1
|
González Rosas Z, Martínez-Jiménez HS, Arroyo-Landín M, Fragoso G, Chávez-Canales M, Hernández M, Rosetti MF, López-Alvarenga JC, Sciutto E, Cárdenas G. Long-Term Neuropsychiatric Sequelae of COVID-19 in an Open Population: A Prospective Pilot Study. J Neuropsychiatry Clin Neurosci 2025:appineuropsych20240040. [PMID: 40384037 DOI: 10.1176/appi.neuropsych.20240040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
OBJECTIVE COVID-19 has been associated with a wide range of systemic and neurological complications, known as long COVID or postacute sequelae of COVID-19 (PASC). Such sequelae can be observed among all infected individuals, even among those with a mild disease course. Dysbiosis, a common condition associated with low-grade inflammation, has been proposed as a potential mechanism of PASC by altering levels of circulating lipopolysaccharide (LPS) and the tryptophan pathway metabolites kynurenine and quinolinic acid, known to affect neurocognitive function. The authors evaluated the evolution of neurological, neurocognitive, and neuropsychiatric COVID-19 sequelae and their relationship with circulating LPS and kynurenine and quinolinic acid levels. METHODS A prospective, longitudinal, and analytical study was conducted. Neurological, neurocognitive, and neuropsychiatric assessments of participants who had recovered from COVID-19 and did not require hospitalization during the acute stages of the infection were performed. Peripheral levels of LPS and tryptophan metabolites were measured 1, 3, 6, and 12 months after infection. RESULTS Of 95 participants recruited, 67 COVID-19-convalescent individuals and 20 COVID-19-free individuals were included. Significantly higher occurrences of asthenia, olfaction and taste alterations, headache, memory dysfunction, and systemic symptoms such as dyspnea, cough, and periodontal diseases were found among participants in the COVID-19-convalescent group compared with participants in the comparison group. A significant decrease in kynurenine levels, which correlated with cognitive impairment, was observed among PASC convalescents. CONCLUSIONS Significant neurocognitive and neuropsychiatric impairments were observed among COVID-19-convalescent individuals, along with decreased kynurenine levels, which recovered during a 12-month follow-up period.
Collapse
Affiliation(s)
- Zeltzin González Rosas
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - Hanna Samara Martínez-Jiménez
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - Manuel Arroyo-Landín
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - Gladis Fragoso
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - María Chávez-Canales
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - Marisela Hernández
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - Marcos Francisco Rosetti
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - Juan Carlos López-Alvarenga
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - Edda Sciutto
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| | - Graciela Cárdenas
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (González Rosas, Fragoso, Hernández, Sciutto); Department of Neurology, Instituto Nacional de Neurología y Neurocirugía, Mexico City (González Rosas, Martínez-Jiménez, Arroyo-Landín, Cárdenas); Department of Neurology, Instituto Nacional de Cardiología Ignacio Chávez and Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Chávez-Canales); Department of Cellular Biology and Physiology, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, and Psychopathology and Development Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City (Rosetti); School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas (López-Alvarenga)
| |
Collapse
|
2
|
Smail SW, Albarzinji N, Salih RH, Taha KO, Hirmiz SM, Ismael HM, Noori MF, Azeez SS, Janson C. Microbiome dysbiosis in SARS-CoV-2 infection: implication for pathophysiology and management strategies of COVID-19. Front Cell Infect Microbiol 2025; 15:1537456. [PMID: 40330025 PMCID: PMC12052750 DOI: 10.3389/fcimb.2025.1537456] [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: 11/30/2024] [Accepted: 03/27/2025] [Indexed: 05/08/2025] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of coronavirus disease 2019 (COVID-19), in late 2019 initiated a global health crisis marked by widespread infection, significant mortality, and long-term health implications. While SARS-CoV-2 primarily targets the respiratory system, recent findings indicate that it also significantly disrupts the human microbiome, particularly the gut microbiota, contributing to disease severity, systemic inflammation, immune dysregulation, and increased susceptibility to secondary infections and chronic conditions. Dysbiosis, or microbial imbalance, exacerbates the clinical outcomes of COVID-19 and has been linked to long-COVID, a condition affecting a significant proportion of survivors and manifesting with over 200 symptoms across multiple organ systems. Despite the growing recognition of microbiome alterations in COVID-19, the precise mechanisms by which SARS-CoV-2 interacts with the microbiome and influences disease progression remain poorly understood. This narrative review investigates the impact of SARS-CoV-2 on host-microbiota dynamics and evaluates its implications in disease severity and for developing personalized therapeutic strategies for COVID-19. Furthermore, it highlights the dual role of the microbiome in modulating disease progression, and as a promising target for advancing diagnostic, prognostic, and therapeutic approaches in managing COVID-19.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- College of Pharmacy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | | | - Rebaz Hamza Salih
- Department of Respiratory Medicine, PAR Private Hospital, Erbil, Kurdistan Region, Iraq
| | - Kalthum Othman Taha
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Sarah Mousa Hirmiz
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Hero M. Ismael
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Marwa Fateh Noori
- College of Pharmacy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Sarkar Sardar Azeez
- Department of Medical Laboratory Technology, Soran Technical College, Erbil Polytechnic University, Erbil, Kurdistan Region, Iraq
| | - Christer Janson
- Department of Medical Science, Respiratory Medicine, and Allergology, Uppsala University and University Hospital, Uppsala, Sweden
| |
Collapse
|
3
|
Mukherjee S, Bayry J. The Yin and Yang of TLR4 in COVID-19. Cytokine Growth Factor Rev 2025; 82:70-85. [PMID: 39490235 DOI: 10.1016/j.cytogfr.2024.10.001] [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: 07/28/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 11/05/2024]
Abstract
Various pattern recognition receptors (PRRs), including toll-like receptors (TLRs), play a crucial role in recognizing invading pathogens as well as damage-associated molecular patterns (DAMPs) released in response to infection. The resulting signaling cascades initiate appropriate immune responses to eliminate these pathogens. Current evidence suggests that SARS-CoV-2-driven activation of TLR4, whether through direct recognition of the spike glycoprotein (alone or in combination with endotoxin) or by sensing various TLR4-activating DAMPs or alarmins released during viral infection, acts as a critical mediator of antiviral immunity. However, TLR4 exerts a dual role in COVID-19, demonstrating both beneficial and deleterious effects. Dysregulated TLR4 signaling is implicated in the proinflammatory consequences linked to the immunopathogenesis of COVID-19. Additionally, TLR4 polymorphisms contribute to severity of the disease. Given its significant immunoregulatory impact on COVID-19 immunopathology and host immunity, TLR4 has emerged as a key target for developing inhibitors and immunotherapeutic strategies to mitigate the adverse effects associated with SARS-CoV-2 and related infections. Furthermore, TLR4 agonists are also being explored as adjuvants to enhance immune responses to SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory (IBIL), Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal 713 340, India.
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Paris 75006, France; Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678 623, India.
| |
Collapse
|
4
|
Tang B, Xie L, Wang Y, Shi Y, Kan W, Feng B, Lin C, Xu Z, Zhu W, Li J, Zhang X, Tian X, Zang Y. Exploratory research on the effective chemical basis of tanreqing injection for treating acute lung injury: In vivo, in vitro and in silico. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118780. [PMID: 39260706 DOI: 10.1016/j.jep.2024.118780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/16/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sepsis-induced acute lung injury (ALI) presents with significant morbidity and mortality in clinical settings. Tanreqing Injection (TRQI) has been clinically recommended for the treatment of ALI; however, the specific active chemical constituents remain unidentified. AIM OF THE STUDY This study aimed to elucidate the potential pharmacologically active components and the underlying mechanisms of TRQI in the treatment of sepsis-induced ALI. MATERIALS AND METHODS High-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) techniques were employed to identify the effective chemical constituents of TRQI. Additionally, an in vitro study was conducted using Raw264.7 macrophage cells stimulated with lipopolysaccharide (LPS) to evaluate the inhibitory effects of TRQI. An acute lung injury model produced by LPS was intraperitoneal injection in mice to assess the ALI-inhibitory effect of TRQI. The lung's pathological characteristics were examined using hematoxylin and eosin staining. Enzyme-linked immunosorbent assay (ELISA) and QPCR were performed to confirm the pharmaceutical effect. Network pharmacology was employed for mechanistic exploration, incorporating GO, and PPI analyses of targets. Src inhibitor and JNK agonist used to investigate the dependence of associated signaling pathways. RESULTS Combining pharmacokinetic characteristics, lung first-pass effect and anti-inflammatory effects, the main components of TRQI for treating sepsis induced ALI were narrowed down to seven compounds: chlorogenic acid, scutellarin, wogonoside, oroxyloside, oroxylin A and baicalein. Network pharmacology indicated that Src/JNK signaling pathway, may be the main regulatory pathway for treatment of actue lung injury. Next by using Src inhibitor, Src inhibition partly diminished the protective effects of TRQI in LPS-injected mice. Pretreatment with JNK agonist anisomycin abolished the protective effects of lung injury in vivo. CONCLUSIONS TRQI is injected, the seven compounds could be presented in vivo, which can improve ALI by inhibiting Src-JNK signaling.
Collapse
Affiliation(s)
- Bixi Tang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China; National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Like Xie
- National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yangyang Wang
- National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yulong Shi
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weijuan Kan
- National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bo Feng
- National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chenxuan Lin
- National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jia Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China; National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Shandong Laboratory of Yantai Drug Discovery,Bohai Rim Advanced Research Institute for Drug Discovery,Yantai, Shandong, 264117, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xiaoting Tian
- National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi Zang
- National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Lingang laboratory, Shanghai, 201203, China.
| |
Collapse
|
5
|
Utrata A, Schmidtner N, Mester P, Schmid S, Müller M, Pavel V, Buechler C. Plasma Lipopolysaccharide-Binding Protein (LBP) Is Induced in Critically Ill Females with Gram-Negative Infections-Preliminary Study. Infect Dis Rep 2025; 17:10. [PMID: 39997462 PMCID: PMC11855555 DOI: 10.3390/idr17010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Men are more susceptible to sepsis than women, but the underlying pathways have not been fully clarified. Lipopolysaccharide-binding protein (LBP) is an acute-phase protein that is highly elevated in sepsis. Experimental evidence shows that LBP increases to a much greater extent in male than in female mice following exposure to lipopolysaccharide. However, gender-specific studies of circulating LBP levels in sepsis patients are scarce. METHODS In the plasma of 189 patients with systemic inflammatory response syndrome (SIRS), sepsis, and septic shock, LBP levels were measured by enzyme-linked immunosorbent assay. RESULTS Patients with liver cirrhosis had reduced circulating LBP levels, regardless of gender. Further analysis within the non-cirrhotic patients showed no significant differences in LBP levels between sexes in patients with SIRS, sepsis, and septic shock. Ventilation, dialysis, and vasopressor therapy had no effect on LBP levels in either sex. A positive correlation between LBP and C-reactive protein was observed in the total cohort, males, and females. Infection with Gram-negative or Gram-positive bacteria had no effect on plasma LBP levels in males. However, female patients with Gram-negative infection had increased plasma LBP levels compared to females with negative and Gram-positive blood cultures, and 70 µg/mL LBP discriminates Gram-negative infections in females with a sensitivity of 88% and a specificity of 74%. Infection with SARS-CoV-2 did not change plasma LBP levels in either men or women. Female patients who did not survive had lower plasma LBP levels compared to female survivors and male non-survivors. CONCLUSIONS This investigation highlights the influence of sex on plasma LBP levels in SIRS/sepsis patients, suggesting that LBP could be a sex-specific biomarker in critically ill patients.
Collapse
|
6
|
An Y, He L, Xu X, Piao M, Wang B, Liu T, Cao H. Gut microbiota in post-acute COVID-19 syndrome: not the end of the story. Front Microbiol 2024; 15:1500890. [PMID: 39777148 PMCID: PMC11703812 DOI: 10.3389/fmicb.2024.1500890] [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: 09/24/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has led to major global health concern. However, the focus on immediate effects was assumed as the tip of iceberg due to the symptoms following acute infection, which was defined as post-acute COVID-19 syndrome (PACS). Gut microbiota alterations even after disease resolution and the gastrointestinal symptoms are the key features of PACS. Gut microbiota and derived metabolites disorders may play a crucial role in inflammatory and immune response after SARS-CoV-2 infection through the gut-lung axis. Diet is one of the modifiable factors closely related to gut microbiota and COVID-19. In this review, we described the reciprocal crosstalk between gut and lung, highlighting the participation of diet and gut microbiota in and after COVID-19 by destroying the gut barrier, perturbing the metabolism and regulating the immune system. Therefore, bolstering beneficial species by dietary supplements, probiotics or prebiotics and fecal microbiota transplantation (FMT) may be a novel avenue for COVID-19 and PACS prevention. This review provides a better understanding of the association between gut microbiota and the long-term consequences of COVID-19, which indicates modulating gut dysbiosis may be a potentiality for addressing this multifaceted condition.
Collapse
Affiliation(s)
| | | | | | | | | | - Tianyu Liu
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
7
|
Fallah A, Sedighian H, Kachuei R, Fooladi AAI. Human microbiome in post-acute COVID-19 syndrome (PACS). CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 8:100324. [PMID: 39717208 PMCID: PMC11665312 DOI: 10.1016/j.crmicr.2024.100324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
The global COVID-19 pandemic, which began in 2019, is still ongoing. SARS-CoV-2, also known as the severe acute respiratory syndrome coronavirus 2, is the causative agent. Diarrhea, nausea, and vomiting are common GI symptoms observed in a significant number of COVID-19 patients. Additionally, the respiratory and GI tracts express high level of transmembrane protease serine 2 (TMPRSS2) and angiotensin-converting enzyme-2 (ACE2), making them primary sites for human microbiota and targets for SARS-CoV-2 infection. A growing body of research indicates that individuals with COVID-19 and post-acute COVID-19 syndrome (PACS) exhibit considerable alterations in their microbiome. In various human disorders, including diabetes, obesity, cancer, ulcerative colitis, Crohn's disease, and several viral infections, the microbiota play a significant immunomodulatory role. In this review, we investigate the potential therapeutic implications of the interactions between host microbiota and COVID-19. Microbiota-derived metabolites and components serve as primary mediators of microbiota-host interactions, influencing host immunity. We discuss the various mechanisms through which these metabolites or components produced by the microbiota impact the host's immune response to SARS-CoV-2 infection. Additionally, we address confounding factors in microbiome studies. Finally, we examine and discuss about a range of potential microbiota-based prophylactic measures and treatments for COVID-19 and PACS, as well as their effects on clinical outcomes and disease severity.
