1
|
Klibus M, Smirnova D, Marcinkevics Z, Rubins U, Grabovskis A, Vanags I, Sabelnikovs O. Quantitative Evaluation of Microcirculatory Alterations in Patients with COVID-19 and Bacterial Septic Shock through Remote Photoplethysmography and Automated Capillary Refill Time Analysis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1680. [PMID: 39459467 PMCID: PMC11509756 DOI: 10.3390/medicina60101680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Sepsis, a leading global health challenge, accounts for around 20% of deaths worldwide. The complexity of sepsis, especially the difference between bacterial and viral etiologies, requires an effective assessment of microcirculation during resuscitation. This study aimed to evaluate the impact of infusion therapy on microcirculation in patients with sepsis, focusing on bacterial- and COVID-19-associated sepsis using remote photoplethysmography (rPPG) and the automated capillary refill time (aCRT). Materials and Methods: This single-center prospective study was conducted in the ICU of Pauls Stradins Clinical University Hospital, including 20 patients with sepsis/septic shock. The patients were selected based on hemodynamic instability and divided into COVID-19 and Bacterial Septic Shock groups. Fluid responsiveness was assessed using the Passive Leg Raising Test (PLRT). Systemic hemodynamics and microcirculation were monitored through MAP CRT, rPPG, and serum lactate levels. Statistical analyses compared responses within and between the groups across different stages of the protocol. Results: The Bacterial group exhibited higher initial serum lactate levels and more pronounced microcirculatory dysfunction than the COVID-19 group. rPPG was more sensitive in detecting perfusion changes, showing significant differences between the groups. The automated CRT demonstrated greater sensitivity compared to the manual CRT, revealing significant differences during PLRT stages between bacterial- and COVID-19-associated sepsis. Both groups had a transient hemodynamic response to PLRT, with subsequent stabilization upon fluid infusion. Conclusions: When managing patients with sepsis in intensive care, monitoring microcirculation is of paramount importance in infusion therapy. Our study highlights the potential of rPPG and aCRT as tools for this purpose. These techniques can be used in conjunction with routine parameters, such as lactate levels and systemic hemodynamic parameters, to provide a comprehensive assessment of a patient's condition.
Collapse
Affiliation(s)
- Mara Klibus
- Department of Clinical Skills and Medical Technology, Rīga Stradiņš University, LV-1007 Riga, Latvia; (M.K.); (D.S.)
- Department of Anaesthesiology and Reanimatology, Rīga Stradiņš University, LV-1007 Riga, Latvia;
- Department of Anaesthesiology and Reanimatology, Pauls Stradins CUH, LV-1002 Riga, Latvia
| | - Darja Smirnova
- Department of Clinical Skills and Medical Technology, Rīga Stradiņš University, LV-1007 Riga, Latvia; (M.K.); (D.S.)
- Department of Anaesthesiology and Reanimatology, Pauls Stradins CUH, LV-1002 Riga, Latvia
| | | | - Uldis Rubins
- Faculty of Science and Technology, University of Latvia, LV-1063 Riga, Latvia; (U.R.); (A.G.)
| | - Andris Grabovskis
- Faculty of Science and Technology, University of Latvia, LV-1063 Riga, Latvia; (U.R.); (A.G.)
| | - Indulis Vanags
- Department of Anaesthesiology and Reanimatology, Rīga Stradiņš University, LV-1007 Riga, Latvia;
| | - Olegs Sabelnikovs
- Department of Clinical Skills and Medical Technology, Rīga Stradiņš University, LV-1007 Riga, Latvia; (M.K.); (D.S.)
