lymphocytes and macrophages within the myocardium. These findings suggest the progression of the disease, which can lead to the development of severe complications, including heart failure. Additionally, SARS-CoV-2 antigen staining detected the presence of virus particles in the myocardium. Thus, we found that SARS-CoV-2 infection is characterized by an exaggerated inflammatory immune response in the heart, which possibly contributes to myocardial remodeling and subsequent fibrosis.
Collapse
Affiliation(s)
- Maryam Yahya Rabbani
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| | - Jay Rappaport
- Division of Pathology, Tulane National Primate Research Center, Covington, LA 70433, USA;
| | - Manish Kumar Gupta
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA;
| |
Collapse
|
40
|
Pillet S, Arunachalam PS, Andreani G, Golden N, Fontenot J, Aye PP, Röltgen K, Lehmicke G, Gobeil P, Dubé C, Trépanier S, Charland N, D'Aoust MA, Russell-Lodrigue K, Monjure C, Blair RV, Boyd SD, Bohm RP, Rappaport J, Villinger F, Landry N, Pulendran B, Ward BJ. Safety, immunogenicity, and protection provided by unadjuvanted and adjuvanted formulations of a recombinant plant-derived virus-like particle vaccine candidate for COVID-19 in nonhuman primates. Cell Mol Immunol 2022; 19:222-233. [PMID: 34983950 PMCID: PMC8727235 DOI: 10.1038/s41423-021-00809-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022] Open
Abstract
Although antivirals are important tools to control severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, effective vaccines are essential to control the current coronavirus disease 2019 (COVID-19) pandemic. Plant-derived virus-like particle (VLP) vaccine candidates have previously demonstrated immunogenicity and efficacy against influenza. Here, we report the immunogenicity and protection induced in rhesus macaques by intramuscular injections of a VLP bearing a SARS-CoV-2 spike protein (CoVLP) vaccine candidate formulated with or without Adjuvant System 03 (AS03) or cytidine-phospho-guanosine (CpG) 1018. Although a single dose of the unadjuvanted CoVLP vaccine candidate stimulated humoral and cell-mediated immune responses, booster immunization (at 28 days after priming) and adjuvant administration significantly improved both responses, with higher immunogenicity and protection provided by the AS03-adjuvanted CoVLP. Fifteen micrograms of CoVLP adjuvanted with AS03 induced a polyfunctional interleukin-2 (IL-2)-driven response and IL-4 expression in CD4 T cells. Animals were challenged by multiple routes (i.e., intratracheal, intranasal, and ocular) with a total viral dose of 106 plaque-forming units of SARS-CoV-2. Lower viral replication in nasal swabs and bronchoalveolar lavage fluid (BALF) as well as fewer SARS-CoV-2-infected cells and immune cell infiltrates in the lungs concomitant with reduced levels of proinflammatory cytokines and chemotactic factors in the BALF were observed in animals immunized with the CoVLP adjuvanted with AS03. No clinical, pathologic, or virologic evidence of vaccine-associated enhanced disease was observed in vaccinated animals. The CoVLP adjuvanted with AS03 was therefore selected for vaccine development and clinical trials.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/adverse effects
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- COVID-19/epidemiology
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/adverse effects
- Disease Models, Animal
- Drug Combinations
- Drug Compounding/methods
- Immunity, Humoral
- Immunogenicity, Vaccine/immunology
- Macaca mulatta
- Male
- Pandemics/prevention & control
- Polysorbates/administration & dosage
- Polysorbates/adverse effects
- Recombinant Proteins/immunology
- Recombinant Proteins/metabolism
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/metabolism
- Squalene/administration & dosage
- Squalene/adverse effects
- Nicotiana/metabolism
- Treatment Outcome
- Vaccination/methods
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/adverse effects
- alpha-Tocopherol/administration & dosage
- alpha-Tocopherol/adverse effects
Collapse
Affiliation(s)
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Jane Fontenot
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | | | - Katharina Röltgen
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | - Robert V Blair
- Tulane National Primate Research Center, Covington, LA, USA
| | - Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Rudolf P Bohm
- Tulane National Primate Research Center, Covington, LA, USA
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - François Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA, USA
| | | | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Brian J Ward
- Medicago Inc., Québec, QC, Canada.