Collapse
Affiliation(s)
- Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Basting CM, Langat R, Broedlow CA, Guerrero CR, Bold TD, Bailey M, Velez A, Schroeder T, Short-Miller J, Cromarty R, Mayer ZJ, Southern PJ, Schacker TW, Safo SE, Bramante CT, Tignanelli CJ, Schifanella L, Klatt NR. SARS-CoV-2 infection is associated with intestinal permeability, systemic inflammation, and microbial dysbiosis in hospitalized patients. Microbiol Spectr 2024; 12:e0068024. [PMID: 39345212 PMCID: PMC11537016 DOI: 10.1128/spectrum.00680-24] [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: 05/22/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) and its associated severity have been linked to uncontrolled inflammation and may be associated with changes in the microbiome of mucosal sites including the gastrointestinal tract and oral cavity. These sites play an important role in host-microbe homeostasis, and disruption of epithelial barrier integrity during COVID-19 may potentially lead to exacerbated inflammation and immune dysfunction. Outcomes in COVID-19 are highly disparate, ranging from asymptomatic to fatal, and the impact of microbial dysbiosis on disease severity is unclear. Here, we obtained plasma, rectal swabs, oropharyngeal swabs, and nasal swabs from 86 patients hospitalized with COVID-19 and 12 healthy volunteers. We performed 16S rRNA sequencing to characterize the microbial communities in the mucosal swabs and measured concentrations of circulating cytokines, markers of gut barrier integrity, and fatty acids in the plasma samples. We compared these plasma concentrations and microbiomes between healthy volunteers and COVID-19 patients, some of whom had unfortunately died by the end of the study enrollment, and performed a correlation analysis between plasma variables and bacterial abundances. Rectal swabs of COVID-19 patients had reduced abundances of several commensal bacteria including Faecalibacterium prausnitzii and an increased abundance of the opportunistic pathogens Eggerthella lenta and Hungatella hathewayi. Furthermore, the oral pathogen Scardovia wiggsiae was more abundant in the oropharyngeal swabs of COVID-19 patients who died. The abundance of both H. hathewayi and S. wiggsiae correlated with circulating inflammatory markers including IL-6, highlighting the possible role of the microbiome in COVID-19 severity and providing potential therapeutic targets for managing COVID-19.IMPORTANCEOutcomes in coronavirus disease 2019 (COVID-19) are highly disparate and are associated with uncontrolled inflammation; however, the individual factors that lead to this uncontrolled inflammation are not fully understood. Here, we report that severe COVID-19 is associated with systemic inflammation, microbial translocation, and microbial dysbiosis. The rectal and oropharyngeal microbiomes of COVID-19 patients were characterized by a decreased abundance of commensal bacteria and an increased abundance of opportunistic pathogens, which positively correlated with markers of inflammation and microbial translocation. These microbial perturbations may, therefore, contribute to disease severity in COVID-19 and highlight the potential for microbiome-based interventions in improving COVID-19 outcomes.
Collapse
Affiliation(s)
| | - Robert Langat
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Candace R. Guerrero
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minnesota, Minneapolis, USA
- College of Biological Sciences, University of Minnesota, Minnesota, Minneapolis, USA
| | - Tyler D. Bold
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | - Melisa Bailey
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Adrian Velez
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ty Schroeder
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jonah Short-Miller
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ross Cromarty
- Masonic Cancer Center, University of Minnesota, Minnesota, Minneapolis, USA
| | - Zachary J. Mayer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minnesota, Minneapolis, USA
- College of Biological Sciences, University of Minnesota, Minnesota, Minneapolis, USA
| | - Peter J. Southern
- Department of Microbiology and Immunology, University of Minnesota, Minnesota, Minneapolis, USA
| | - Timothy W. Schacker
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | - Sandra E. Safo
- Department of Biostatistics and Health Data Science, University of Minnesota, Minnesota, Minneapolis, USA
| | - Carolyn T. Bramante
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | | | - Luca Schifanella
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- National Cancer Institute, Center for Cancer Research, Vaccine Branch, Animal Models and Retroviral Vaccines Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Nichole R. Klatt
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
9
|
Adhikari A, Maddumage J, Eriksson EM, Annesley SJ, Lawson VA, Bryant VL, Gras S. Beyond acute infection: mechanisms underlying post-acute sequelae of COVID-19 (PASC). Med J Aust 2024; 221 Suppl 9:S40-S48. [PMID: 39489518 DOI: 10.5694/mja2.52456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 08/10/2024] [Indexed: 11/05/2024]
Abstract
Immune dysregulation is a key aspect of post-acute sequelae of coronavirus disease 2019 (PASC), also known as long COVID, with sustained activation of immune cells, T cell exhaustion, skewed B cell profiles, and disrupted immune communication thereby resulting in autoimmune-related complications. The gut is emerging as a critical link between microbiota, metabolism and overall dysfunction, potentially sharing similarities with other chronic fatigue conditions and PASC. Immunothrombosis and neurological signalling dysfunction emphasise the complex interplay between the immune system, blood clotting, and the central nervous system in the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Clear research gaps in the design of PASC studies, especially in the context of longitudinal research, stand out as significant areas of concern.
Collapse
Affiliation(s)
- Anurag Adhikari
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC
- Kathmandu Research Institute for Biological Sciences, Lalitpur, Nepal
| | - Janesha Maddumage
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC
| | - Emily M Eriksson
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
| | | | - Victoria A Lawson
- Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC
| | - Vanessa L Bryant
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
- Royal Melbourne Hospital, Melbourne, VIC
- University of Melbourne, Melbourne, VIC
| | - Stephanie Gras
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC
- Monash University, Melbourne, VIC
| |
Collapse
|
10
|
de Farias Fraga G, da Silva Rodrigues F, Jantsch J, Silva Dias V, Milczarski V, Wickert F, Pereira Medeiros C, Eller S, Gatto Barschak A, Giovenardi M, Padilha Guedes R. Omega-3 Attenuates Disrupted Neurotransmission and Partially Protects Metabolic Dysfunction Caused by Obesity in Wistar Rats. Neurochem Res 2024; 49:2763-2773. [PMID: 38960951 DOI: 10.1007/s11064-024-04201-0] [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: 12/22/2023] [Revised: 05/02/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
Omega-3 (n3) is a polyunsaturated fatty acid well known for its anti-inflammatory and neuroprotective properties. Obesity is linked to chronic inflammation that disrupts metabolism, the intestine physiology and the central nervous system functioning. This study aims to determine if n3 supplementation can interfere with the effects of obesity on the mitochondrial activity, intestinal barrier, and neurotransmitter levels in the brain of Wistar rats that received cafeteria diet (CAF). We examined adipose tissue, skeletal muscle, plasma, intestine, and the cerebral cortex of four groups: CT (control diet), CTn3 (control diet with n3 supplementation), CAF, and CAFn3 (CAF and n3). Diets were offered for 13 weeks, with n3 supplementation in the final 5 weeks. Adipose tissue Electron Transport Chain complexes I, II, and III showed higher activity in CAF groups, as did complexes III and IV in skeletal muscle. Acetate levels in plasma were reduced in CAF groups, and Lipopolysaccharide (LPS) was higher in the CAF group but reduced in CAFn3 group. Claudin-5 in the intestine was lower in CAF groups, with no n3 supplementation effect. In the cerebral cortex, dopamine levels were decreased with CAF, which was reversed by n3. DOPAC, a dopamine metabolite, also showed a supplementation effect, and HVA, a diet effect. Serotonin levels increased in the CAF group that received supplementation. Therefore, we demonstrate disturbances in mitochondria, plasma, intestine and brain of rats submitted to CAF and the potential benefit of n3 supplementation in endotoxemia and neurotransmitter levels.
Collapse
Affiliation(s)
- Gabriel de Farias Fraga
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Victor Silva Dias
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Vitória Milczarski
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Fernanda Wickert
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Camila Pereira Medeiros
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Sarah Eller
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Alethéa Gatto Barschak
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Marcia Giovenardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil.
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil.
| |
Collapse
|
11
|
Wilson JD, Dworsky-Fried M, Ismail N. Neurodevelopmental implications of COVID-19-induced gut microbiome dysbiosis in pregnant women. J Reprod Immunol 2024; 165:104300. [PMID: 39004033 DOI: 10.1016/j.jri.2024.104300] [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: 04/04/2024] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
The global public health emergency of COVID-19 in January 2020 prompted a surge in research focusing on the pathogenesis and clinical manifestations of the virus. While numerous reports have been published on the acute effects of COVID-19 infection, the review explores the multifaceted long-term implications of COVID-19, with a particular focus on severe maternal COVID-19 infection, gut microbiome dysbiosis, and neurodevelopmental disorders in offspring. Severe COVID-19 infection has been associated with heightened immune system activation and gastrointestinal symptoms. Severe COVID-19 may also result in gut microbiome dysbiosis and a compromised intestinal mucosal barrier, often referred to as 'leaky gut'. Increased gut permeability facilitates the passage of inflammatory cytokines, originating from the inflamed intestinal mucosa and gut, into the bloodstream, thereby influencing fetal development during pregnancy and potentially elevating the risk of neurodevelopmental disorders such as autism and schizophrenia. The current review discusses the role of cytokine signaling molecules, microglia, and synaptic pruning, highlighting their potential involvement in the pathogenesis of neurodevelopmental disorders following maternal COVID-19 infection. Additionally, this review addresses the potential of probiotic interventions to mitigate gut dysbiosis and inflammatory responses associated with COVID-19, offering avenues for future research in optimizing maternal and fetal health outcomes.
Collapse
Affiliation(s)
- Jacob D Wilson
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada
| | - Michaela Dworsky-Fried
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada
| | - Nafissa Ismail
- NISE Laboratory, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, Ontario K1N 9A4, Canada; LIFE Research Institute, Ottawa, Ontario K1N 6N5, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada.
| |
Collapse
|
12
|
Dos Santos Medeiros SMDFR, Sousa Lino BMN, Perez VP, Sousa ESS, Campana EH, Miyajima F, Carvalho-Silva WHV, Dejani NN, de Sousa Fernandes MS, Yagin FH, Al-Hashem F, Elkholi SM, Alyami H, Souto FO. Predictive biomarkers of mortality in patients with severe COVID-19 hospitalized in intensive care unit. Front Immunol 2024; 15:1416715. [PMID: 39281667 PMCID: PMC11401048 DOI: 10.3389/fimmu.2024.1416715] [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: 04/12/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Objectives This study was performed to identify predictive markers of worse outcomes in patients with severe COVID-19 in an intensive care unit. Methods Sixty patients with severe COVID-19, hospitalized in the Intensive Care Unit (ICU) between March and July 2021, were stratified into two groups according to the outcome survivors and non-survivors. After admission to the ICU, blood samples were collected directly for biomarker analysis. Routine hematological and biochemical biomarkers, as well as serum levels of cytokines, chemokines, and immunoglobulins, were investigated. Results Lymphopenia, neutrophilia, and thrombocytopenia were more pronounced in non-surviving patients, while the levels of CRP, AST, creatinine, ferritin, AST, troponin I, urea, magnesium, and potassium were higher in the non-surviving group than the survival group. In addition, serum levels of IL-10, CCL2, CXCL9, and CXCL10 were significantly increased in patients who did not survive. These changes in the biomarkers evaluated were associated with increased mortality in patients with severe COVID-19. Conclusion The present study confirmed and expanded the validity of laboratory biomarkers as indicators of mortality in severe COVID-19.
Collapse
Affiliation(s)
- Sandrelli Meridiana de Fátima Ramos Dos Santos Medeiros
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Molecular Biology Laboratory (LABIMOL), Medical Sciences Center, Federal University of Paraíba (CCM/UFPB), João Pessoa, Paraíba, Brazil
| | | | - Vinícius Pietta Perez
- Molecular Biology Laboratory (LABIMOL), Medical Sciences Center, Federal University of Paraíba (CCM/UFPB), João Pessoa, Paraíba, Brazil
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba (CCS/UFPB), João Pessoa, Paraíba, Brazil
| | - Eduardo Sérgio Soares Sousa
- Molecular Biology Laboratory (LABIMOL), Medical Sciences Center, Federal University of Paraíba (CCM/UFPB), João Pessoa, Paraíba, Brazil
- Department of Obstetrics and Gynecology, Medical Sciences Center, Federal University of Paraíba (CCM/UFPB), João Pessoa, Paraíba, Brazil
| | - Eloiza Helena Campana
- Molecular Biology Laboratory (LABIMOL), Medical Sciences Center, Federal University of Paraíba (CCM/UFPB), João Pessoa, Paraíba, Brazil
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Paraíba (CCS/UFPB), João Pessoa, Paraíba, Brazil
| | | | | | - Naiara Naiana Dejani
- Molecular Biology Laboratory (LABIMOL), Medical Sciences Center, Federal University of Paraíba (CCM/UFPB), João Pessoa, Paraíba, Brazil
- Department of Physiology and Pathology, Health Sciences Center, Federal University of Paraíba (CCS/UFPB), João Pessoa, Paraíba, Brazil
| | | | - Fatma Hilal Yagin
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, Malatya, Türkiye
| | - Fahaid Al-Hashem
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Safaa M Elkholi
- Department of Rehabilitation Sciences, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Hanan Alyami
- Department of Medical and Surgical Nursing, College of Nursing, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Fabrício Oliveira Souto
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| |
Collapse
|
13
|
Boncheva I, Poudrier J, Falcone EL. Role of the intestinal microbiota in host defense against respiratory viral infections. Curr Opin Virol 2024; 66:101410. [PMID: 38718575 DOI: 10.1016/j.coviro.2024.101410] [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: 01/17/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 06/07/2024]
Abstract
Viral infections, including those affecting the respiratory tract, can alter the composition of the intestinal microbiota, which, in turn, can significantly influence both innate and adaptive immune responses, resulting in either enhanced pathogen clearance or exacerbation of the infection, possibly leading to inflammatory complications. A deeper understanding of the interplay between the intestinal microbiota and host immune responses in the context of respiratory viral infections (i.e. the gut-lung axis) is necessary to develop new treatments. This review highlights key mechanisms by which the intestinal microbiota, including its metabolites, can act locally or at distant organs to combat respiratory viruses. Therapeutics aimed at harnessing the microbiota to prevent and/or help treat respiratory viral infections represent a promising avenue for future investigation.
Collapse
Affiliation(s)
- Idia Boncheva
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Johanne Poudrier
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada; Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Emilia L Falcone
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada; Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada; Department of Medicine, Université de Montréal, Montreal, QC, Canada; Department of Microbiology and Infectious Diseases, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.
| |
Collapse
|
14
|
Invernici F, Bernasconi A, Ceri S. Searching COVID-19 Clinical Research Using Graph Queries: Algorithm Development and Validation. J Med Internet Res 2024; 26:e52655. [PMID: 38814687 PMCID: PMC11176882 DOI: 10.2196/52655] [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: 11/26/2023] [Revised: 03/06/2024] [Accepted: 03/30/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Since the beginning of the COVID-19 pandemic, >1 million studies have been collected within the COVID-19 Open Research Dataset, a corpus of manuscripts created to accelerate research against the disease. Their related abstracts hold a wealth of information that remains largely unexplored and difficult to search due to its unstructured nature. Keyword-based search is the standard approach, which allows users to retrieve the documents of a corpus that contain (all or some of) the words in a target list. This type of search, however, does not provide visual support to the task and is not suited to expressing complex queries or compensating for missing specifications. OBJECTIVE This study aims to consider small graphs of concepts and exploit them for expressing graph searches over existing COVID-19-related literature, leveraging the increasing use of graphs to represent and query scientific knowledge and providing a user-friendly search and exploration experience. METHODS We considered the COVID-19 Open Research Dataset corpus and summarized its content by annotating the publications' abstracts using terms selected from the Unified Medical Language System and the Ontology of Coronavirus Infectious Disease. Then, we built a co-occurrence network that includes all relevant concepts mentioned in the corpus, establishing connections when their mutual information is relevant. A sophisticated graph query engine was built to allow the identification of the best matches of graph queries on the network. It also supports partial matches and suggests potential query completions using shortest paths. RESULTS We built a large co-occurrence network, consisting of 128,249 entities and 47,198,965 relationships; the GRAPH-SEARCH interface allows users to explore the network by formulating or adapting graph queries; it produces a bibliography of publications, which are globally ranked; and each publication is further associated with the specific parts of the query that it explains, thereby allowing the user to understand each aspect of the matching. CONCLUSIONS Our approach supports the process of query formulation and evidence search upon a large text corpus; it can be reapplied to any scientific domain where documents corpora and curated ontologies are made available.