- Department of Anaesthesiology and Reanimatology, Rīga Stradiņš University, LV-1007 Riga, Latvia;
- Department of Anaesthesiology and Reanimatology, Pauls Stradins CUH, LV-1002 Riga, Latvia
| |
Collapse
|
2
|
Ávila-Gómez P, Shingai Y, Dash S, Liu C, Callegari K, Meyer H, Khodarkovskaya A, Aburakawa D, Uchida H, Faraco G, Garcia-Bonilla L, Anrather J, Lee FS, Iadecola C, Sanchez T. Molecular and Functional Alterations in the Cerebral Microvasculature in an Optimized Mouse Model of Sepsis-Associated Cognitive Dysfunction. eNeuro 2024; 11:ENEURO.0426-23.2024. [PMID: 39266325 PMCID: PMC11439565 DOI: 10.1523/eneuro.0426-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/20/2024] [Accepted: 07/02/2024] [Indexed: 09/14/2024] Open
Abstract
Systemic inflammation has been implicated in the development and progression of neurodegenerative conditions such as cognitive impairment and dementia. Recent clinical studies indicate an association between sepsis, endothelial dysfunction, and cognitive decline. However, the investigations of the role and therapeutic potential of the cerebral microvasculature in sepsis-induced cognitive dysfunction have been limited by the lack of standardized experimental models for evaluating the alterations in the cerebral microvasculature and cognition induced by the systemic inflammatory response. Herein, we validated a mouse model of endotoxemia that recapitulates key pathophysiology related to sepsis-induced cognitive dysfunction, including the induction of an acute systemic hyperinflammatory response, blood-brain barrier (BBB) leakage, neurovascular inflammation, and memory impairment after recovery from the systemic inflammation. In the acute phase, we identified novel molecular (e.g., upregulation of plasmalemma vesicle-associated protein, PLVAP, a driver of endothelial permeability, and the procoagulant plasminogen activator inhibitor-1, PAI-1) and functional perturbations (i.e., albumin and small-molecule BBB leakage) in the cerebral microvasculature along with neuroinflammation. Remarkably, small-molecule BBB permeability, elevated levels of PAI-1, intra-/perivascular fibrin/fibrinogen deposition, and microglial activation persisted 1 month after recovery from sepsis. We also highlight molecular neuronal alterations of potential clinical relevance following systemic inflammation including changes in neurofilament phosphorylation and decreases in postsynaptic density protein 95 and brain-derived neurotrophic factor, suggesting diffuse axonal injury, synapse degeneration, and impaired neurotrophism. Our study serves as a standardized mouse model to support future mechanistic studies of sepsis-associated cognitive dysfunction and to identify novel endothelial therapeutic targets for this devastating condition.
Collapse
Affiliation(s)
- Paulo Ávila-Gómez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
| | - Yuto Shingai
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
| | - Sabyasachi Dash
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
| | - Catherine Liu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
| | - Keri Callegari
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
| | - Heidi Meyer
- Department of Psychiatry, Weill Cornell Medicine, New York, New York 10065
| | - Anne Khodarkovskaya
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
| | - Daiki Aburakawa
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
| | - Hiroki Uchida
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
| | - Giuseppe Faraco
- Department of Neuroscience, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Lidia Garcia-Bonilla
- Department of Neuroscience, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Josef Anrather
- Department of Neuroscience, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, New York 10065
| | - Costantino Iadecola
- Department of Neuroscience, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Teresa Sanchez
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York 10065
- Department of Neuroscience, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| |
Collapse
|
3
|
Meshref M, Ghaith HS, Hammad MA, Shalaby MMM, Ayasra F, Monib FA, Attia MS, Ebada MA, Elsayed H, Shalash A, Bahbah EI. The Role of RIN3 Gene in Alzheimer's Disease Pathogenesis: a Comprehensive Review. Mol Neurobiol 2024; 61:3528-3544. [PMID: 37995081 PMCID: PMC11087354 DOI: 10.1007/s12035-023-03802-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023]
Abstract
Alzheimer's disease (AD) is a globally prevalent form of dementia that impacts diverse populations and is characterized by progressive neurodegeneration and impairments in executive memory. Although the exact mechanisms underlying AD pathogenesis remain unclear, it is commonly accepted that the aggregation of misfolded proteins, such as amyloid plaques and neurofibrillary tau tangles, plays a critical role. Additionally, AD is a multifactorial condition influenced by various genetic factors and can manifest as either early-onset AD (EOAD) or late-onset AD (LOAD), each associated with specific gene variants. One gene of particular interest in both EOAD and LOAD is RIN3, a guanine nucleotide exchange factor. This gene plays a multifaceted role in AD pathogenesis. Firstly, upregulation of RIN3 can result in endosomal enlargement and dysfunction, thereby facilitating the accumulation of beta-amyloid (Aβ) peptides in the brain. Secondly, RIN3 has been shown to impact the PICLAM pathway, affecting transcytosis across the blood-brain barrier. Lastly, RIN3 has implications for immune-mediated responses, notably through its influence on the PTK2B gene. This review aims to provide a concise overview of AD and delve into the role of the RIN3 gene in its pathogenesis.