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
41
|
Puthia M, Tanner L, Petruk G, Schmidtchen A. Experimental Model of Pulmonary Inflammation Induced by SARS-CoV-2 Spike Protein and Endotoxin. ACS Pharmacol Transl Sci 2022; 5:141-148. [PMID: 35774232 PMCID: PMC9239546 DOI: 10.1021/acsptsci.1c00219] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Indexed: 01/27/2023]
Abstract
COVID-19 is characterized by a dysregulated and excessive inflammatory response and, in severe cases, acute respiratory distress syndrome. We have recently demonstrated a previously unknown high-affinity interaction between the SARS-CoV-2 spike (S) protein and bacterial lipopolysaccharide (LPS), leading to the boosting of inflammation. Here we present a mouse inflammation model employing the coadministration of aerosolized S protein together with LPS to the lungs. Using NF-κB-RE-Luc reporter and C57BL/6 mice followed by combinations of bioimaging, cytokine, chemokine, fluorescence-activated cell sorting, and histochemistry analyses, we show that the model yields severe pulmonary inflammation and a cytokine profile similar to that observed in COVID-19. Therefore, the model offers utility for analyses of the pathophysiological features of COVID-19 and the development of new treatments.
Collapse
Affiliation(s)
- Manoj Puthia
- Division
of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden,
| | - Lloyd Tanner
- Division
of Respiratory Medicine and Allergology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Ganna Petruk
- Division
of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Artur Schmidtchen
- Division
of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden,Bispebjerg
Hospital, Department of Biomedical Sciences, University of Copenhagen, DK-2400 Copenhagen, Denmark
| |
Collapse
|
42
|
Grau-Expósito J, Perea D, Suppi M, Massana N, Vergara A, Soler MJ, Trinite B, Blanco J, García-Pérez J, Alcamí J, Serrano-Mollar A, Rosado J, Falcó V, Genescà M, Buzon MJ. Evaluation of SARS-CoV-2 entry, inflammation and new therapeutics in human lung tissue cells. PLoS Pathog 2022; 18:e1010171. [PMID: 35025963 PMCID: PMC8791477 DOI: 10.1371/journal.ppat.1010171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/26/2022] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
The development of physiological models that reproduce SARS-CoV-2 infection in primary human cells will be instrumental to identify host-pathogen interactions and potential therapeutics. Here, using cell suspensions directly from primary human lung tissues (HLT), we have developed a rapid platform for the identification of viral targets and the expression of viral entry factors, as well as for the screening of viral entry inhibitors and anti-inflammatory compounds. The direct use of HLT cells, without long-term cell culture and in vitro differentiation approaches, preserves main immune and structural cell populations, including the most susceptible cell targets for SARS-CoV-2; alveolar type II (AT-II) cells, while maintaining the expression of proteins involved in viral infection, such as ACE2, TMPRSS2, CD147 and AXL. Further, antiviral testing of 39 drug candidates reveals a highly reproducible method, suitable for different SARS-CoV-2 variants, and provides the identification of new compounds missed by conventional systems, such as VeroE6. Using this method, we also show that interferons do not modulate ACE2 expression, and that stimulation of local inflammatory responses can be modulated by different compounds with antiviral activity. Overall, we present a relevant and rapid method for the study of SARS-CoV-2.