Collapse
Affiliation(s)
- Francesco Invernici
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Anna Bernasconi
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Stefano Ceri
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
15
|
Duve K, Petakh P, Kamyshnyi O. COVID-19-associated encephalopathy: connection between neuroinflammation and microbiota-gut-brain axis. Front Microbiol 2024; 15:1406874. [PMID: 38863751 PMCID: PMC11165208 DOI: 10.3389/fmicb.2024.1406874] [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: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/13/2024] Open
Abstract
While neurological complications of COVID-19, such as encephalopathy, are relatively rare, their potential significant impact on long-term morbidity is substantial, especially given the large number of infected patients. Two proposed hypotheses for the pathogenesis of this condition are hypoxia and the uncontrolled release of proinflammatory cytokines. The gut microbiota plays an important role in regulating immune homeostasis and overall gut health, including its effects on brain health through various pathways collectively termed the gut-brain axis. Recent studies have shown that COVID-19 patients exhibit gut dysbiosis, but how this dysbiosis can affect inflammation in the central nervous system (CNS) remains unclear. In this context, we discuss how dysbiosis could contribute to neuroinflammation and provide recent data on the features of neuroinflammation in COVID-19 patients.
Collapse
Affiliation(s)
- Khrystyna Duve
- Department of Neurology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Oleksandr Kamyshnyi
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| |
Collapse
|
16
|
Wei Y, Yu W, Zhang Z, Liu S, Xue J, Wu C, Gao Z, Guo S. Comparative analysis of oropharyngeal microbiota in healthcare workers post-COVID-19. Front Cell Infect Microbiol 2024; 14:1347345. [PMID: 38828262 PMCID: PMC11140064 DOI: 10.3389/fcimb.2024.1347345] [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: 11/30/2023] [Accepted: 04/25/2024] [Indexed: 06/05/2024] Open
Abstract
Background To date, more than 770 million individuals have become coronavirus disease 2019 (COVID-19) convalescents worldwide. Emerging evidence highlights the influence of COVID-19 on the oral microbiome during both acute and convalescent disease phases. Front-line healthcare workers are at an elevated risk of exposure to viral infections, and the effects of COVID-19 on their oral microbiome remain relatively unexplored. Methods Oropharyngeal swab specimens, collected one month after a negative COVID-19 test from a cohort comprising 55 healthcare workers, underwent 16S rRNA sequencing. We conducted a comparative analysis between this post-COVID-19 cohort and the pre-infection dataset from the same participants. Community composition analysis, indicator species analysis, alpha diversity assessment, beta diversity exploration, and functional prediction were evaluated. Results The Shannon and Simpson indexes of the oral microbial community declined significantly in the post-COVID-19 group when compared with the pre-infection cohort. Moreover, there was clear intergroup clustering between the two groups. In the post-COVID-19 group, the phylum Firmicutes showed a significant increase. Further, there were clear differences in relative abundance of several bacterial genera in contrast with the pre-infection group, including Streptococcus, Gemella, Granulicatella, Capnocytophaga, Leptotrichia, Fusobacterium, and Prevotella. We identified Gemella enrichment in the post-COVID-19 group, potentially serving as a recovery period performance indicator. Functional prediction revealed lipopolysaccharide biosynthesis downregulation in the post-COVID-19 group, an outcome with host inflammatory response modulation and innate defence mechanism implications. Conclusion During the recovery phase of COVID-19, the oral microbiome diversity of front-line healthcare workers failed to fully return to its pre-infection state. Despite the negative COVID-19 test result one month later, notable disparities persisted in the composition and functional attributes of the oral microbiota.
Collapse
Affiliation(s)
- Yue Wei
- Nursing of school, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenyi Yu
- Department of Respiratory and Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Zhixia Zhang
- Nursing Department, Linfen Central Hospital, Shanxi, China
| | - Siqin Liu
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianbo Xue
- Department of Respiratory and Critical Care Medicine, Peking University People’s Hospital, Beijing, China
| | - Chunyan Wu
- Nursing of school, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhancheng Gao
- Department of Respiratory and Critical Care Medicine, Peking University People’s Hospital, Beijing, China
- Institute of Chest and Lung Diseases, Shanxi Medical University, Linfen, Shanxi, China
| | - Shuming Guo
- Nursing of school, Shanxi Medical University, Taiyuan, Shanxi, China
- Nursing Department, Linfen Central Hospital, Shanxi, China
| |
Collapse
|
17
|
Madruga MP, Grun LK, Santos LSMD, Friedrich FO, Antunes DB, Rocha MEF, Silva PL, Dorneles GP, Teixeira PC, Oliveira TF, Romão PRT, Santos L, Moreira JCF, Michaelsen VS, Cypel M, Antunes MOB, Jones MH, Barbé-Tuana FM, Bauer ME. Excess of body weight is associated with accelerated T-cell senescence in hospitalized COVID-19 patients. Immun Ageing 2024; 21:17. [PMID: 38454515 PMCID: PMC10921685 DOI: 10.1186/s12979-024-00423-6] [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: 12/27/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Several risk factors have been involved in the poor clinical progression of coronavirus disease-19 (COVID-19), including ageing, and obesity. SARS-CoV-2 may compromise lung function through cell damage and paracrine inflammation; and obesity has been associated with premature immunosenescence, microbial translocation, and dysfunctional innate immune responses leading to poor immune response against a range of viruses and bacterial infections. Here, we have comprehensively characterized the immunosenescence, microbial translocation, and immune dysregulation established in hospitalized COVID-19 patients with different degrees of body weight. RESULTS Hospitalised COVID-19 patients with overweight and obesity had similarly higher plasma LPS and sCD14 levels than controls (all p < 0.01). Patients with obesity had higher leptin levels than controls. Obesity and overweight patients had similarly higher expansions of classical monocytes and immature natural killer (NK) cells (CD56+CD16-) than controls. In contrast, reduced proportions of intermediate monocytes, mature NK cells (CD56+CD16+), and NKT were found in both groups of patients than controls. As expected, COVID-19 patients had a robust expansion of plasmablasts, contrasting to lower proportions of major T-cell subsets (CD4 + and CD8+) than controls. Concerning T-cell activation, overweight and obese patients had lower proportions of CD4+CD38+ cells than controls. Contrasting changes were reported in CD25+CD127low/neg regulatory T cells, with increased and decreased proportions found in CD4+ and CD8+ T cells, respectively. There were similar proportions of T cells expressing checkpoint inhibitors across all groups. We also investigated distinct stages of T-cell differentiation (early, intermediate, and late-differentiated - TEMRA). The intermediate-differentiated CD4 + T cells and TEMRA cells (CD4+ and CD8+) were expanded in patients compared to controls. Senescent T cells can also express NK receptors (NKG2A/D), and patients had a robust expansion of CD8+CD57+NKG2A+ cells than controls. Unbiased immune profiling further confirmed the expansions of senescent T cells in COVID-19. CONCLUSIONS These findings suggest that dysregulated immune cells, microbial translocation, and T-cell senescence may partially explain the increased vulnerability to COVID-19 in subjects with excess of body weight.
Collapse
Affiliation(s)
- Mailton Prestes Madruga
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Lucas Kich Grun
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Letícya Simone Melo Dos Santos
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | | | - Douglas Bitencourt Antunes
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Marcella Elesbão Fogaça Rocha
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Pedro Luis Silva
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Gilson P Dorneles
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Paula Coelho Teixeira
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Tiago Franco Oliveira
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Pedro R T Romão
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Lucas Santos
- Centro de Estudos em Estresse Oxidativo - Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (IB-UFRGS), Porto Alegre, RS, Brazil
| | - José Claudio Fonseca Moreira
- Centro de Estudos em Estresse Oxidativo - Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (IB-UFRGS), Porto Alegre, RS, Brazil
| | - Vinicius Schenk Michaelsen
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Marcelo Cypel
- Toronto General Hospital Research Institute, Department of Surgery, University Health Network, University of Toronto, Toronto, Canada
| | - Marcos Otávio Brum Antunes
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Marcus Herbert Jones
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Florencia María Barbé-Tuana
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil
| | - Moisés Evandro Bauer
- Laboratory of Immunobiology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6681, building 12 (4th floor), Porto Alegre, 90619-900, RS, Brazil.
| |
Collapse
|
18
|
Huang K, Li Z, He X, Dai J, Huang B, Shi Y, Fan D, Zhang Z, Liu Y, Li N, Zhang Z, Peng J, Liu C, Zeng R, Cen Z, Wang T, Yang W, Cen M, Li J, Yuan S, Zhang L, Hu D, Huang S, Chen P, Lai P, Lin L, Wen J, Zhao Z, Huang X, Yuan L, Zhou L, Wu H, Huang L, Feng K, Wang J, Liao B, Cai W, Deng X, Li Y, Li J, Hu Z, Yang L, Li J, Zhuo Y, Zhang F, Lin L, Luo Y, Zhang W, Ni Q, Hong X, Chang G, Zhang Y, Guan D, Cai W, Lu Y, Li F, Yan L, Ren M, Li L, Chen S. Gut microbial co-metabolite 2-methylbutyrylcarnitine exacerbates thrombosis via binding to and activating integrin α2β1. Cell Metab 2024; 36:598-616.e9. [PMID: 38401546 DOI: 10.1016/j.cmet.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/08/2023] [Accepted: 01/25/2024] [Indexed: 02/26/2024]
Abstract
Thrombosis represents the leading cause of death and disability upon major adverse cardiovascular events (MACEs). Numerous pathological conditions such as COVID-19 and metabolic disorders can lead to a heightened thrombotic risk; however, the underlying mechanisms remain poorly understood. Our study illustrates that 2-methylbutyrylcarnitine (2MBC), a branched-chain acylcarnitine, is accumulated in patients with COVID-19 and in patients with MACEs. 2MBC enhances platelet hyperreactivity and thrombus formation in mice. Mechanistically, 2MBC binds to integrin α2β1 in platelets, potentiating cytosolic phospholipase A2 (cPLA2) activation and platelet hyperresponsiveness. Genetic depletion or pharmacological inhibition of integrin α2β1 largely reverses the pro-thrombotic effects of 2MBC. Notably, 2MBC can be generated in a gut-microbiota-dependent manner, whereas the accumulation of plasma 2MBC and its thrombosis-aggravating effect are largely ameliorated following antibiotic-induced microbial depletion. Our study implicates 2MBC as a metabolite that links gut microbiota dysbiosis to elevated thrombotic risk, providing mechanistic insight and a potential therapeutic strategy for thrombosis.
Collapse
Affiliation(s)
- Kan Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China; Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xi He
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Jun Dai
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Bingding Huang
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, Guangdong 518118, China
| | - Yongxia Shi
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Dongxiao Fan
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zefeng Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Yunchong Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Na Li
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zhongyu Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Jiangyun Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Chenshu Liu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Renli Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Zhipeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Wenchao Yang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Meifeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Jingyu Li
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, Guangdong 518118, China
| | - Shuai Yuan
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Lu Zhang
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Dandan Hu
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Shuxiang Huang
- Guangzhou Customs District Technology Center, Guangzhou, Guangdong 510700, China
| | - Pin Chen
- National Supercomputer Center in Guangzhou, School of Computer Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Peilong Lai
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Liyan Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Jielu Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Zhengde Zhao
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xiuyi Huang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lining Yuan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Lifang Zhou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Haoliang Wu
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lihua Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China
| | - Kai Feng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Jian Wang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Baolin Liao
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Weiping Cai
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Xilong Deng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Yueping Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Jianping Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Zhongwei Hu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Li Yang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Jiaojiao Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Youguang Zhuo
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Fuchun Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China
| | - Lin Lin
- Department of Respiratory Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Yifeng Luo
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Sun Yat-sen University, Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Wei Zhang
- Wuhan Metware Biotechnology Co., Ltd., Wuhan, Hubei 430070, China
| | - Qianlin Ni
- Wuhan Metware Biotechnology Co., Ltd., Wuhan, Hubei 430070, China
| | - Xiqiang Hong
- Wuhan Metware Biotechnology Co., Ltd., Wuhan, Hubei 430070, China
| | - Guangqi Chang
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yang Zhang
- School of Public Health, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Dongxian Guan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Weikang Cai
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568, USA
| | - Yutong Lu
- National Supercomputer Center in Guangzhou, School of Computer Science and Engineering, Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Fang Li
- Department of Obstetrics and Gynecology, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510620, China
| | - Li Yan
- Guangdong Clinical Research Center for Metabolic Diseases (Diabetes), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Meng Ren
- Guangdong Clinical Research Center for Metabolic Diseases (Diabetes), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China.
| | - Linghua Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510060, China.
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China; Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, Guangdong 528200, China.
| |
Collapse
|
19
|
Wang L, Cao JB, Xia BB, Li YJ, Zhang X, Mo GX, Wang RJ, Guo SQ, Zhang YQ, Xiao K, Zhu GF, Liu PF, Song LC, Ma XH, Xiang PC, Wang J, Liu YH, Xie F, Zhang XD, Li XX, Sun WL, Cao Y, Wang KF, Zhang WH, Zhao WC, Yan P, Chen JC, Yang YW, Yu ZK, Tang JS, Xiao L, Zhou JM, Xie LX, Wang J. Metatranscriptome of human lung microbial communities in a cohort of mechanically ventilated COVID-19 Omicron patients. Signal Transduct Target Ther 2023; 8:432. [PMID: 37949875 PMCID: PMC10638395 DOI: 10.1038/s41392-023-01684-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/21/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023] Open
Abstract
The Omicron variant of the severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) infected a substantial proportion of Chinese population, and understanding the factors underlying the severity of the disease and fatality is valuable for future prevention and clinical treatment. We recruited 64 patients with invasive ventilation for COVID-19 and performed metatranscriptomic sequencing to profile host transcriptomic profiles, plus viral, bacterial, and fungal content, as well as virulence factors and examined their relationships to 28-day mortality were examined. In addition, the bronchoalveolar lavage fluid (BALF) samples from invasive ventilated hospital/community-acquired pneumonia patients (HAP/CAP) sampled in 2019 were included for comparison. Genomic analysis revealed that all Omicron strains belong to BA.5 and BF.7 sub-lineages, with no difference in 28-day mortality between them. Compared to HAP/CAP cohort, invasive ventilated COVID-19 patients have distinct host transcriptomic and microbial signatures in the lower respiratory tract; and in the COVID-19 non-survivors, we found significantly lower gene expressions in pathways related viral processes and positive regulation of protein localization to plasma membrane, higher abundance of opportunistic pathogens including bacterial Alloprevotella, Caulobacter, Escherichia-Shigella, Ralstonia and fungal Aspergillus sydowii and Penicillium rubens. Correlational analysis further revealed significant associations between host immune responses and microbial compositions, besides synergy within viral, bacterial, and fungal pathogens. Our study presents the relationships of lower respiratory tract microbiome and transcriptome in invasive ventilated COVID-19 patients, providing the basis for future clinical treatment and reduction of fatality.