Collapse
Affiliation(s)
- Mostafa Meshref
- Department of Neurology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | | | | | | | - Faris Ayasra
- Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | | | - Mohamed S Attia
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | | | - Hanaa Elsayed
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ali Shalash
- Department of Neurology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eshak I Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt.
| |
Collapse
|
4
|
Ávila-Gómez P, Shingai Y, Dash S, Liu C, Callegari K, Meyer H, Khodarkovskaya A, Aburakawa D, Uchida H, Faraco G, Garcia-Bonilla L, Anrather J, Lee FS, Iadecola C, Sanchez T. Molecular and functional alterations in the cerebral microvasculature in an optimized mouse model of sepsis-associated cognitive dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596050. [PMID: 38853992 PMCID: PMC11160628 DOI: 10.1101/2024.05.28.596050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Systemic inflammation has been implicated in the development and progression of neurodegenerative conditions such as cognitive impairment and dementia. Recent clinical studies indicate an association between sepsis, endothelial dysfunction, and cognitive decline. However, the investigations of the role and therapeutic potential of the cerebral microvasculature in systemic inflammation-induced cognitive dysfunction have been limited by the lack of standardized experimental models for evaluating the alterations in the cerebral microvasculature and cognition induced by the systemic inflammatory response. Herein, we validated a mouse model of endotoxemia that recapitulates key pathophysiology related to sepsis-induced cognitive dysfunction, including the induction of an acute systemic hyperinflammatory response, blood-brain barrier (BBB) leakage, neurovascular inflammation, and memory impairment after recovery from the systemic inflammatory response. In the acute phase, we identified novel molecular (e.g. upregulation of plasmalemma vesicle associated protein, a driver of endothelial permeability, and the pro-coagulant plasminogen activator inhibitor-1, PAI-1) and functional perturbations (i.e., albumin and small molecule BBB leakage) in the cerebral microvasculature along with neuroinflammation. Remarkably, small molecule BBB permeability, elevated levels of PAI-1, intra/perivascular fibrin/fibrinogen deposition and microglial activation persisted 1 month after recovery from sepsis. We also highlight molecular neuronal alterations of potential clinical relevance following systemic inflammation including changes in neurofilament phosphorylation and decreases in postsynaptic density protein 95 and brain-derived neurotrophic factor suggesting diffuse axonal injury, synapse degeneration and impaired neurotrophism. Our study serves as a standardized model to support future mechanistic studies of sepsis-associated cognitive dysfunction and to identify novel endothelial therapeutic targets for this devastating condition. SIGNIFICANCE The limited knowledge of how systemic inflammation contributes to cognitive decline is a major obstacle to the development of novel therapies for dementia and other neurodegenerative diseases. Clinical evidence supports a role for the cerebral microvasculature in sepsis-induced neurocognitive dysfunction, but the investigation of the underlying mechanisms has been limited by the lack of standardized experimental models. Herein, we optimized a mouse model that recapitulates important pathophysiological aspects of systemic inflammation-induced cognitive decline and identified key alterations in the cerebral microvasculature associated with cognitive dysfunction. Our study provides a reliable experimental model for mechanistic studies and therapeutic discovery of the impact of systemic inflammation on cerebral microvascular function and the development and progression of cognitive impairment.