Collapse
Affiliation(s)
- Judith Grau-Expósito
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - David Perea
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Marina Suppi
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Núria Massana
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Ander Vergara
- Nephrology Research Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Maria José Soler
- Nephrology Research Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Benjamin Trinite
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Autonomous University of Barcelona (UAB), Badalona, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Autonomous University of Barcelona (UAB), Badalona, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Javier García-Pérez
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José Alcamí
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Clinic HIV Unit, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Anna Serrano-Mollar
- Experimental Pathology Department, Institut d’Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Joel Rosado
- Thoracic Surgery and Lung Transplantation Department, Vall d’Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d’Hebron, VHIR Task Force COVID-19, Barcelona, Spain
| | - Vicenç Falcó
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Meritxell Genescà
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| | - Maria J. Buzon
- Infectious Diseases Department, Vall d’Hebron Research Institute (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, VHIR Task Force COVID-19, Barcelona, Spain
| |
Collapse
|
43
|
Awad K, Maghraby AS, Abd-Elshafy DN, Bahgat MM. Carbohydrates Metabolic Signatures in Immune Cells: Response to Infection. Front Immunol 2022; 13:912899. [PMID: 35983037 PMCID: PMC9380592 DOI: 10.3389/fimmu.2022.912899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/01/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Metabolic reprogramming in immune cells is diverse and distinctive in terms of complexity and flexibility in response to heterogeneous pathogenic stimuli. We studied the carbohydrate metabolic changes in immune cells in different types of infectious diseases. This could help build reasonable strategies when understanding the diagnostics, prognostics, and biological relevance of immune cells under alternative metabolic burdens. METHODS Search and analysis were conducted on published peer-reviewed papers on immune cell metabolism of a single pathogen infection from the four known types (bacteria, fungi, parasites, and viruses). Out of the 131 selected papers based on the PIC algorithm (pathogen type/immune cell/carbohydrate metabolism), 30 explored immune cell metabolic changes in well-studied bacterial infections, 17 were on fungal infections of known medical importance, and 12 and 57 were on parasitic and viral infections, respectively. RESULTS AND DISCUSSION While carbohydrate metabolism in immune cells is signaled by glycolytic shift during a bacterial or viral infection, it is widely evident that effector surface proteins are expressed on the surface of parasites and fungi to modulate metabolism in these cells. CONCLUSIONS Carbohydrate metabolism in immune cells can be categorized according to the pathogen or the disease type. Accordingly, this classification can be used to adopt new strategies in disease diagnosis and treatment.
Collapse
Affiliation(s)
- Kareem Awad
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Cairo, Egypt
- *Correspondence: Kareem Awad, ; Mahmoud Mohamed Bahgat, ,
| | - Amany Sayed Maghraby
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Cairo, Egypt
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt
| | - Dina Nadeem Abd-Elshafy
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt
- Department of Water Pollution Research, Institute of Environmental Research, National Research Center, Cairo, Egypt
| | - Mahmoud Mohamed Bahgat
- Department of Therapeutic Chemistry, Institute of Pharmaceutical and Drug Industries Research, National Research Center, Cairo, Egypt
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt
- *Correspondence: Kareem Awad, ; Mahmoud Mohamed Bahgat, ,
| |
Collapse
|
44
|
Veenhuis RT, Zeiss CJ. Animal Models of COVID-19 II. Comparative Immunology. ILAR J 2021; 62:17-34. [PMID: 33914873 PMCID: PMC8135340 DOI: 10.1093/ilar/ilab010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/03/2020] [Accepted: 12/20/2020] [Indexed: 12/22/2022] Open
Abstract
Developing strong animal models is essential for furthering our understanding of how the immune system functions in response to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. The alarming speed at which SARS-CoV-2 has spread, and the high mortality rate of severe Coronavirus Disease 2019 (COVID-19), has required both basic science and clinical research to move at an unprecedented pace. Models previously developed to study the immune response against SARS-CoV have been rapidly deployed to now study SARS-CoV-2. To date, both small and large animal models are remarkably consistent when infected with SARS-CoV-2; however, certain models have proven more useful when answering specific immunological questions than others. Small animal models, such as Syrian hamsters, ferrets, and mice carrying the hACE2 transgene, appear to reliably recapitulate the initial cytokine surge seen in COVID-19 as well as show significant innate and adaptive cell infiltration in to the lung early in infection. Additionally, these models develop strong antibody responses to the virus, are protected from reinfection, and genetically modified versions exist that can be used to ask specific immunological questions. Large animal models such as rhesus and cynomologus macaques and African green monkeys are critical to understanding how the immune system responds to SARS-CoV-2 infection because they are considered to be the most similar to humans. These models are considered the gold standard for assessing vaccine efficacy and protection, and recapitulate the initial cytokine surge, immune cell infiltration into the lung, certain aspects of thrombosis, and the antibody and T-cell response to the virus. In this review, we discuss both small and large animal model studies previously used in SARS-CoV-2 research that may be useful in elucidating the immunological contributions to hallmark syndromes observed with COVID-19.