Collapse
Affiliation(s)
- Lin Wang
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Jia-Bao Cao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bin-Bin Xia
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yue-Juan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuan Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Faculty of Biological Science and Technology, Baotou Teacher's College, Baotou, 014030, China
| | - Guo-Xin Mo
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Rui-Juan Wang
- Department of Respiratory Medicine, PLA Strategic Support Force Medical Center, Beijing, 100101, China
| | - Si-Qi Guo
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yu-Qing Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kun Xiao
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Guang-Fa Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Peng-Fei Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Li-Cheng Song
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Xi-Hui Ma
- Respiratory Research Institute, Department of Pulmonary & Critical Care Medicine, Beijing Key Laboratory of OTIR, the 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Ping-Chao Xiang
- Shougang hospital of Peking University, Beijing, 100144, China
| | - Jiang Wang
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Yu-Hong Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Fei Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Xu-Dong Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiang-Xin Li
- Department of Respiratory Medicine, Beijing Changping Hospital, Beijing, 102200, China
| | - Wan-Lu Sun
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yan Cao
- Pulmonary research institute, Senior Department of Respiratory and Critical Care Medicine, the 8th medical center of Chinese PLA general hospital, Beijing, 100091, China
| | - Kai-Fei Wang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wen-Hui Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei-Chao Zhao
- Department of Respiratory Medicine, PLA Strategic Support Force Medical Center, Beijing, 100101, China
| | - Peng Yan
- China Aerospace Science & Industry Corporation 731 hospital, Beijing, 100074, China
| | - Ji-Chao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Beijing, 100049, China
| | - Yu-Wei Yang
- Respiratory Research Institute, Department of Pulmonary & Critical Care Medicine, Beijing Key Laboratory of OTIR, the 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Zhong-Kuo Yu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Jing-Si Tang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Li Xiao
- Respiratory Research Institute, Department of Pulmonary & Critical Care Medicine, Beijing Key Laboratory of OTIR, the 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Jie-Min Zhou
- Vision Medicals Center for Infectious Diseases, Guangzhou, 510700, China
| | - Li-Xin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China.
| | - Jun Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
20
|
Bourumeau W, Tremblay K, Jourdan G, Girard C, Laprise C. Bacterial Biomarkers of the Oropharyngeal and Oral Cavity during SARS-CoV-2 Infection. Microorganisms 2023; 11:2703. [PMID: 38004715 PMCID: PMC10673573 DOI: 10.3390/microorganisms11112703] [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: 09/26/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
(1) Background: Individuals with COVID-19 display different forms of disease severity and the upper respiratory tract microbiome has been suggested to play a crucial role in the development of its symptoms. (2) Methods: The present study analyzed the microbial profiles of the oral cavity and oropharynx of 182 COVID-19 patients compared to 75 unaffected individuals. The samples were obtained from gargle screening samples. 16S rRNA amplicon sequencing was applied to analyze the samples. (3) Results: The present study shows that SARS-CoV-2 infection induced significant differences in bacterial community assemblages, with Prevotella and Veillonella as biomarkers for positive-tested people and Streptococcus and Actinomyces for negative-tested people. It also suggests a state of dysbiosis on the part of the infected individuals due to significant differences in the bacterial community in favor of a microbiome richer in opportunistic pathogens. (4) Conclusions: SARS-CoV-2 infection induces dysbiosis in the upper respiratory tract. The identification of these opportunistic pathogenic biomarkers could be a new screening and prevention tool for people with prior dysbiosis.
Collapse
Affiliation(s)
- William Bourumeau
- Département des Sciences Fondamentales, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada; (W.B.); (C.G.)
- Centre Intersectoriel en Santé Durable, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada;
| | - Karine Tremblay
- Pharmacology-Physiology Department, Université de Sherbrooke, Saguenay, QC J1K 2R1, Canada;
- Research Centre of Centre Intégré Universitaire de Santé et de Services Sociaux du Saguenay–Lac-Saint-Jean (CIUSSS-SLSJ), Saguenay, QC G7H 7K9, Canada
| | - Guillaume Jourdan
- Centre Intersectoriel en Santé Durable, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada;
| | - Catherine Girard
- Département des Sciences Fondamentales, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada; (W.B.); (C.G.)
- Centre Intersectoriel en Santé Durable, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada;
| | - Catherine Laprise
- Département des Sciences Fondamentales, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada; (W.B.); (C.G.)
- Centre Intersectoriel en Santé Durable, Université du Québec à Chicoutimi, Saguenay, QC G7H 2B1, Canada;
| |
Collapse
|
21
|
Brandenburg K, Ferrer-Espada R, Martinez-de-Tejada G, Nehls C, Fukuoka S, Mauss K, Weindl G, Garidel P. A Comparison between SARS-CoV-2 and Gram-Negative Bacteria-Induced Hyperinflammation and Sepsis. Int J Mol Sci 2023; 24:15169. [PMID: 37894850 PMCID: PMC10607443 DOI: 10.3390/ijms242015169] [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: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Sepsis is a life-threatening condition caused by the body's overwhelming response to an infection, such as pneumonia or urinary tract infection. It occurs when the immune system releases cytokines into the bloodstream, triggering widespread inflammation. If not treated, it can lead to organ failure and death. Unfortunately, sepsis has a high mortality rate, with studies reporting rates ranging from 20% to over 50%, depending on the severity and promptness of treatment. According to the World Health Organization (WHO), the annual death toll in the world is about 11 million. One of the main toxins responsible for inflammation induction are lipopolysaccharides (LPS, endotoxin) from Gram-negative bacteria, which rank among the most potent immunostimulants found in nature. Antibiotics are consistently prescribed as a part of anti-sepsis-therapy. However, antibiotic therapy (i) is increasingly ineffective due to resistance development and (ii) most antibiotics are unable to bind and neutralize LPS, a prerequisite to inhibit the interaction of endotoxin with its cellular receptor complex, namely Toll-like receptor 4 (TLR4)/MD-2, responsible for the intracellular cascade leading to pro-inflammatory cytokine secretion. The pandemic virus SARS-CoV-2 has infected hundreds of millions of humans worldwide since its emergence in 2019. The COVID-19 (Coronavirus disease-19) caused by this virus is associated with high lethality, particularly for elderly and immunocompromised people. As of August 2023, nearly 7 million deaths were reported worldwide due to this disease. According to some reported studies, upregulation of TLR4 and the subsequent inflammatory signaling detected in COVID-19 patients "mimics bacterial sepsis". Furthermore, the immune response to SARS-CoV-2 was described by others as "mirror image of sepsis". Similarly, the cytokine profile in sera from severe COVID-19 patients was very similar to those suffering from the acute respiratory distress syndrome (ARDS) and sepsis. Finally, the severe COVID-19 infection is frequently accompanied by bacterial co-infections, as well as by the presence of significant LPS concentrations. In the present review, we will analyze similarities and differences between COVID-19 and sepsis at the pathophysiological, epidemiological, and molecular levels.
Collapse
Affiliation(s)
- Klaus Brandenburg
- Brandenburg Antiinfektiva, c/o Forschungszentrum Borstel, Leibniz-Lungenzentrum, Parkallee 10, 23845 Borstel, Germany; (K.B.); (K.M.)
| | - Raquel Ferrer-Espada
- Department of Microbiology, University of Navarra, IdiSNA (Navarra Institute for Health Research), Irunlarrea 1, E-31008 Pamplona, Spain;
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Guillermo Martinez-de-Tejada
- Department of Microbiology, University of Navarra, IdiSNA (Navarra Institute for Health Research), Irunlarrea 1, E-31008 Pamplona, Spain;
| | - Christian Nehls
- Forschungszentrum Borstel, FG Biophysik, Parkallee 10, 23845 Borstel, Germany;
| | - Satoshi Fukuoka
- National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu 761-0395, Japan;
| | - Karl Mauss
- Brandenburg Antiinfektiva, c/o Forschungszentrum Borstel, Leibniz-Lungenzentrum, Parkallee 10, 23845 Borstel, Germany; (K.B.); (K.M.)
- Sylter Klinik Karl Mauss, Dr.-Nicolas-Strasse 3, 25980 Westerland (Sylt), Germany
| | - Günther Weindl
- Pharmazeutisches Institut, Abteilung Pharmakologie und Toxikologie, Universität Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany;
| | - Patrick Garidel
- Physikalische Chemie, Martin-Luther-Universität Halle-Wittenberg, 06108 Halle (Saale), Germany
| |
Collapse
|
22
|
Zheng A, Huang N, Bean D, Rayapaneni S, Deeney J, Sagar M, Hamilton JA. Resolvin E1 heals injured cardiomyocytes: Therapeutic implications and H-FABP as a readout for cardiovascular disease & systemic inflammation. Prostaglandins Leukot Essent Fatty Acids 2023; 197:102586. [PMID: 37604082 PMCID: PMC11203388 DOI: 10.1016/j.plefa.2023.102586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/23/2023]
Abstract
The purpose of this study is to investigate heart-fatty acid binding protein (H-FABP) leakage from cardiomyocytes as a quantitative measure of cell membrane damage and to test healing by Resolvin E1 (RVE1) as a potential therapeutic for patients with inflammatory diseases (cardiovascular disease and comorbidities) with high morbidity and mortality. Our quantitative ELISA assays demonstrated H-FABP as a sensitive and reliable biomarker for measuring cardiomyocyte damage induced by lipopolysaccharide (LPS) and healing by RvE1, a specialized pro-resolving mediator (SPM) derived from the Omega-3 fatty acid, eicosapentaenoic acid (EPA), a dietary nutrient that balances inflammation to restore homeostasis. RvE1 reduced leakage of H-FABP by up to 86%, which supports our hypothesis that inflammation as a mechanism of injury can be targeted for therapy. H-FABP as a blood biomarker was tested in 40 patients admitted to Boston Medical Center for respiratory distress, (20 patients with and 20 patients without COVID infection). High levels of H-FABP correlated with clinically diagnosed CVD, diabetes, and end-stage renal disease (ESRD) in both patient groups. The level of H-FABP indicates not only CVD damage but is a valuable measure for patients with increased inflammation disease comorbidities.
Collapse
Affiliation(s)
- A Zheng
- Boston University, United States of America
| | - N Huang
- Boston University School of Medicine, United States of America
| | - D Bean
- Boston University School of Medicine, United States of America
| | | | - Jude Deeney
- Boston University School of Medicine, United States of America
| | - M Sagar
- Boston Medical Center, United States of America
| | | |
Collapse
|
23
|
Isaza-Correa J, Ryan L, Kelly L, Allen J, Melo A, Jones J, Huggard D, Ryan E, Ó Maoldomhnaigh C, Geoghehan S, Gavin P, Leahy TR, Butler K, Freyne B, Molloy EJ. Innate immune dysregulation in multisystem inflammatory syndrome in children (MIS-C). Sci Rep 2023; 13:16463. [PMID: 37777557 PMCID: PMC10542373 DOI: 10.1038/s41598-023-43390-6] [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: 06/12/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023] Open
Abstract
MIS-C is a systemic inflammation disorder with poorly characterised immunopathological mechanisms. We compared changes in the systemic immune response in children with MIS-C (n = 12, 5-13 years) to healthy controls (n = 14, 5-15 years). Analysis was done in whole blood treated with LPS. Expression of CD11b and Toll-like receptor-4 (TLR4) in neutrophils and monocytes were analysed by flow cytometry. Serum cytokines (IL-1β, IL-2, IL-6, IL-8, IL-10, IL-Ira, TNF-α, TNF-β, IFN-Υ, VEGF, EPO and GM-CSF) and mRNA levels of inflammasome molecules (NLRP3, ASC and IL-1β) were evaluated. Subpopulations of lymphocytes (CD3+, CD19+, CD56+, CD4+, CD8+, TCR Vδ1+, TCR Vδ2+) were assessed at basal levels. Absolute counts of neutrophils and NLR were high in children with MIS-C while absolute counts of lymphocytes were low. Children with MIS-C had increased levels of IL-6, IL-10, TNF-β and VEGF serum cytokines at the basal level, and significantly increased TNF-β post-LPS, compared to controls. IL-1RA and EPO decreased at baseline and post-LPS in MIS-C patients compared to controls. The percentage of CD3+ cells, NK cells and Vδ1 was lower while B cells were higher in children with MIS-C than in controls. Dysregulated immune response in children with MIS-C was evident and may be amenable to immunomodulation.
Collapse
Affiliation(s)
- Johana Isaza-Correa
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | - Laura Ryan
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | - Lynne Kelly
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | - John Allen
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | - Ashanty Melo
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | - Jennifer Jones
- Infectious Diseases/Immunology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Dean Huggard
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | - Emer Ryan
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | | | - Sarah Geoghehan
- Infectious Diseases/Immunology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Patrick Gavin
- Infectious Diseases/Immunology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Timothy Ronan Leahy
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland
- Infectious Diseases/Immunology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Karina Butler
- Infectious Diseases/Immunology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Bridget Freyne
- Infectious Diseases/Immunology, Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland.
- Infectious Diseases/Immunology, Children's Health Ireland at Crumlin, Dublin, Ireland.
- Neonatology, Children's Health Ireland at Crumlin, Dublin, Ireland.
- Neurodisability, Children's Health Ireland at Tallaght, Dublin, Ireland.
- Neonatology, The Coombe Hospital, Dublin, Ireland.
- Discipline of Paediatrics, Trinity Centre for Health Sciences, Children's Hospital Ireland (CHI) at Tallaght, Tallaght University Hospital, Dublin 24, Ireland.
| |
Collapse
|
24
|
Plummer AM, Matos YL, Lin HC, Ryman SG, Birg A, Quinn DK, Parada AN, Vakhtin AA. Gut-brain pathogenesis of post-acute COVID-19 neurocognitive symptoms. Front Neurosci 2023; 17:1232480. [PMID: 37841680 PMCID: PMC10568482 DOI: 10.3389/fnins.2023.1232480] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/01/2023] [Indexed: 10/17/2023] Open
Abstract
Approximately one third of non-hospitalized coronavirus disease of 2019 (COVID-19) patients report chronic symptoms after recovering from the acute stage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Some of the most persistent and common complaints of this post-acute COVID-19 syndrome (PACS) are cognitive in nature, described subjectively as "brain fog" and also objectively measured as deficits in executive function, working memory, attention, and processing speed. The mechanisms of these chronic cognitive sequelae are currently not understood. SARS-CoV-2 inflicts damage to cerebral blood vessels and the intestinal wall by binding to angiotensin-converting enzyme 2 (ACE2) receptors and also by evoking production of high levels of systemic cytokines, compromising the brain's neurovascular unit, degrading the intestinal barrier, and potentially increasing the permeability of both to harmful substances. Such substances are hypothesized to be produced in the gut by pathogenic microbiota that, given the profound effects COVID-19 has on the gastrointestinal system, may fourish as a result of intestinal post-COVID-19 dysbiosis. COVID-19 may therefore create a scenario in which neurotoxic and neuroinflammatory substances readily proliferate from the gut lumen and encounter a weakened neurovascular unit, gaining access to the brain and subsequently producing cognitive deficits. Here, we review this proposed PACS pathogenesis along the gut-brain axis, while also identifying specific methodologies that are currently available to experimentally measure each individual component of the model.
Collapse
Affiliation(s)
- Allison M. Plummer
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Yvette L. Matos
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
| | - Henry C. Lin
- Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM, United States
- Section of Gastroenterology, New Mexico Veterans Affairs Health Care System, Albuquerque, NM, United States
| | - Sephira G. Ryman
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
- Nene and Jamie Koch Comprehensive Movement Disorder Center, Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| | - Aleksandr Birg
- Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM, United States
- Section of Gastroenterology, New Mexico Veterans Affairs Health Care System, Albuquerque, NM, United States
| | - Davin K. Quinn
- Department of Psychiatry and Behavioral Sciences, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Alisha N. Parada
- Division of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Andrei A. Vakhtin
- The Mind Research Network/Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, United States
| |
Collapse
|
25
|
Granato G, Gesmundo I, Pedrolli F, Kasarla R, Begani L, Banfi D, Bruno S, Lopatina T, Brizzi MF, Cai R, Sha W, Ghigo E, Schally AV, Granata R. Growth hormone-releasing hormone antagonist MIA-602 inhibits inflammation induced by SARS-CoV-2 spike protein and bacterial lipopolysaccharide synergism in macrophages and human peripheral blood mononuclear cells. Front Immunol 2023; 14:1231363. [PMID: 37649486 PMCID: PMC10462983 DOI: 10.3389/fimmu.2023.1231363] [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: 05/30/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
COVID-19 is characterized by an excessive inflammatory response and macrophage hyperactivation, leading, in severe cases, to alveolar epithelial injury and acute respiratory distress syndrome. Recent studies have reported that SARS-CoV-2 spike (S) protein interacts with bacterial lipopolysaccharide (LPS) to boost inflammatory responses in vitro, in macrophages and peripheral blood mononuclear cells (PBMCs), and in vivo. The hypothalamic hormone growth hormone-releasing hormone (GHRH), in addition to promoting pituitary GH release, exerts many peripheral functions, acting as a growth factor in both malignant and non-malignant cells. GHRH antagonists, in turn, display potent antitumor effects and antinflammatory activities in different cell types, including lung and endothelial cells. However, to date, the antinflammatory role of GHRH antagonists in COVID-19 remains unexplored. Here, we examined the ability of GHRH antagonist MIA-602 to reduce inflammation in human THP-1-derived macrophages and PBMCs stimulated with S protein and LPS combination. Western blot and immunofluorescence analysis revealed the presence of GHRH receptor and its splice variant SV1 in both THP-1 cells and PBMCs. Exposure of THP-1 cells to S protein and LPS combination increased the mRNA levels and protein secretion of TNF-α and IL-1β, as well as IL-8 and MCP-1 gene expression, an effect hampered by MIA-602. Similarly, MIA-602 hindered TNF-α and IL-1β secretion in PBMCs and reduced MCP-1 mRNA levels. Mechanistically, MIA-602 blunted the S protein and LPS-induced activation of inflammatory pathways in THP-1 cells, such as NF-κB, STAT3, MAPK ERK1/2 and JNK. MIA-602 also attenuated oxidative stress in PBMCs, by decreasing ROS production, iNOS and COX-2 protein levels, and MMP9 activity. Finally, MIA-602 prevented the effect of S protein and LPS synergism on NF-кB nuclear translocation and activity. Overall, these findings demonstrate a novel antinflammatory role for GHRH antagonists of MIA class and suggest their potential development for the treatment of inflammatory diseases, such as COVID-19 and related comorbidities.