Collapse
|
5
|
Galiatsatos P, Garibaldi B, Yao D, Xu Y, Perin J, Shahu A, Jackson JW, Piggott D, Falade-Nwulia O, Shubella J, Michtalik H, Belcher HME, Hansel NN, Golden S. Lack of racial and ethnic disparities in mortality in minority patients hospitalised with COVID-19 in a mid-Atlantic healthcare system. BMJ Open Respir Res 2024; 11:e002310. [PMID: 38692710 PMCID: PMC11086483 DOI: 10.1136/bmjresp-2024-002310] [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: 01/12/2024] [Accepted: 04/11/2024] [Indexed: 05/03/2024] Open
Abstract
INTRODUCTION In the USA, minoritised communities (racial and ethnic) have suffered disproportionately from COVID-19 compared with non-Hispanic white communities. In a large cohort of patients hospitalised for COVID-19 in a healthcare system spanning five adult hospitals, we analysed outcomes of patients based on race and ethnicity. METHODS This was a retrospective cohort analysis of patients 18 years or older admitted to five hospitals in the mid-Atlantic area between 4 March 2020 and 27 May 2022 with confirmed COVID-19. Participants were divided into four groups based on their race/ethnicity: non-Hispanic black, non-Hispanic white, Latinx and other. Propensity score weighted generalised linear models were used to assess the association between race/ethnicity and the primary outcome of in-hospital mortality. RESULTS Of the 9651 participants in the cohort, more than half were aged 18-64 years old (56%) and 51% of the cohort were females. Non-Hispanic white patients had higher mortality (p<0.001) and longer hospital length-of-stay (p<0.001) than Latinx and non-Hispanic black patients. DISCUSSION In this large multihospital cohort of patients admitted with COVID-19, non-Hispanic black and Hispanic patients did not have worse outcomes than white patients. Such findings likely reflect how the complex range of factors that resulted in a life-threatening and disproportionate impact of incidence on certain vulnerable populations by COVID-19 in the community was offset through admission at well-resourced hospitals and healthcare systems. However, there continues to remain a need for efforts to address the significant pre-existing race and ethnicity inequities highlighted by the COVID-19 pandemic to be better prepared for future public health emergencies.
Collapse
Affiliation(s)
| | | | - Dapeng Yao
- Johns Hopkins University, Baltimore, Maryland, USA
| | - Yanxun Xu
- Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Jamie Perin
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andi Shahu
- Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - John W Jackson
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Franchini S, Mette F, Landoni G, Setti E, Ferrante L, Calcaterra B, Pagliula G, Barbieri A, Fontani D, Borio G, Citro M, Farolfi F, Suma G, Monti G, Colombo S, Dagna L, Rovere-Querini P, DE Cobelli F, Castagna A, Ciceri F, Zangrillo A, Tresoldi M, Secchi A, Etteri M. Gas-exchange deficit and systemic hypoperfusion in COVID-19 and non-COVID-19 young adult patients with pneumonia. Panminerva Med 2024; 66:27-35. [PMID: 35119247 DOI: 10.23736/s0031-0808.22.04562-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Lung damage leading to gas-exchange deficit and sepsis leading to systemic hypoperfusion are well-known features of severe pneumonia. Although frequently described in COVID-19, their prognostic impact in COVID-19-related pneumonia versus COVID-19-unrelated pneumonia has never been compared. This study assesses fundamental gas-exchange and hemodynamic parameters and explores their prognostic impact in COVID-19 pneumonia and non-COVID-19 pneumonia. METHODS We prospectively evaluated arterial pO2/FiO2, alveolar to arterial O2 gradient, shock index, and serum lactate in 126 COVID-19 pneumonia patients, aged 18-65, presenting to the emergency department with acute, non-hypercapnic respiratory failure. As a control group we identified 1:1 age-, sex-, and pO2/FiO2-matched COVID-19-urelated pneumonia patients. Univariate and multivariable predictors of 30-day survival were identified in both groups. RESULTS COVID-19 patients showed lower arterial serum lactate concentration (P<0.001) and shock index (P<0.001) values as compared to non-COVID-19 patients. While we did not observe differences in lactate concentration or in shock index values in deceased vs. surviving COVID-19 patients (P=0.7 and P=0.6, respectively), non-COVID-19 deceased patients showed significantly higher lactate and shock index than non-COVID-19 survivors (P<0.001 and P=0.03). The pO2/FiO2 was the most powerful determinant of survival by Cox regression multivariate analysis in COVID-19 patients (P=0.006), while it was lactate in non-COVID-19 patients (P=0.001). CONCLUSIONS As compared to COVID19-unrelated pneumonia, COVID-19 pneumonia outcome seems more strictly correlated to the extent of lung damage, rather than to the systemic circulatory and metabolic derangements typical of sepsis.