Collapse
Affiliation(s)
- Rebecca T Veenhuis
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Caroline J Zeiss
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
45
|
Patania OM, Chiba S, Halfmann PJ, Hatta M, Maemura T, Bernard KA, Kawaoka Y, Crawford LK. Pulmonary lesions induced by SARS-CoV-2 infection in domestic cats. Vet Pathol 2021; 59:696-706. [PMID: 34963403 PMCID: PMC9208068 DOI: 10.1177/03009858211066840] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of coronavirus disease 2019, which ranges from fatal disease in some to mild or subclinical in most affected individuals. Many recovered human patients report persistent respiratory signs; however, lung disease in post-acute infection is poorly understood. Our objective was to describe histologic lung lesions and viral loads following experimental SARS-CoV-2 infection in 11 cats. Microscopic evaluation at 3, 6, 10, or 28 days postinoculation (DPI) identified mild to moderate patchy interstitial pneumonia, bronchiolar epithelial damage, and occlusive histiocytic bronchiolitis. Based on immunohistochemistry, alveolar septal thickening was due to CD204-positive macrophages, fewer B and T lymphocytes, type II pneumocytes, and capillary proliferation with a relative dearth of fibrosis. In blood vessel endothelium, there was reactive hypertrophy or vacuolar degeneration and increased MHC II expression at all time points. Unexpectedly, one cat from the 28 DPI group had severe subacute regionally extensive lymphohistiocytic pneumonia with multifocal consolidation, vasculitis, and alveolar fibrin. Reverse transcriptase-quantitative polymerase chain reaction identified SARS-CoV-2 RNA within the lung at 3 and 6 DPI, and viral RNA was below the limit of detection at 10 and 28 DPI, suggesting that pulmonary lesions persist beyond detection of viral RNA. These findings clarify our comparative understanding of disease induced by SARS-CoV-2 and suggest that cats can serve as an informative model to study post-acute pulmonary sequelae.
Collapse
Affiliation(s)
| | - Shiho Chiba
- University of Wisconsin-Madison, Madison, WI, USA
| | | | - Masato Hatta
- University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Yoshihiro Kawaoka
- University of Wisconsin-Madison, Madison, WI, USA.,University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
46
|
Madden PJ, Arif MS, Becker ME, McRaven MD, Carias AM, Lorenzo-Redondo R, Xiao S, Midkiff CC, Blair RV, Potter EL, Martin-Sancho L, Dodson A, Martinelli E, Todd JPM, Villinger FJ, Chanda SK, Aye PP, Roy CJ, Roederer M, Lewis MG, Veazey RS, Hope TJ. Development of an In Vivo Probe to Track SARS-CoV-2 Infection in Rhesus Macaques. Front Immunol 2021; 12:810047. [PMID: 35003140 PMCID: PMC8739270 DOI: 10.3389/fimmu.2021.810047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 01/02/2023] Open
Abstract
Infection with the novel coronavirus, SARS-CoV-2, results in pneumonia and other respiratory symptoms as well as pathologies at diverse anatomical sites. An outstanding question is whether these diverse pathologies are due to replication of the virus in these anatomical compartments and how and when the virus reaches those sites. To answer these outstanding questions and study the spatiotemporal dynamics of SARS-CoV-2 infection a method for tracking viral spread in vivo is needed. We developed a novel, fluorescently labeled, antibody-based in vivo probe system using the anti-spike monoclonal antibody CR3022 and demonstrated that it could successfully identify sites of SARS-CoV-2 infection in a rhesus macaque model of COVID-19. Our results showed that the fluorescent signal from our antibody-based probe could differentiate whole lungs of macaques infected for 9 days from those infected for 2 or 3 days. Additionally, the probe signal corroborated the frequency and density of infected cells in individual tissue blocks from infected macaques. These results provide proof of concept for the use of in vivo antibody-based probes to study SARS-CoV-2 infection dynamics in rhesus macaques.