Collapse
Affiliation(s)
- Giuseppina Granato
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Iacopo Gesmundo
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Francesca Pedrolli
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Ramesh Kasarla
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Laura Begani
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Dana Banfi
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Stefania Bruno
- Department of Medical Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Tatiana Lopatina
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Renzhi Cai
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States
- South Florida VA Foundation for Research and Education, Veterans Affairs Medical Center, Miami, FL, United States
| | - Wei Sha
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States
| | - Ezio Ghigo
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Andrew V. Schally
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, United States
- South Florida VA Foundation for Research and Education, Veterans Affairs Medical Center, Miami, FL, United States
- Department of Medicine, Divisions of Medical/Oncology and Endocrinology, and the Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Riccarda Granata
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Turin, Turin, Italy
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
26
|
Sfera A, Rahman L, Zapata-Martín Del Campo CM, Kozlakidis Z. Long COVID as a Tauopathy: Of "Brain Fog" and "Fusogen Storms". Int J Mol Sci 2023; 24:12648. [PMID: 37628830 PMCID: PMC10454863 DOI: 10.3390/ijms241612648] [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: 07/13/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Long COVID, also called post-acute sequelae of SARS-CoV-2, is characterized by a multitude of lingering symptoms, including impaired cognition, that can last for many months. This symptom, often called "brain fog", affects the life quality of numerous individuals, increasing medical complications as well as healthcare expenditures. The etiopathogenesis of SARS-CoV-2-induced cognitive deficit is unclear, but the most likely cause is chronic inflammation maintained by a viral remnant thriving in select body reservoirs. These viral sanctuaries are likely comprised of fused, senescent cells, including microglia and astrocytes, that the pathogen can convert into neurotoxic phenotypes. Moreover, as the enteric nervous system contains neurons and glia, the virus likely lingers in the gastrointestinal tract as well, accounting for the intestinal symptoms of long COVID. Fusogens are proteins that can overcome the repulsive forces between cell membranes, allowing the virus to coalesce with host cells and enter the cytoplasm. In the intracellular compartment, the pathogen hijacks the actin cytoskeleton, fusing host cells with each other and engendering pathological syncytia. Cell-cell fusion enables the virus to infect the healthy neighboring cells. We surmise that syncytia formation drives cognitive impairment by facilitating the "seeding" of hyperphosphorylated Tau, documented in COVID-19. In our previous work, we hypothesized that the SARS-CoV-2 virus induces premature endothelial senescence, increasing the permeability of the intestinal and blood-brain barrier. This enables the migration of gastrointestinal tract microbes and/or their components into the host circulation, eventually reaching the brain where they may induce cognitive dysfunction. For example, translocated lipopolysaccharides or microbial DNA can induce Tau hyperphosphorylation, likely accounting for memory problems. In this perspective article, we examine the pathogenetic mechanisms and potential biomarkers of long COVID, including microbial cell-free DNA, interleukin 22, and phosphorylated Tau, as well as the beneficial effect of transcutaneous vagal nerve stimulation.
Collapse
Affiliation(s)
- Adonis Sfera
- Paton State Hospital, 3102 Highland Ave, Patton, CA 92369, USA
- School of Behavioral Health, Loma Linda University, 11139 Anderson St., Loma Linda, CA 92350, USA
- Department of Psychiatry, University of California, Riverside 900 University Ave, Riverside, CA 92521, USA
| | - Leah Rahman
- Department of Neuroscience, University of Oregon, 222 Huestis Hall, Eugene, OR 97401, USA
| | | | - Zisis Kozlakidis
- International Agency for Research on Cancer, World Health Organization, 69000 Lyon, France
| |
Collapse
|
27
|
Guo C, Yi B, Wu J, Lu J. The microbiome in post-acute infection syndrome (PAIS). Comput Struct Biotechnol J 2023; 21:3904-3911. [PMID: 37602232 PMCID: PMC10432703 DOI: 10.1016/j.csbj.2023.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/03/2023] [Accepted: 08/03/2023] [Indexed: 08/22/2023] Open
Abstract
Post-Acute Infection Syndrome (PAIS) is a relatively new medical terminology that represents prolonged sequelae symptoms after acute infection by numerous pathogenic agents. Imposing a substantial public health burden worldwide, PASC (post-acute sequelae of COVID-19 infection) and ME/CFS (myalgic encephalomyelitis/chronic fatigue syndrome) are two of the most recognized and prevalent PAIS conditions. The presences of prior infections and similar symptom profiles in PAIS reflect a plausible common etiopathogenesis. The human microbiome is known to play an essential role in health and disease. In this review, we reviewed and summarized available research on oral and gut microbiota alterations in patients with different infections or PAIS conditions. We discussed key theories about the associations between microbiome dysbiosis and PAIS disease development, aiming to explore the mechanistic roles and potential functions the microbiome may have in the process. Additionally, we discuss the areas of knowledge gaps and propose the potential clinical applications of the microbiome for prevention and treatment of PAIS conditions.
Collapse
Affiliation(s)
- Cheng Guo
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, USA
| | - Boyang Yi
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Jianyong Wu
- School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Jiahai Lu
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
- Hainan Key Novel Thinktank “Hainan Medical University ‘One Health’ Research Center”, Haikou, China
| |
Collapse
|
28
|
Araújo JR, Serafim T, Ismael S, Calhau C, Faria A, Teixeira D. Intestinal Alkaline Phosphatase Activity and Efficiency Are Altered in Severe COVID-19 Patients. GASTRO HEP ADVANCES 2023; 2:911-917. [PMID: 39130768 PMCID: PMC11307804 DOI: 10.1016/j.gastha.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/11/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Although gut inflammation and dysbiosis have been implicated in the pathophysiology of severe cases of coronavirus disease 2019 (COVID-19), the role of intestinal anti-inflammatory enzymes, such as alkaline phosphatase, is still underexplored. Therefore, the aim of this study was to compare intestinal alkaline phosphatase (iALP) activity and its proinflammatory substrate - bacterial lipopolysaccharide (LPS) - concentration between mild-to-moderate and severe COVID-19 patients. Methods Stool samples collected from 53 mild-to-moderate and 57 severe adult COVID-19 patients, previously enrolled in a national multicentre cross-sectional study (NCT04355741), were analysed for iALP activity and LPS concentration. Results iALP activity decreased by 40% in severe compared to mild-to-moderate COVID-19 patients (median [interquartile range] of 120.6 [25.2-593.1] nmol pNP/min/g of protein vs 202.8 [102.1-676.1] nmol pNP/min/g of protein; P = .04) after adjustment for clinical and gut microbiota parameters. Regarding fecal LPS, its concentration was found to be decreased in severe patients (mean ± standard error of mean of 18,118 ± 1225 EU/g of feces vs 22,508 ± 1203 EU/g of feces; P = .01), although this parameter did not correlate with plasma levels of C-reactive protein (P = .08), a sensitive biomarker of systemic inflammation. In contrast, fecal ALP activity / LPS concentration ratio, an indicator of iALP efficiency, was found to be increased in severe compared to mild-to-moderate COVID-19 patients (P = .04). Conclusion Changes in iALP kinetic parameters found in severe COVID-19 patients may represent a potential mechanism to counterbalance alterations in gut homeostasis (eg inflammation and dysbiosis) associated with COVID-19 severity.
Collapse
Affiliation(s)
- João R. Araújo
- Nutrition & Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
- Nutrition & Metabolism, CINTESIS@RISE, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Thainá Serafim
- Nutrition & Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Shámila Ismael
- Nutrition & Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
- Nutrition & Metabolism, CINTESIS@RISE, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
- CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Conceição Calhau
- Nutrition & Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
- Nutrition & Metabolism, CINTESIS@RISE, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Ana Faria
- Nutrition & Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
- Nutrition & Metabolism, CINTESIS@RISE, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
- CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Diana Teixeira
- Nutrition & Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
- Nutrition & Metabolism, CINTESIS@RISE, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
- CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| |
Collapse
|
29
|
Jantsch J, Rodrigues FDS, Fraga GDF, Eller S, Silveira AK, Moreira JCF, Giovernardi M, Guedes RP. Calorie restriction mitigates metabolic, behavioral and neurochemical effects of cafeteria diet in aged male rats. J Nutr Biochem 2023:109371. [PMID: 37169228 DOI: 10.1016/j.jnutbio.2023.109371] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023]
Abstract
Besides metabolic dysfunctions, elderly individuals with obesity are at special risk of developing cognitive decline and psychiatric disturbances. Restricted calorie consumption could be an efficient strategy to improve metabolic function after obesity. However, its effects on anxiety-like behaviors in aged rats submitted to an obesogenic diet are unknown. For this purpose, 42 Wistar rats (18-months old) were divided into four groups: Control (CT), calorie restriction (CR), cafeteria diet (CAF), and CAF+CR (CAF/CR). CT, CR, and CAF groups received the diets for 8 weeks. CAF/CR group was submitted to the CAF menu for 7 weeks and then switched to a standard diet on a CR regimen, receiving 30% lower calories than consumed by the CT, for another 5 weeks. CAF's menu consisted of ultra-processed foods such as cookies, chocolate, sausage, and bologna. Body weight, visceral adiposity, and biochemical blood analysis were evaluated for obesity diagnosis. The profile of gut microbiota was investigated, along with circulating levels of LPS. Neurochemical parameters, such as neurotransmitter levels, were dosed. Anxiety-like behaviors were accessed using open field (OF) and elevated plus maze (EPM) tests. As expected, CR reduced weight gain and improved glucose homeostasis. Gut microbiome disturbance was found in CAF-fed animals accompanied by increased levels of LPS. However, CR after CAF mitigated several harmful responses. The obesogenic diet triggered anxiety-like manifestations in the OF and EPM tests that were not evidenced in the CAF/CR group. These findings indicate that CR can be a promising strategy for the neurological effects of obesity in aged rats.
Collapse
Affiliation(s)
- Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Gabriel de Farias Fraga
- Biomedical Science Undergraduate Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Sarah Eller
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Alexandre Kleber Silveira
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil
| | - José Cláudio Fonseca Moreira
- Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil
| | - Márcia Giovernardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil; Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, Brazil.
| |
Collapse
|
30
|
Kang JH, Jang M, Seo SJ, Choi A, Shin D, Seo S, Lee SH, Kim HN. Mechanobiological Adaptation to Hyperosmolarity Enhances Barrier Function in Human Vascular Microphysiological System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206384. [PMID: 36808839 PMCID: PMC10161024 DOI: 10.1002/advs.202206384] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/27/2023] [Indexed: 05/06/2023]
Abstract
In infectious disease such as sepsis and COVID-19, blood vessel leakage treatment is critical to prevent fatal progression into multi-organ failure and ultimately death, but the existing effective therapeutic modalities that improve vascular barrier function are limited. Here, this study reports that osmolarity modulation can significantly improve vascular barrier function, even in an inflammatory condition. 3D human vascular microphysiological systems and automated permeability quantification processes for high-throughput analysis of vascular barrier function are utilized. Vascular barrier function is enhanced by >7-folds with 24-48 h hyperosmotic exposure (time window of emergency care; >500 mOsm L-1 ) but is disrupted after hypo-osmotic exposure (<200 mOsm L-1 ). By integrating genetic and protein level analysis, it is shown that hyperosmolarity upregulates vascular endothelial-cadherin, cortical F-actin, and cell-cell junction tension, indicating that hyperosmotic adaptation mechanically stabilizes the vascular barrier. Importantly, improved vascular barrier function following hyperosmotic exposure is maintained even after chronic exposure to proinflammatory cytokines and iso-osmotic recovery via Yes-associated protein signaling pathways. This study suggests that osmolarity modulation may be a unique therapeutic strategy to proactively prevent infectious disease progression into severe stages via vascular barrier function protection.
Collapse
Affiliation(s)
- Joon Ho Kang
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Minjeong Jang
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Su Jin Seo
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Department of Chemical EngineeringKwangwoon UniversitySeoul01897Republic of Korea
| | - Andrew Choi
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
| | - Daeeun Shin
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- School of Mechanical EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
| | - Suyoung Seo
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Program in Nano Science and TechnologyGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul08826Republic of Korea
| | - Soo Hyun Lee
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and TechnologySeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolUniversity of Science and Technology (UST)Seoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
31
|
Grewal T, Buechler C. Adipokines as Diagnostic and Prognostic Markers for the Severity of COVID-19. Biomedicines 2023; 11:1302. [PMID: 37238973 PMCID: PMC10215701 DOI: 10.3390/biomedicines11051302] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Accumulating evidence implicates obesity as a risk factor for increased severity of disease outcomes in patients infected with severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). Obesity is associated with adipose tissue dysfunction, which not only predisposes individuals to metabolic complications, but also substantially contributes to low-grade systemic inflammation, altered immune cell composition, and compromised immune function. This seems to impact the susceptibility and outcome of diseases caused by viruses, as obese people appear more vulnerable to developing infections and they recover later from infectious diseases than normal-weight individuals. Based on these findings, increased efforts to identify suitable diagnostic and prognostic markers in obese Coronavirus disease 2019 (COVID-19) patients to predict disease outcomes have been made. This includes the analysis of cytokines secreted from adipose tissues (adipokines), which have multiple regulatory functions in the body; for instance, modulating insulin sensitivity, blood pressure, lipid metabolism, appetite, and fertility. Most relevant in the context of viral infections, adipokines also influence the immune cell number, with consequences for overall immune cell activity and function. Hence, the analysis of the circulating levels of diverse adipokines in patients infected with SARS-CoV-2 have been considered to reveal diagnostic and prognostic COVID-19 markers. This review article summarizes the findings aimed to correlate the circulating levels of adipokines with progression and disease outcomes of COVID-19. Several studies provided insights on chemerin, adiponectin, leptin, resistin, and galectin-3 levels in SARS-CoV-2-infected patients, while limited information is yet available on the adipokines apelin and visfatin in COVID-19. Altogether, current evidence points at circulating galectin-3 and resistin levels being of diagnostic and prognostic value in COVID-19 disease.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053 Regensburg, Germany
| |
Collapse
|
32
|
Zhu W, Wang M, Jin L, Yang B, Bai B, Mutsinze RN, Zuo W, Chattipakorn N, Huh JY, Liang G, Wang Y. Licochalcone A protects against LPS-induced inflammation and acute lung injury by directly binding with myeloid differentiation factor 2 (MD2). Br J Pharmacol 2023; 180:1114-1131. [PMID: 36480410 DOI: 10.1111/bph.15999] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a challenging clinical syndrome that leads to various respiratory sequelae and even high mortality in patients with severe disease. The novel pharmacological strategies and therapeutic drugs are urgently needed. Natural products have played a fundamental role and provided an abundant pool in drug discovery. EXPERIMENTAL APPROACH A compound library containing 160 natural products was used to screen potential anti-inflammatory compounds. Mice with LPS-induced ALI was then used to verify the preventive and therapeutic effects of the selected compounds. KEY RESULTS Licochalcone A was discovered from the anti-inflammatory screening of natural products in macrophages. A qPCR array validated the inflammation-regulatory effects of licochalcone A and indicated that the potential targets of licochalcone A may be the upstream proteins in LPS pro-inflammatory signalling. Further studies showed that licochalcone A directly binds to myeloid differentiation factor 2 (MD2), an assistant protein of toll-like receptor 4 (TLR4), to block both LPS-induced TRIF- and MYD88-dependent pathways. LEU61 and PHE151 in MD2 protein are the two key residues that contribute to the binding of MD2 to licochalcone A. In vivo, licochalcone A treatment alleviated ALI in LPS-challenged mice through significantly reducing immunocyte infiltration, suppressing activation of TLR4 pathway and inflammatory cytokine induction. CONCLUSION AND IMPLICATIONS In summary, our study identified MD2 as a direct target of licochalcone A for its anti-inflammatory activity and suggested that licochalcone A might serve as a novel MD2 inhibitor and a potential drug for developing ALI/ARDS therapy.