Collapse
Affiliation(s)
- Stefano Franchini
- Emergency Department, IRCCS San Raffaele Scientific Institute, Milan, Italy -
| | - Francesca Mette
- Emergency Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Eleonora Setti
- Emergency Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Ferrante
- Emergency Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Barbara Calcaterra
- Emergency Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gaia Pagliula
- Emergency Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Dario Fontani
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Giorgia Borio
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Marta Citro
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Farolfi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Gloria Suma
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Giacomo Monti
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sergio Colombo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Patrizia Rovere-Querini
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Internal Medicine, Diabetes, and Endocrinology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco DE Cobelli
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Castagna
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Department of Infectious Diseases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Fabio Ciceri
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Alberto Zangrillo
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Moreno Tresoldi
- General Medicine and Advanced Care Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Antonio Secchi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | |
Collapse
|
7
|
Lai J, Liang J, Chen K, Guan B, Chen Z, Chen L, Fan J, Zhang Y, Li Q, Su J, Chen Q, Lin J. Carrimycin ameliorates lipopolysaccharide and cecal ligation and puncture-induced sepsis in mice. Chin J Nat Med 2024; 22:235-248. [PMID: 38553191 DOI: 10.1016/s1875-5364(24)60600-x] [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: 07/14/2023] [Indexed: 04/02/2024]
Abstract
Carrimycin (CA), sanctioned by China's National Medical Products Administration (NMPA) in 2019 for treating acute bronchitis and sinusitis, has recently been observed to exhibit multifaceted biological activities, encompassing anti-inflammatory, antiviral, and anti-tumor properties. Despite these applications, its efficacy in sepsis treatment remains unexplored. This study introduces a novel function of CA, demonstrating its capacity to mitigate sepsis induced by lipopolysaccharide (LPS) and cecal ligation and puncture (CLP) in mice models. Our research employed in vitro assays, real-time quantitative polymerase chain reaction (RT-qPCR), and RNA-seq analysis to establish that CA significantly reduces the levels of pro-inflammatory cytokines, namely tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6), in response to LPS stimulation. Additionally, Western blotting and immunofluorescence assays revealed that CA impedes Nuclear Factor Kappa B (NF-κB) activation in LPS-stimulated RAW264.7 cells. Complementing these findings, in vivo experiments demonstrated that CA effectively alleviates LPS- and CLP-triggered organ inflammation in C57BL/6 mice. Further insights were gained through 16S sequencing, highlighting CA's pivotal role in enhancing gut microbiota diversity and modulating metabolic pathways, particularly by augmenting the production of short-chain fatty acids in mice subjected to CLP. Notably, a comparative analysis revealed that CA's anti-inflammatory efficacy surpasses that of equivalent doses of aspirin (ASP) and TIENAM. Collectively, these findings suggest that CA exhibits significant therapeutic potential in sepsis treatment. This discovery provides a foundational theoretical basis for the clinical application of CA in sepsis management.
Collapse
Affiliation(s)
- Junzhong Lai
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Jiadi Liang
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China; Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Kunsen Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Biyun Guan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Zhirong Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Linqin Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Jiqiang Fan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Yong Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Qiumei Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Jingqian Su
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou 350117, China.
| | - Jizhen Lin
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou 350001, China; The Department of Otolaryngology, Head & Neck Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
8
|