Collapse
Affiliation(s)
- Patrick J. Madden
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Muhammad S. Arif
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Mark E. Becker
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Michael D. McRaven
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ann M. Carias
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Institute for Global Health, Chicago, IL, United States
| | - Sixia Xiao
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Cecily C. Midkiff
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Robert V. Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Elizabeth Lake Potter
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Laura Martin-Sancho
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | | | - Elena Martinelli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - John-Paul M. Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Francois J. Villinger
- New Iberia Research Center, University of Louisiana-Lafayette, New Iberia, LA, United States
| | - Sumit K. Chanda
- Immunity and Pathogenesis Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Pyone Pyone Aye
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Chad J. Roy
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | - Ronald S. Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, United States
| | - Thomas J. Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
47
|
O’Connor MA, Erasmus JH, Randall S, Archer J, Lewis TB, Brown B, Fredericks M, Groenier S, Iwayama N, Ahrens C, Garrison W, Wangari S, Guerriero KA, Fuller DH. A Single Dose SARS-CoV-2 Replicon RNA Vaccine Induces Cellular and Humoral Immune Responses in Simian Immunodeficiency Virus Infected and Uninfected Pigtail Macaques. Front Immunol 2021; 12:800723. [PMID: 34992610 PMCID: PMC8724308 DOI: 10.3389/fimmu.2021.800723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
The ongoing COVID-19 vaccine rollout is critical for reducing SARS-CoV-2 infections, hospitalizations, and deaths worldwide. Unfortunately, massive disparities exist in getting vaccines to vulnerable populations, including people living with HIV. Preliminary studies indicate that COVID-19 mRNA vaccines are safe and immunogenic in people living with HIV that are virally suppressed with potent antiretroviral therapy but may be less efficacious in immunocompromised individuals. This raises the concern that COVID-19 vaccines may be less effective in resource poor settings with limited access to antiretroviral therapy. Here, we evaluated the immunogenicity of a single dose COVID-19 replicon RNA vaccine expressing Spike protein (A.1) from SARS-CoV-2 (repRNA-CoV2S) in immunocompromised, SIV infected and immune competent, naïve pigtail macaques. Moderate vaccine-specific cellular Th1 T-cell responses and binding and neutralizing antibodies were induced by repRNA-CoV2S in SIV infected animals and naïve animals. Furthermore, vaccine immunogenicity was elicited even among the animals with the highest SIV viral burden or lowest peripheral CD4 counts prior to immunization. This study provides evidence that a SARS-CoV-2 repRNA vaccine could be employed to induce strong immunity against COVID-19 in HIV infected and other immunocompromised individuals.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/virology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- COVID-19 Vaccines/immunology
- Cells, Cultured
- Disease Models, Animal
- Host-Pathogen Interactions
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunocompromised Host
- Immunogenicity, Vaccine
- Macaca nemestrina
- Male
- Simian Acquired Immunodeficiency Syndrome/blood
- Simian Acquired Immunodeficiency Syndrome/immunology
- Simian Acquired Immunodeficiency Syndrome/virology
- Simian Immunodeficiency Virus/immunology
- Simian Immunodeficiency Virus/pathogenicity
- Spike Glycoprotein, Coronavirus/administration & dosage
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/virology
- Time Factors
- Vaccination
- Vaccine Efficacy
- mRNA Vaccines/administration & dosage
- mRNA Vaccines/genetics
- mRNA Vaccines/immunology
Collapse
Affiliation(s)
- Megan A. O’Connor
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Jesse H. Erasmus
- Department of Microbiology, University of Washington, Seattle, WA, United States
- HDT Bio, Seattle, WA, United States
| | - Samantha Randall
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Jacob Archer