Collapse
Affiliation(s)
- Weiwei Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,College of Pharmacy, Chonnam National University, Gwangju, Korea
| | - Minxiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Leiming Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bin Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bin Bai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Rumbidzai Natasha Mutsinze
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Zuo
- Affiliated Xiangshan Hospital of Wenzhou Medical University (Xiangshan First People's Hospital Medical and Health Group), Xiangshan, China
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Joo Young Huh
- College of Pharmacy, Chonnam National University, Gwangju, Korea
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,College of Pharmacy, Chonnam National University, Gwangju, Korea.,Affiliated Xiangshan Hospital of Wenzhou Medical University (Xiangshan First People's Hospital Medical and Health Group), Xiangshan, China
| |
Collapse
|
33
|
Tsounis EP, Triantos C, Konstantakis C, Marangos M, Assimakopoulos SF. Intestinal barrier dysfunction as a key driver of severe COVID-19. World J Virol 2023; 12:68-90. [PMID: 37033148 PMCID: PMC10075050 DOI: 10.5501/wjv.v12.i2.68] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/08/2022] [Accepted: 01/16/2023] [Indexed: 03/21/2023] Open
Abstract
The intestinal lumen harbors a diverse consortium of microorganisms that participate in reciprocal crosstalk with intestinal immune cells and with epithelial and endothelial cells, forming a multi-layered barrier that enables the efficient absorption of nutrients without an excessive influx of pathogens. Despite being a lung-centered disease, severe coronavirus disease 2019 (COVID-19) affects multiple systems, including the gastrointestinal tract and the pertinent gut barrier function. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can inflict either direct cytopathic injury to intestinal epithelial and endothelial cells or indirect immune-mediated damage. Alternatively, SARS-CoV-2 undermines the structural integrity of the barrier by modifying the expression of tight junction proteins. In addition, SARS-CoV-2 induces profound alterations to the intestinal microflora at phylogenetic and metabolomic levels (dysbiosis) that are accompanied by disruption of local immune responses. The ensuing dysregulation of the gut-lung axis impairs the ability of the respiratory immune system to elicit robust and timely responses to restrict viral infection. The intestinal vasculature is vulnerable to SARS-CoV-2-induced endothelial injury, which simultaneously triggers the activation of the innate immune and coagulation systems, a condition referred to as “immunothrombosis” that drives severe thrombotic complications. Finally, increased intestinal permeability allows an aberrant dissemination of bacteria, fungi, and endotoxin into the systemic circulation and contributes, to a certain degree, to the over-exuberant immune responses and hyper-inflammation that dictate the severe form of COVID-19. In this review, we aim to elucidate SARS-CoV-2-mediated effects on gut barrier homeostasis and their implications on the progression of the disease.
Collapse
Affiliation(s)
- Efthymios P Tsounis
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Christos Konstantakis
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Markos Marangos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Patras 26504, Greece
| | - Stelios F Assimakopoulos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Patras 26504, Greece
| |
Collapse
|
34
|
Presepsin as a diagnostic and prognostic biomarker of severe bacterial infections and COVID-19. Sci Rep 2023; 13:3814. [PMID: 36882572 PMCID: PMC9990570 DOI: 10.1038/s41598-023-30807-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 03/01/2023] [Indexed: 03/09/2023] Open
Abstract
We aimed to develop presepsin as a marker of diagnosis of severe infections of either bacterial and viral origin. The derivation cohort was recruited from 173 hospitalized patients with acute pancreatitis or post-operative fever or infection suspicion aggravated by at least one sign of the quick sequential organ failure assessment (qSOFA). The first validation cohort was recruited from 57 admissions at the emergency department with at least one qSOFA sign and the second validation cohort from 115 patients with COVID-19 pneumonia. Presepsin was measured in plasma by the PATHFAST assay. Concentrations more than 350 pg/ml had sensitivity 80.2% for sepsis diagnosis in the derivation cohort (adjusted odds ratio 4.47; p < 0.0001). In the derivation cohort, sensitivity for 28-day mortality prognosis was 91.5% (adjusted odds ratio 6.82; p: 0.001). Concentrations above 350 pg/ml had sensitivity 93.3% for the diagnosis of sepsis in the first validation cohort; this was 78.3% in the second validation cohort of COVID-19 aiming at the early diagnosis of acute respiratory distress syndrome necessitating mechanical ventilation. The respective sensitivity for 28-day mortality was 85.7% and 92.3%. Presepsin may be a universal biomarker for the diagnosis of severe infections of bacterial origin and prediction of unfavorable outcome.
Collapse
|
35
|
Samsudin F, Raghuvamsi P, Petruk G, Puthia M, Petrlova J, MacAry P, Anand GS, Bond PJ, Schmidtchen A. SARS-CoV-2 spike protein as a bacterial lipopolysaccharide delivery system in an overzealous inflammatory cascade. J Mol Cell Biol 2023; 14:6761401. [PMID: 36240490 PMCID: PMC9940780 DOI: 10.1093/jmcb/mjac058] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/11/2022] [Indexed: 11/14/2022] Open
Abstract
Accumulating evidence indicates a potential role for bacterial lipopolysaccharide (LPS) in the overactivation of the immune response during SARS-CoV-2 infection. LPS is recognized by Toll-like receptor 4, mediating proinflammatory effects. We previously reported that LPS directly interacts with SARS-CoV-2 spike (S) protein and enhances proinflammatory activities. Using native gel electrophoresis and hydrogen-deuterium exchange mass spectrometry, we showed that LPS binds to multiple hydrophobic pockets spanning both the S1 and S2 subunits of the S protein. Molecular simulations validated by a microscale thermophoresis binding assay revealed that LPS binds to the S2 pocket with a lower affinity compared to S1, suggesting a role as an intermediate in LPS transfer. Congruently, nuclear factor-kappa B (NF-κB) activation in monocytic THP-1 cells is strongly boosted by S2. Using NF-κB reporter mice followed by bioimaging, a boosting effect was observed for both S1 and S2, with the former potentially facilitated by proteolysis. The Omicron S variant binds to LPS, but with reduced affinity and LPS boosting in vitro and in vivo. Taken together, the data provide a molecular mechanism by which S protein augments LPS-mediated hyperinflammation.
Collapse
Affiliation(s)
- Firdaus Samsudin
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore
| | - Palur Raghuvamsi
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Ganna Petruk
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Manoj Puthia
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Jitka Petrlova
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden
| | - Paul MacAry
- Life Sciences Institute, Centre for Life Sciences, National University of Singapore, Singapore 117546, Singapore
| | - Ganesh S Anand
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.,Department of Chemistry, The Pennsylvania State University, PA 16801, USA
| | - Peter J Bond
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Artur Schmidtchen
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-22184 Lund, Sweden.,Copenhagen Wound Healing Center, Bispebjerg Hospital, Department of Biomedical Sciences, University of Copenhagen, DK-2400 Copenhagen, Denmark
| |
Collapse
|
36
|
Dorneles GP, Teixeira PC, Peres A, Rodrigues Júnior LC, da Fonseca SG, Monteiro MC, Eller S, Oliveira TF, Wendland EM, Romão PRT. Endotoxin tolerance and low activation of TLR-4/NF-κB axis in monocytes of COVID-19 patients. J Mol Med (Berl) 2023; 101:183-195. [PMID: 36790534 PMCID: PMC9930695 DOI: 10.1007/s00109-023-02283-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/20/2022] [Accepted: 01/03/2023] [Indexed: 02/16/2023]
Abstract
Higher endotoxin in the circulation may indicate a compromised state of host immune response against coinfections in severe COVID-19 patients. We evaluated the inflammatory response of monocytes from COVID-19 patients after lipopolysaccharide (LPS) challenge. Whole blood samples of healthy controls, patients with mild COVID-19, and patients with severe COVID-19 were incubated with LPS for 2 h. Severe COVID-19 patients presented higher LPS and sCD14 levels in the plasma than healthy controls and mild COVID-19 patients. In non-stimulated in vitro condition, severe COVID-19 patients presented higher inflammatory cytokines and PGE-2 levels and CD14 + HLA-DRlow monocytes frequency than controls. Moreover, severe COVID-19 patients presented higher NF-κB p65 phosphorylation in CD14 + HLA-DRlow, as well as higher expression of TLR-4 and NF-κB p65 phosphorylation in CD14 + HLA-DRhigh compared to controls. The stimulation of LPS in whole blood of severe COVID-19 patients leads to lower cytokine production but higher PGE-2 levels compared to controls. Endotoxin challenge with both concentrations reduced the frequency of CD14 + HLA-DRlow in severe COVID-19 patients, but the increases in TLR-4 expression and NF-κB p65 phosphorylation were more pronounced in both CD14 + monocytes of healthy controls and mild COVID-19 patients compared to severe COVID-19 group. We conclude that acute SARS-CoV-2 infection is associated with diminished endotoxin response in monocytes. KEY MESSAGES: Severe COVID-19 patients had higher levels of LPS and systemic IL-6 and TNF-α. Severe COVID-19 patients presented higher CD14+HLA-DRlow monocytes. Increased TLR-4/NF-κB axis was identified in monocytes of severe COVID-19. Blunted production of cytokines after whole blood LPS stimulation in severe COVID-19. Lower TLR-4/NF-κB activation in monocytes after LPS stimulation in severe COVID-19.
Collapse
Affiliation(s)
- Gilson P Dorneles
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, Porto Alegre, RS, 245, 90050-170, Brazil
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Paula C Teixeira
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, Porto Alegre, RS, 245, 90050-170, Brazil
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Alessandra Peres
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, Porto Alegre, RS, 245, 90050-170, Brazil
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Luiz Carlos Rodrigues Júnior
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, Porto Alegre, RS, 245, 90050-170, Brazil
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | | | - Marta Chagas Monteiro
- Graduate Program in Pharmaceutical Science, Health Science Institute, Universidade Federal Do Pará, Belém, Pará, Brazil
| | - Sarah Eller
- Pharmacosciences Department, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Tiago F Oliveira
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Eliana M Wendland
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Pediatrics, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Pedro R T Romão
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, Porto Alegre, RS, 245, 90050-170, Brazil.
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
37
|
Wadowski PP, Panzer B, Józkowicz A, Kopp CW, Gremmel T, Panzer S, Koppensteiner R. Microvascular Thrombosis as a Critical Factor in Severe COVID-19. Int J Mol Sci 2023; 24:2492. [PMID: 36768817 PMCID: PMC9916726 DOI: 10.3390/ijms24032492] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Platelet-endothelial interactions have a critical role in microcirculatory function, which maintains tissue homeostasis. The subtle equilibrium between platelets and the vessel wall is disturbed by the coronavirus disease 2019 (COVID-19), which affects all three components of Virchow's triad (endothelial injury, stasis and a hypercoagulable state). Endotheliitis, vasculitis, glycocalyx degradation, alterations in blood flow and viscosity, neutrophil extracellular trap formation and microparticle shedding are only few pathomechanisms contributing to endothelial damage and microthrombosis resulting in capillary plugging and tissue ischemia. In the following opinion paper, we discuss major pathological processes leading to microvascular endothelial activation and thrombosis formation as a possible major adverse factor driving the deterioration of patient disease course in severe COVID-19.
Collapse
Affiliation(s)
- Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
- Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Benjamin Panzer
- Department of Cardiology, Wilhelminenspital, 1160 Vienna, Austria
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Gremmel
- Institute of Antithrombotic Therapy in Cardiovascular Disease, Karl Landsteiner Society, 3100 St. Pölten, Austria
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
| | - Simon Panzer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
38
|
Bacorn M, Romero-Soto HN, Levy S, Chen Q, Hourigan SK. The Gut Microbiome of Children during the COVID-19 Pandemic. Microorganisms 2022; 10:microorganisms10122460. [PMID: 36557713 PMCID: PMC9783902 DOI: 10.3390/microorganisms10122460] [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: 11/19/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
The gut microbiome has been shown to play a critical role in maintaining a healthy state. Dysbiosis of the gut microbiome is involved in modulating disease severity and potentially contributes to long-term outcomes in adults with COVID-19. Due to children having a significantly lower risk of severe illness and limited sample availability, much less is known about the role of the gut microbiome in children with COVID-19. It is well recognized that the developing gut microbiome of children differs from that of adults, but it is unclear if this difference contributes to the different clinical presentations and complications. In this review, we discuss the current knowledge of the gut microbiome in children with COVID-19, with gut microbiome dysbiosis being found in pediatric COVID-19 but specific taxa change often differing from those described in adults. Additionally, we discuss possible mechanisms of how the gut microbiome may mediate the presentation and complications of COVID-19 in children and the potential role for microbial therapeutics.
Collapse
|
39
|
Winzig F, Gandhi S, Lee A, Würstle S, Stanley GL, Capuano I, Neuringer I, Koff JL, Turner PE, Chan BK. Inhaled Bacteriophage Therapy for Multi-Drug Resistant Achromobacter. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2022; 95:413-427. [PMID: 36568830 PMCID: PMC9765334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The rise of antimicrobial resistant (AMR) bacteria is a global public health threat. AMR Achromobacter bacteria pose a challenging clinical problem, particularly for those with cystic fibrosis (CF) who are predisposed to chronic bacterial lung infections. Lytic bacteriophages (phages) offer a potential alternative to treat AMR infections, with the possible benefit that phage selection for resistance in target bacteria might coincide with reduced pathogenicity. The result is a genetic "trade-off," such as increased sensitivity to chemical antibiotics, and/or decreased virulence of surviving bacteria that are phage resistant. Here, we show that two newly discovered lytic phages against Achromobacter were associated with stabilization of respiratory status when deployed to treat a chronic pulmonary infection in a CF patient using inhaled (nebulized) phage therapy. The two phages demonstrate traits that could be generally useful in their development as therapeutics, especially the possibility that the phages can select for clinically useful trade-offs if bacteria evolve phage resistance following therapy. We discuss the limitations of the current study and suggest further work that should explore whether the phages could be generally useful in targeting pulmonary or other Achromobacter infections in CF patients.