- Department of Microbiology, University of Washington, Seattle, WA, United States
- HDT Bio, Seattle, WA, United States
| | - Thomas B. Lewis
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Brieann Brown
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Megan Fredericks
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Skyler Groenier
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Naoto Iwayama
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Chul Ahrens
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - William Garrison
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Solomon Wangari
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Kathryn A. Guerriero
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| |
Collapse
|
48
|
Bi Z, Hong W, Yang J, Lu S, Peng X. Animal models for SARS-CoV-2 infection and pathology. MedComm (Beijing) 2021; 2:548-568. [PMID: 34909757 PMCID: PMC8662225 DOI: 10.1002/mco2.98] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 02/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiology of coronavirus disease 2019 (COVID-19) pandemic. Current variants including Alpha, Beta, Gamma, Delta, and Lambda increase the capacity of infection and transmission of SARS-CoV-2, which might disable the in-used therapies and vaccines. The COVID-19 has now put an enormous strain on health care system all over the world. Therefore, the development of animal models that can capture characteristics and immune responses observed in COVID-19 patients is urgently needed. Appropriate models could accelerate the testing of therapeutic drugs and vaccines against SARS-CoV-2. In this review, we aim to summarize the current animal models for SARS-CoV-2 infection, including mice, hamsters, nonhuman primates, and ferrets, and discuss the details of transmission, pathology, and immunology induced by SARS-CoV-2 in these animal models. We hope this could throw light to the increased usefulness in fundamental studies of COVID-19 and the preclinical analysis of vaccines and therapeutic agents.
Collapse
Affiliation(s)
- Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduSichuanChina
| | - Shuaiyao Lu
- National Kunming High‐level Biosafety Primate Research CenterInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeYunnanChina
| | - Xiaozhong Peng
- National Kunming High‐level Biosafety Primate Research CenterInstitute of Medical BiologyChinese Academy of Medical Sciences and Peking Union Medical CollegeYunnanChina
| |
Collapse
|
49
|
Melton A, Doyle-Meyers LA, Blair RV, Midkiff C, Melton HJ, Russell-Lodrigue K, Aye PP, Schiro F, Fahlberg M, Szeltner D, Spencer S, Beddingfield BJ, Goff K, Golden N, Penney T, Picou B, Hensley K, Chandler KE, Plante JA, Plante KS, Weaver SC, Roy CJ, Hoxie JA, Gao H, Montefiori DC, Mankowski JL, Bohm RP, Rappaport J, Maness NJ. The pigtail macaque (Macaca nemestrina) model of COVID-19 reproduces diverse clinical outcomes and reveals new and complex signatures of disease. PLoS Pathog 2021; 17:e1010162. [PMID: 34929014 PMCID: PMC8722729 DOI: 10.1371/journal.ppat.1010162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/03/2022] [Accepted: 12/01/2021] [Indexed: 01/08/2023] Open
Abstract
The novel coronavirus SARS-CoV-2, the causative agent of COVID-19 disease, has killed over five million people worldwide as of December 2021 with infections rising again due to the emergence of highly transmissible variants. Animal models that faithfully recapitulate human disease are critical for assessing SARS-CoV-2 viral and immune dynamics, for understanding mechanisms of disease, and for testing vaccines and therapeutics. Pigtail macaques (PTM, Macaca nemestrina) demonstrate a rapid and severe disease course when infected with simian immunodeficiency virus (SIV), including the development of severe cardiovascular symptoms that are pertinent to COVID-19 manifestations in humans. We thus proposed this species may likewise exhibit severe COVID-19 disease upon infection with SARS-CoV-2. Here, we extensively studied a cohort of SARS-CoV-2-infected PTM euthanized either 6- or 21-days after respiratory viral challenge. We show that PTM demonstrate largely mild-to-moderate COVID-19 disease. Pulmonary infiltrates were dominated by T cells, including CD4+ T cells that upregulate CD8 and express cytotoxic molecules, as well as virus-targeting T cells that were predominantly CD4+. We also noted increases in inflammatory and coagulation markers in blood, pulmonary pathologic lesions, and the development of neutralizing antibodies. Together, our data demonstrate that SARS-CoV-2 infection of PTM recapitulates important features of COVID-19 and reveals new immune and viral dynamics and thus may serve as a useful animal model for studying pathogenesis and testing vaccines and therapeutics.