Collapse
Affiliation(s)
- Franziska Winzig
- Department of Ecology and Evolutionary Biology, Yale
University, New Haven, CT, USA
- Center for Phage Biology & Therapy, Yale
University, New Haven, CT, USA
- Technische Universität München, München, Germany
| | - Shiv Gandhi
- Department of Ecology and Evolutionary Biology, Yale
University, New Haven, CT, USA
- Center for Phage Biology & Therapy, Yale
University, New Haven, CT, USA
- Department of Internal Medicine, Section of Infectious
Disease, Yale School of Medicine, New Haven, CT, USA
| | - Alina Lee
- Department of Ecology and Evolutionary Biology, Yale
University, New Haven, CT, USA
- Center for Phage Biology & Therapy, Yale
University, New Haven, CT, USA
| | - Silvia Würstle
- Department of Ecology and Evolutionary Biology, Yale
University, New Haven, CT, USA
- Center for Phage Biology & Therapy, Yale
University, New Haven, CT, USA
- Department of Internal Medicine II, University Hospital
rechts der Isar, School of Medicine, Technische Universität München, München,
Germany
| | - Gail L. Stanley
- Center for Phage Biology & Therapy, Yale
University, New Haven, CT, USA
- Department of Internal Medicine, Section of Pulmonary,
Critical Care, & Sleep Medicine, Yale School of Medicine, New Haven, CT,
USA
| | - Isabella Capuano
- Department of Internal Medicine, Section of Pulmonary,
Critical Care, & Sleep Medicine, Yale School of Medicine, New Haven, CT,
USA
- Cornell University, Ithaca, NY, USA
| | | | - Jonathan L. Koff
- Center for Phage Biology & Therapy, Yale
University, New Haven, CT, USA
- Department of Internal Medicine, Section of Pulmonary,
Critical Care, & Sleep Medicine, Yale School of Medicine, New Haven, CT,
USA
- To whom all correspondence should be addressed:
Paul E. Turner, ; ORCID:
https://www.orcid.org/0000-0003-3490-7498. Benjamin K. Chan,
. Jonathan L. Koff,
| | - Paul E. Turner
- Department of Ecology and Evolutionary Biology, Yale
University, New Haven, CT, USA
- Center for Phage Biology & Therapy, Yale
University, New Haven, CT, USA
- Program in Microbiology, Yale School of Medicine, New
Haven, CT, USA
- To whom all correspondence should be addressed:
Paul E. Turner, ; ORCID:
https://www.orcid.org/0000-0003-3490-7498. Benjamin K. Chan,
. Jonathan L. Koff,
| | - Benjamin K. Chan
- Department of Ecology and Evolutionary Biology, Yale
University, New Haven, CT, USA
- Center for Phage Biology & Therapy, Yale
University, New Haven, CT, USA
- To whom all correspondence should be addressed:
Paul E. Turner, ; ORCID:
https://www.orcid.org/0000-0003-3490-7498. Benjamin K. Chan,
. Jonathan L. Koff,
| |
Collapse
|
40
|
Long COVID and the Neuroendocrinology of Microbial Translocation Outside the GI Tract: Some Treatment Strategies. ENDOCRINES 2022. [DOI: 10.3390/endocrines3040058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Similar to previous pandemics, COVID-19 has been succeeded by well-documented post-infectious sequelae, including chronic fatigue, cough, shortness of breath, myalgia, and concentration difficulties, which may last 5 to 12 weeks or longer after the acute phase of illness. Both the psychological stress of SARS-CoV-2 infection and being diagnosed with COVID-19 can upregulate cortisol, a stress hormone that disrupts the efferocytosis effectors, macrophages, and natural killer cells, leading to the excessive accumulation of senescent cells and disruption of biological barriers. This has been well-established in cancer patients who often experience unrelenting fatigue as well as gut and blood–brain barrier dysfunction upon treatment with senescence-inducing radiation or chemotherapy. In our previous research from 2020 and 2021, we linked COVID-19 to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) via angiotensin II upregulation, premature endothelial senescence, intestinal barrier dysfunction, and microbial translocation from the gastrointestinal tract into the systemic circulation. In 2021 and 2022, these hypotheses were validated and SARS-CoV-2-induced cellular senescence as well as microbial translocation were documented in both acute SARS-CoV-2 infection, long COVID, and ME/CFS, connecting intestinal barrier dysfunction to disabling fatigue and specific infectious events. The purpose of this narrative review is to summarize what is currently known about host immune responses to translocated gut microbes and how these responses relate to fatiguing illnesses, including long COVID. To accomplish this goal, we examine the role of intestinal and blood–brain barriers in long COVID and other illnesses typified by chronic fatigue, with a special emphasis on commensal microbes functioning as viral reservoirs. Furthermore, we discuss the role of SARS-CoV-2/Mycoplasma coinfection in dysfunctional efferocytosis, emphasizing some potential novel treatment strategies, including the use of senotherapeutic drugs, HMGB1 inhibitors, Toll-like receptor 4 (TLR4) blockers, and membrane lipid replacement.
Collapse
|
41
|
Assante G, Tourna A, Carpani R, Ferrari F, Prati D, Peyvandi F, Blasi F, Bandera A, Le Guennec A, Chokshi S, Patel VC, Cox IJ, Valenti L, Youngson NA. Reduced circulating FABP2 in patients with moderate to severe COVID-19 may indicate enterocyte functional change rather than cell death. Sci Rep 2022; 12:18792. [PMID: 36335131 PMCID: PMC9637119 DOI: 10.1038/s41598-022-23282-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022] Open
Abstract
The gut is of importance in the pathology of COVID-19 both as a route of infection, and gut dysfunction influencing the severity of disease. Systemic changes caused by SARS-CoV-2 gut infection include alterations in circulating levels of metabolites, nutrients and microbial products which alter immune and inflammatory responses. Circulating plasma markers for gut inflammation and damage such as zonulin, lipopolysaccharide and β-glycan increase in plasma along with severity of disease. However, Intestinal Fatty Acid Binding Protein / Fatty Acid Binding Protein 2 (I-FABP/FABP2), a widely used biomarker for gut cell death, has paradoxically been shown to be reduced in moderate to severe COVID-19. We also found this pattern in a pilot cohort of mild (n = 18) and moderately severe (n = 19) COVID-19 patients in Milan from March to June 2020. These patients were part of the first phase of COVID-19 in Europe and were therefore all unvaccinated. After exclusion of outliers, patients with more severe vs milder disease showed reduced FABP2 levels (median [IQR]) (124 [368] vs. 274 [558] pg/mL, P < 0.01). A reduction in NMR measured plasma relative lipid-CH3 levels approached significance (median [IQR]) (0.081 [0.011] vs. 0.073 [0.024], P = 0.06). Changes in circulating lipid levels are another feature commonly observed in severe COVID-19 and a weak positive correlation was observed in the more severe group between reduced FABP2 and reduced relative lipid-CH3 and lipid-CH2 levels. FABP2 is a key regulator of enterocyte lipid import, a process which is inhibited by gut SARS-CoV-2 infection. We propose that the reduced circulating FABP2 in moderate to severe COVID-19 is a marker of infected enterocyte functional change rather than gut damage, which could also contribute to the development of hypolipidemia in patients with more severe disease.
Collapse
Affiliation(s)
- G Assante
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - A Tourna
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - R Carpani
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - F Ferrari
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - D Prati
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - F Peyvandi
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - F Blasi
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - A Bandera
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - A Le Guennec
- Randall Centre for Cell & Molecular Biophysics, King's College, London, UK
| | - S Chokshi
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - V C Patel
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
- Institute of Liver Studies, King's College Hospital, London, UK
| | - I J Cox
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK.
- Faculty of Life Sciences & Medicine, King's College, London, UK.
| | - L Valenti
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy.
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy.
| | - N A Youngson
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK.
- Faculty of Life Sciences & Medicine, King's College, London, UK.
| |
Collapse
|
42
|
Kiselevskiy MV, Anisimova NY, Bilan MI, Usov AI, Ustyuzhanina NE, Petkevich AA, Shubina IZ, Morozevich GE, Nifantiev NE. Prospects for the Use of Marine Sulfated Fucose-Rich Polysaccharides in Treatment and Prevention of COVID-19 and Post-COVID-19 Syndrome. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022; 48:1109-1122. [PMID: 36325402 PMCID: PMC9584273 DOI: 10.1134/s1068162022060152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/18/2022] [Accepted: 07/29/2022] [Indexed: 01/03/2023]
Abstract
Symptoms of the new coronavirus infection that appeared in 2019 (COVID-19) range from low fever and fatigue to acute pneumonia and multiple organ failure. The clinical picture of COVID-19 is heterogeneous and involves most physiological systems; therefore, drugs with a wide spectrum of mechanism of action are required. The choice of the treatment strategy for post-COVID-19 syndrome is still a challenge to be resolved. Polysaccharides with a high fucose content derived from seaweed and marine animals can form the basis for the subsequent development of promising agents for the treatment of COVID-19 and post-COVID-19 syndrome. This class of biopolymers is characterized by a variety of biological activities, including antiviral, antithrombotic, anticoagulant, hemo-stimulating, anti-inflammatory and immune-regulatory. Low molecular weight derivatives of these polysaccharides, as well as synthetic oligosaccharides with a sufficient amount and sulfation type may be considered as the most promising compounds due to their better bioavailability, which undoubtedly increases their therapeutic potential.
Collapse
Affiliation(s)
- M. V. Kiselevskiy
- Blokhin National Medical Research Center of Oncology, 115552 Moscow, Russia
| | - N. Yu. Anisimova
- Blokhin National Medical Research Center of Oncology, 115552 Moscow, Russia
| | - M. I. Bilan
- Laboratory of Glycoconjugate Chemistry, Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A. I. Usov
- Laboratory of Glycoconjugate Chemistry, Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia
| | - N. E. Ustyuzhanina
- Laboratory of Glycoconjugate Chemistry, Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A. A. Petkevich
- Blokhin National Medical Research Center of Oncology, 115552 Moscow, Russia
| | - I. Zh. Shubina
- Blokhin National Medical Research Center of Oncology, 115552 Moscow, Russia
| | - G. E. Morozevich
- Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia
| | - N. E. Nifantiev
- Laboratory of Glycoconjugate Chemistry, Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
43
|
Körper S, Schrezenmeier EV, Rincon-Arevalo H, Grüner B, Zickler D, Weiss M, Wiesmann T, Zacharowski K, Kalbhenn J, Bentz M, Dollinger MM, Paul G, Lepper PM, Ernst L, Wulf H, Zinn S, Appl T, Jahrsdörfer B, Rojewski M, Lotfi R, Dörner T, Jungwirth B, Seifried E, Fürst D, Schrezenmeier H. Cytokine levels associated with favorable clinical outcome in the CAPSID randomized trial of convalescent plasma in patients with severe COVID-19. Front Immunol 2022; 13:1008438. [PMID: 36275695 PMCID: PMC9582990 DOI: 10.3389/fimmu.2022.1008438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/12/2022] [Indexed: 12/03/2022] Open
Abstract
Objectives To determine the profile of cytokines in patients with severe COVID-19 who were enrolled in a trial of COVID-19 convalescent plasma (CCP). Methods Patients were randomized to receive standard treatment and 3 CCP units or standard treatment alone (CAPSID trial, ClinicalTrials.gov NCT04433910). The primary outcome was a dichotomous composite outcome (survival and no longer severe COVID-19 on day 21). Time to clinical improvement was a key secondary endpoint. The concentrations of 27 cytokines were measured (baseline, day 7). We analyzed the change and the correlation between serum cytokine levels over time in different subgroups and the prediction of outcome in receiver operating characteristics (ROC) analyses and in multivariate models. Results The majority of cytokines showed significant changes from baseline to day 7. Some were strongly correlated amongst each other (at baseline the cluster IL-1ß, IL-2, IL-6, IL-8, G-CSF, MIP-1α, the cluster PDGF-BB, RANTES or the cluster IL-4, IL-17, Eotaxin, bFGF, TNF-α). The correlation matrix substantially changed from baseline to day 7. The heatmaps of the absolute values of the correlation matrix indicated an association of CCP treatment and clinical outcome with the cytokine pattern. Low levels of IP-10, IFN-γ, MCP-1 and IL-1ß on day 0 were predictive of treatment success in a ROC analysis. In multivariate models, low levels of IL-1ß, IFN-γ and MCP-1 on day 0 were significantly associated with both treatment success and shorter time to clinical improvement. Low levels of IP-10, IL-1RA, IL-6, MCP-1 and IFN-γ on day 7 and high levels of IL-9, PDGF and RANTES on day 7 were predictive of treatment success in ROC analyses. Low levels of IP-10, MCP-1 and high levels of RANTES, on day 7 were associated with both treatment success and shorter time to clinical improvement in multivariate models. Conclusion This analysis demonstrates a considerable dynamic of cytokines over time, which is influenced by both treatment and clinical course of COVID-19. Levels of IL-1ß and MCP-1 at baseline and MCP-1, IP-10 and RANTES on day 7 were associated with a favorable outcome across several endpoints. These cytokines should be included in future trials for further evaluation as predictive factors.
Collapse
Affiliation(s)
- Sixten Körper
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Eva Vanessa Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health Charité Universitätsmedizin Berlin, Berlin Institute of Health (BIH) Academy, Berlin, Germany
| | - Hector Rincon-Arevalo
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Instituto de Investigaciones Médicas, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Beate Grüner
- Division of Infectious Diseases, University Hospital and Medical Center Ulm, Ulm, Germany
| | - Daniel Zickler
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Manfred Weiss
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Thomas Wiesmann
- Department of Anaesthesiology and Intensive Care Medicine, Phillips-University Marburg, Marburg, Germany
| | - Kai Zacharowski
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Johannes Kalbhenn
- Clinic of Anesthesiology and Intensive Care Medicine University Medical Center of Freiburg, Freiburg, Germany
| | - Martin Bentz
- Department of Internal Medicine III, Hospital of Karlsruhe, Karlsruhe, Germany
| | | | - Gregor Paul
- Department of Gastroenterology, Hepatology, Pneumology and Infectious Diseases, Klinikum Stuttgart, Stuttgart, Germany
| | - Philipp M. Lepper
- Department of Internal Medicine V – Pneumology, Allergology, Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Lucas Ernst
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hinnerk Wulf
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Sebastian Zinn
- Department of Anaesthesiology and Intensive Care Medicine, Phillips-University Marburg, Marburg, Germany
| | - Thomas Appl
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Bernd Jahrsdörfer
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Markus Rojewski
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Ramin Lotfi
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany
| | - Bettina Jungwirth
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Erhard Seifried
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen, Frankfurt, Germany
| | - Daniel Fürst
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
- *Correspondence: Hubert Schrezenmeier,
| |
Collapse
|
44
|
Postiga IAL, Teixeira PC, Neves CAM, Santana Filho P, Marmett B, Carvalho R, Peres A, Rotta L, Thompson CE, Dorneles GP, Romão PRT. Systemic redox imbalance in severe COVID-19 patients. Cell Biochem Funct 2022; 40:694-705. [PMID: 35980161 PMCID: PMC9538604 DOI: 10.1002/cbf.3735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/19/2022] [Accepted: 07/27/2022] [Indexed: 11/11/2022]
Abstract
The aim of this study was to evaluate the systemic redox state and inflammatory markers in intensive care unit (ICU) or non-ICU severe COVID-19 patients during the hospitalization period. Blood samples were collected at hospital admission (T1) (Controls and COVID-19 patients), 5-7 days after admission (T2: 5-7 days after hospital admission), and at the discharge time from the hospital (T3: 0-72 h before leaving hospital or death) to analyze systemic oxidative stress markers and inflammatory variables. The reactive oxygen species (ROS) production and mitochondrial membrane potential (MMP) were analyzed in peripheral granulocytes and monocytes. THP-1 human monocytic cell line was incubated with plasma from non-ICU and ICU COVID-19 patients and cell viability and apoptosis rate were analyzed. Higher total antioxidant capacity, protein oxidation, lipid peroxidation, and IL-6 at hospital admission were identified in both non-ICU and ICU COVID-19 patients. ICU COVID-19 patients presented increased C-reactive protein, ROS levels, and protein oxidation over hospitalization period compared to non-ICU patients, despite increased antioxidant status. Granulocytes and monocytes of non-ICU and ICU COVID-19 patients presented lower MMP and higher ROS production compared to the healthy controls, with the highest values found in ICU COVID-19 group. Finally, the incubation of THP-1 cells with plasma acquired from ICU COVID-19 patients at T3 hospitalization period decreased cell viability and apoptosis rate. In conclusion, disturbance in redox state is a hallmark of severe COVID-19 and is associated with cell damage and death.