Collapse
Affiliation(s)
- Alexandra Melton
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Biomedical Science Training Program, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Lara A. Doyle-Meyers
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Robert V. Blair
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Cecily Midkiff
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Hunter J. Melton
- Florida State University, Department of Statistics, Tallahassee, Florida, United States of America
| | - Kasi Russell-Lodrigue
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Pyone P. Aye
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Faith Schiro
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Marissa Fahlberg
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Dawn Szeltner
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Skye Spencer
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | | | - Kelly Goff
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Nadia Golden
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Toni Penney
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Breanna Picou
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Krystle Hensley
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Kristin E. Chandler
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Jessica A. Plante
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kenneth S. Plante
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Scott C. Weaver
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Chad J. Roy
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - James A. Hoxie
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Hongmei Gao
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - David C. Montefiori
- Duke University Medical Center, Duke Human Vaccine Institute, Durham, North Carolina, United States of America
| | - Joseph L. Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Rudolf P. Bohm
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Nicholas J. Maness
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
50
|
Douillard V, Castelli EC, Mack SJ, Hollenbach JA, Gourraud PA, Vince N, Limou S. Current HLA Investigations on SARS-CoV-2 and Perspectives. Front Genet 2021; 12:774922. [PMID: 34912378 PMCID: PMC8667766 DOI: 10.3389/fgene.2021.774922] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/08/2021] [Indexed: 01/11/2023] Open
Abstract
The rapid, global spread of the SARS-CoV-2 virus during the current pandemic has triggered numerous efforts in clinical and research settings to better understand the host genetics' interactions and the severity of COVID-19. Due to the established major role played by MHC/HLA polymorphism in infectious disease course and susceptibility, immunologists and geneticists have teamed up to investigate its contribution to the SARS-CoV-2 infection and COVID-19 progression. A major goal of the Covid-19|HLA & Immunogenetics Consortium is to support and unify these efforts. Here, we present a review of HLA immunogenomics studies in the SARS-CoV-2 pandemic and reflect on the role of various HLA data, their limitation and future perspectives.
Collapse
Affiliation(s)
- Venceslas Douillard
- Université de Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | | | - Steven J. Mack
- Division of Allergy, Immunology and Bone Marrow Transplantation, Department of Pediatrics, School of Medicine, University of California, San Francisco, CA, United States
| | - Jill A. Hollenbach
- Department of Neurology, University of California, San Francisco and Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States
| | - Pierre-Antoine Gourraud
- Université de Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Nicolas Vince
- Université de Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Sophie Limou
- Université de Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
- Ecole Centrale de Nantes, Department of Computer Sciences and Mathematics in Biology, Nantes, France
| | | |
Collapse
|