Collapse
Affiliation(s)
- Isabelle A L Postiga
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Paula C Teixeira
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Carla Andretta Moreira Neves
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Paulo Santana Filho
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Bruna Marmett
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Roseana Carvalho
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Alessandra Peres
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Liane Rotta
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Claudia Elizabeth Thompson
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Gilson P Dorneles
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Pedro R T Romão
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
45
|
Hassel B, Niehusmann P, Halvorsen B, Dahlberg D. Pro-inflammatory cytokines in cystic glioblastoma: A quantitative study with a comparison with bacterial brain abscesses. With an MRI investigation of displacement and destruction of the brain tissue surrounding a glioblastoma. Front Oncol 2022; 12:846674. [PMID: 35965529 PMCID: PMC9372434 DOI: 10.3389/fonc.2022.846674] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Cystic glioblastomas are aggressive primary brain tumors that may both destroy and displace the surrounding brain tissue as they grow. The mechanisms underlying these tumors’ destructive effect could include exposure of brain tissue to tumor-derived cytokines, but quantitative cytokine data are lacking. Here, we provide quantitative data on leukocyte markers and cytokines in the cyst fluid from 21 cystic glioblastomas, which we compare to values in 13 brain abscess pus samples. The concentration of macrophage/microglia markers sCD163 and MCP-1 was higher in glioblastoma cyst fluid than in brain abscess pus; lymphocyte marker sCD25 was similar in cyst fluid and pus, whereas neutrophil marker myeloperoxidase was higher in pus. Median cytokine levels in glioblastoma cyst fluid were high (pg/mL): TNF-α: 32, IL-6: 1064, IL-8: 23585, tissue factor: 28, the chemokine CXCL1: 639. These values were not significantly different from values in pus, pointing to a highly pro-inflammatory glioblastoma environment. In contrast, levels of IFN-γ, IL-1β, IL-2, IL-4, IL-10, IL-12, and IL-13 were higher in pus than in glioblastoma cyst fluid. Based on the quantitative data, we show for the first time that the concentrations of cytokines in glioblastoma cyst fluid correlate with blood leukocyte levels, suggesting an important interaction between glioblastomas and the circulation. Preoperative MRI of the cystic glioblastomas confirmed both destruction and displacement of brain tissue, but none of the cytokine levels correlated with degree of brain tissue displacement or peri-tumoral edema, as could be assessed by MRI. We conclude that cystic glioblastomas are highly pro-inflammatory environments that interact with the circulation and that they both displace and destroy brain tissue. These observations point to the need for neuroprotective strategies in glioblastoma therapy, which could include an anti-inflammatory approach.
Collapse
Affiliation(s)
- Bjørnar Hassel
- Department of Neurohabilitation, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Defence Research Establishment (FFI), Kjeller, Norway
- *Correspondence: Bjørnar Hassel,
| | - Pitt Niehusmann
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Daniel Dahlberg
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
46
|
Romão PR, Teixeira PC, Schipper L, da Silva I, Santana Filho P, Júnior LCR, Peres A, Gonçalves da Fonseca S, Chagas Monteiro M, Lira FS, Andrey Cipriani Frade M, Comerlato J, Comerlato C, Sant'Anna FH, Bessel M, Abreu CM, Wendland EM, Dorneles GP. Viral load is associated with mitochondrial dysfunction and altered monocyte phenotype in acute severe SARS-CoV-2 infection. Int Immunopharmacol 2022; 108:108697. [PMID: 35405594 PMCID: PMC8920784 DOI: 10.1016/j.intimp.2022.108697] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/21/2022] [Accepted: 03/10/2022] [Indexed: 02/07/2023]
Abstract
Monocytes play a major role in the initial innate immune response to SARS-CoV-2. Although viral load may correlate with several clinical outcomes in COVID-19, much less is known regarding their impact on innate immune phenotype. We evaluated the monocyte phenotype and mitochondrial function in severe COVID-19 patients (n = 22) with different viral burden (determined by the median of viral load of the patients) at hospital admission. Severe COVID-19 patients presented lower frequency of CD14 + CD16- classical monocytes and CD39 expression on CD14 + monocytes, and higher frequency of CD14 + CD16 + intermediate and CD14-CD16 + nonclassical monocytes as compared to healthy controls independently of viral load. COVID-19 patients with high viral load exhibited increased GM-CSF, PGE-2 and lower IFN-α as compared to severe COVID-19 patients with low viral load (p < 0.05). CD14 + monocytes of COVID-19 patients with high viral load presented higher expression of PD-1 but lower HLA-DR on the cell surface than severe COVID-19 patients with low viral load. All COVID-19 patients presented decreased monocyte mitochondria membrane polarization, but high SARS-CoV-2 viral load was associated with increased mitochondrial reactive oxygen species. In this sense, higher viral load induces mitochondrial reactive oxygen species generation associated with exhaustion profile in CD14 + monocytes of severe COVID-19 patients. Altogether, these data shed light on new pathological mechanisms involving SARS-CoV-2 viral load on monocyte activation and mitochondrial function, which were associated with COVID-19 severity.
Collapse
Affiliation(s)
- Pedro Rt Romão
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| | - Paula C Teixeira
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Lucas Schipper
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Igor da Silva
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Paulo Santana Filho
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Luiz Carlos Rodrigues Júnior
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Alessandra Peres
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | | | - Marta Chagas Monteiro
- Graduate Program in Pharmaceutical Science, Health Science Institute, Federal University of Pará/UFPA, Belém, Pará, Brazil
| | - Fabio S Lira
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente 19060-900, SP, Brazil
| | - Marco Andrey Cipriani Frade
- Dermatology Division, Department of Medical Clinics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | | | | | - Marina Bessel
- Hospital Moinhos de Vento, Porto Alegre, Rio Grande do Sul, Brazil
| | - Celina Monteiro Abreu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Eliana M Wendland
- Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Pediatrics, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| | - Gilson P Dorneles
- Laboratory of Cellular and Molecular Immunology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
47
|
Dorneles GP, Teixeira PC, da Silva IM, Schipper LL, Santana Filho PC, Rodrigues Junior LC, Bonorino C, Peres A, Fonseca SG, Monteiro MC, Boeck CR, Eller S, Oliveira TF, Wendland EM, Romão PRT. Alterations in CD39/CD73 axis of T cells associated with COVID-19 severity. J Cell Physiol 2022; 237:3394-3407. [PMID: 35754396 PMCID: PMC9349448 DOI: 10.1002/jcp.30805] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/11/2022] [Accepted: 05/20/2022] [Indexed: 12/14/2022]
Abstract
Purinergic signaling modulates immune function and is involved in the immunopathogenesis of several viral infections. This study aimed to investigate alterations in purinergic pathways in coronavirus disease 2019 (COVID‐19) patients. Mild and severe COVID‐19 patients had lower extracellular adenosine triphosphate and adenosine levels, and higher cytokines than healthy controls. Mild COVID‐19 patients presented lower frequencies of CD4+CD25+CD39+ (activated/memory regulatory T cell [mTreg]) and increased frequencies of high‐differentiated (CD27−CD28−) CD8+ T cells compared with healthy controls. Severe COVID‐19 patients also showed higher frequencies of CD4+CD39+, CD4+CD25−CD39+ (memory T effector cell), and high‐differentiated CD8+ T cells (CD27−CD28−), and diminished frequencies of CD4+CD73+, CD4+CD25+CD39+ mTreg cell, CD8+CD73+, and low‐differentiated CD8+ T cells (CD27+CD28+) in the blood in relation to mild COVID‐19 patients and controls. Moreover, severe COVID‐19 patients presented higher expression of PD‐1 on low‐differentiated CD8+ T cells. Both severe and mild COVID‐19 patients presented higher frequencies of CD4+Annexin‐V+ and CD8+Annexin‐V+ T cells, indicating increased T‐cell apoptosis. Plasma samples collected from severe COVID‐19 patients were able to decrease the expression of CD73 on CD4+ and CD8+ T cells of a healthy donor. Interestingly, the in vitro incubation of peripheral blood mononuclear cell from severe COVID‐19 patients with adenosine reduced the nuclear factor‐κB activation in T cells and monocytes. Together, these data add new knowledge to the COVID‐19 immunopathology through purinergic regulation.
Collapse
Affiliation(s)
- Gilson P Dorneles
- Laboratory of Cellular and Molecular Immunology, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Paula C Teixeira
- Laboratory of Cellular and Molecular Immunology, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Igor M da Silva
- Laboratory of Cellular and Molecular Immunology, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Lucas L Schipper
- Laboratory of Cellular and Molecular Immunology, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Paulo C Santana Filho
- Laboratory of Cellular and Molecular Immunology, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Luiz C Rodrigues Junior
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Cristina Bonorino
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Alessandra Peres
- Laboratory of Cellular and Molecular Immunology, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Simone G Fonseca
- Institute of Tropical Pathology and Public Health, Universidade Federal de Goiás, Goiânia, Brazil
| | - Marta C Monteiro
- Graduate Program in Pharmaceutical Science, Health Science Institute, Federal University of Pará/UFPA, Belém, Pará, Brazil
| | - Carina R Boeck
- Graduate Program in Nanosciences and Health Sciences and Life, Universidade Franciscana-UFN, Santa Maria, Brazil
| | - Sarah Eller
- Pharmacosciences Department, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Tiago F Oliveira
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Pharmacosciences Department, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Eliana M Wendland
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Graduate Program in Pediatrics, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Pedro R T Romão
- Laboratory of Cellular and Molecular Immunology, Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.,Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
48
|
Rentería I, García-Suárez PC, Moncada-Jiménez J, Machado-Parra JP, Antunes BM, Lira FS, Jiménez-Maldonado A. Unhealthy Dieting During the COVID-19 Pandemic: An Opinion Regarding the Harmful Effects on Brain Health. Front Nutr 2022; 9:876112. [PMID: 35571935 PMCID: PMC9097874 DOI: 10.3389/fnut.2022.876112] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Since 2020, the world has been suffering from a pandemic that has affected thousands of people regardless of socio-economic conditions, forcing the population to adopt different strategies to prevent and control the advance of the disease, one of which is social distancing. Even though social distancing is a safe strategy to reduce the spread of COVID-19, it is also the cause of a rising sedentary behavior. This behavior develops an excess of fat tissue that leads to metabolic and inflammatory disruption related to chronic diseases and mental health disorders, such as anxiety, depression, and sleep issues. Furthermore, the adoption of dietary patterns involving the consumption of ultra-processed foods, higher in fats and sugars, and the reduction of fresh and healthy foods may play a role in the progress of the disease. In this perspective, we will discuss how an unhealthy diet can affect brain function and, consequently, be a risk factor for mental health diseases.
Collapse
Affiliation(s)
- Iván Rentería
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
| | - Patricia Concepción García-Suárez
- Facultad de Deportes, Universidad Autónoma de Baja California, Ensenada, Mexico
- Department of Health, Sports and Exercise Sciences, University of Kansas, Lawrence, KS, United States
| | - José Moncada-Jiménez
- Human Movement Sciences Research Center (CIMOHU), University of Costa Rica, San José, Costa Rica
| | | | | | - Fabio Santos Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, Paulista State University, UNESP, Presidente Prudente, São Paulo, Brazil
| | | |
Collapse
|
49
|
Cannabinoids Alleviate the LPS-Induced Cytokine Storm via Attenuating NLRP3 Inflammasome Signaling and TYK2-Mediated STAT3 Signaling Pathways In Vitro. Cells 2022; 11:cells11091391. [PMID: 35563697 PMCID: PMC9103143 DOI: 10.3390/cells11091391] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 12/15/2022] Open
Abstract
Cannabinoids, mainly cannabidiol (CBD) and Δ9-tetrahydrocannabinol (THC), are the most studied group of compounds obtained from Cannabis sativa because of their several pharmaceutical properties. Current evidence suggests a crucial role of cannabinoids as potent anti-inflammatory agents for the treatment of chronic inflammatory diseases; however, the mechanisms remain largely unclear. Cytokine storm, a dysregulated severe inflammatory response by our immune system, is involved in the pathogenesis of numerous chronic inflammatory disorders, including coronavirus disease 2019 (COVID-19), which results in the accumulation of pro-inflammatory cytokines. Therefore, we hypothesized that CBD and THC reduce the levels of pro-inflammatory cytokines by inhibiting key inflammatory signaling pathways. The nucleotide-binding and oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling has been implicated in a variety of chronic inflammatory diseases, which results in the release of pyroptotic cytokines, interleukin-1β (IL-1β) and IL-18. Likewise, the activation of the signal transducer and activator of transcription-3 (STAT3) causes increased expression of pro-inflammatory cytokines. We studied the effects of CBD and THC on lipopolysaccharide (LPS)-induced inflammatory response in human THP-1 macrophages and primary human bronchial epithelial cells (HBECs). Our results revealed that CBD and, for the first time, THC significantly inhibited NLRP3 inflammasome activation following LPS + ATP stimulation, leading to a reduction in the levels of IL-1β in THP-1 macrophages and HBECs. CBD attenuated the phosphorylation of nuclear factor-κB (NF-κB), and both cannabinoids inhibited the generation of oxidative stress post-LPS. Our multiplex ELISA data revealed that CBD and THC significantly diminished the levels of IL-6, IL-8, and tumor necrosis factor-α (TNF-α) after LPS treatment in THP-1 macrophages and HBECs. In addition, the phosphorylation of STAT3 was significantly downregulated by CBD and THC in THP-1 macrophages and HBECs, which was in turn attributed to the reduced phosphorylation of tyrosine kinase-2 (TYK2) by CBD and THC after LPS stimulation in these cells. Overall, CBD and THC were found to be effective in alleviating the LPS-induced cytokine storm in human macrophages and primary HBECs, at least via modulation of NLRP3 inflammasome and STAT3 signaling pathways. The encouraging results from this study warrant further investigation of these cannabinoids in vivo.
Collapse
|
50
|
Hamdy A, Leonardi A. Superantigens and SARS-CoV-2. Pathogens 2022; 11:390. [PMID: 35456065 PMCID: PMC9026686 DOI: 10.3390/pathogens11040390] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 12/31/2022] Open
Abstract
It has been posited SARS-CoV-2 contains at least one unique superantigen-like motif not found in any other SARS or endemic coronaviruses. Superantigens are potent antigens that can send the immune system into overdrive. SARS-CoV-2 causes many of the biological and clinical consequences of a superantigen, and, in the context of reinfection and waning immunity, it is important to better understand the impact of a widely circulating, airborne pathogen that may be a superantigen, superantigen-like or trigger a superantigenic host response. Urgent research is needed to better understand the long-term risks being taken by governments whose policies enable widespread transmission of a potential superantigenic pathogen, and to more clearly define the vaccination and public health policies needed to protect against the consequences of repeat exposure to the pathogen.
Collapse
Affiliation(s)
- Adam Hamdy
- Panres Pandemic Research, Newport TF10 8PG, UK
| | - Anthony Leonardi
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA;
| |
Collapse
|