Elnosary ME, Shreadah MA, Ashour ML, Nabil-Adam A. Predictions based on inflammatory cytokine profiling of Egyptian COVID-19 with 2 potential therapeutic effects of certain marine-derived compounds. Int Immunopharmacol 2024; 126:111072. [PMID: 38006751 DOI: 10.1016/j.intimp.2023.111072] [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: 11/12/2022] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUNDS A worldwide coronavirus pandemic has affected many healthcare systems in 2019 (COVID-19). Following viral activation, cytokines and chemokines are released, causing inflammation and tissue death, particularly in the lungs, resulting in severe COVID-19 symptoms such as pneumonia and ARDS. COVID-19 induces the release of several chemokines and cytokines in different organs, such as the cardiovascular system and lungs. RESEARCH IDEA COVID-19 and its more severe effects, such as an elevated risk of death, are more common in patients with metabolic syndrome and the elderly. Cytokine storm and COVID-19 severity may be mitigated by immunomodulation targeting NF-κB activation in conjunction with TNF- α -inhibition. In severe cases of COVID-19, inhibiting the NF-κB/TNF- α, the pathway may be employed as a therapeutic option. MATERIAL AND METHODS The study will elaborate on the Egyptian pattern for COVID-19 patients in the first part of our study. An Egyptian patient with COVID-19 inflammatory profiling will be discussed in the second part of this article using approved marine drugs selected to inhabit the significant inflammatory signals. A biomarker profiling study is currently being performed on Egyptian patients with SARS-COV-2. According to the severity of the infection, participants were divided into four groups. The First Group was non-infected with SARS-CoV-2 (Control, n = 16), the Second Group was non-intensive care patients (non-ICU, n = 16), the Third Group was intensive care patients (ICU, n = 16), and the Fourth Group was ICU with endotracheal intubation (ICU + EI, n = 16). To investigate COVID-19 inflammatory biomarkers for Egyptian patients, several inflammatory, oxidative, antioxidant, and anti-inflammatory biomarkers were measured. The following are examples of blood tests: CRP, Ferritin, D-dimer, TNF-α, IL-8, IL-6., IL-Ib, CD8, NF-κB, MDA, and total antioxidants. RESULTS AND DISCUSSION The results of the current study revealed many logical findings, such as the elevation of CRP, Ferritin, D-dimer, TNF- α, CD8, IL-6, IL-, NF-κB, and MDA. Where a significant increase showed in ICU group results (23.05 ± 0.30, 2.35 ± 0.86, 433.4 ± 159.3, 26.67 ± 3.51, 7.52 ± 1.48, 7.49 ± 1.04, 5.76 ± 1.31, 7.41 ± 0.73) respectively, and also ICU group results (54.75 ± 3.44, 0.65 ± 0.13, 460.2 ± 121.42, 27.43 ± 2.52, 8.63 ± 2.68, 10.65 ± 2.75, 5.93 ± 1.4, 10.64 ± 0.86) respectively, as well as ICU + EI group results (117.63 ± 11.89, 1.22 ± 0.65, 918.8 ± 159.27, 26.68 ± 2.00, 6.68 ± 1.08, 11.68 ± 6.16, 6.23 ± 0.07, 22.41 ± 1.39),respectively.The elevation in laboratory biomarkers of cytokines storm in three infected groups with remarkable increases in the ICU + EI group was due to the elevation of oxidative stress and inflammatory storm molecules, which lead to highly inflammatory responses, specifically in severe patients of COVID-19. Another approach to be used in the current study is investigating new computational drug compounds for SARS-COV-2 protective agents from the marine environment. The results revealed that (Imatinib and Indinavir) had the highest affinity toward Inflammatory molecules and COVID-19 proteins (PDB ID: -7CZ4 and 7KJR), which may be used in the future as possible COVID-19 drug candidates. CONCLUSION The investigated inflammatory biomarkers in Egyptian COVID-19 patients showed a strong correlation between IL6, TNF-α, NF-κB, CRB, DHL, and ferritin as COVID-19 biomarkers and determined the severity of the infection. Also, the oxidative /antioxidant showed good biomarkers for infection recovery and progression of the patients.
Collapse
Affiliation(s)
- Mohamed E Elnosary
- Al-Azhar University, Faculty of Science, Botany and Microbiology Department, 11884 Nasr City, Cairo, Egypt.
| | - Mohamed Attia Shreadah
- Marine Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Egypt
| | - Mohamed L Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Abbasia, Cairo 11566, Egypt; Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia.
| | - Asmaa Nabil-Adam
- Marine Biotechnology and Natural Products Laboratory, National Institute of Oceanography & Fisheries, Egypt.
| |
Collapse
|
9
|
Skei NV, Nilsen TIL, Mohus RM, Prescott HC, Lydersen S, Solligård E, Damås JK, Gustad LT. Trends in mortality after a sepsis hospitalization: a nationwide prospective registry study from 2008 to 2021. Infection 2023; 51:1773-1786. [PMID: 37572240 PMCID: PMC10665235 DOI: 10.1007/s15010-023-02082-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Few studies have reported on mortality beyond one year after sepsis. We aim to describe trends in short- and long-term mortality among patients admitted with sepsis, and to describe the association between clinical characteristics and mortality for improved monitoring, treatment and prognosis. METHODS Patients ≥ 18 years admitted to all Norwegian hospitals (2008-2021) with a first sepsis episode were identified using Norwegian Patient Registry and International Classification of Diseases 10th Revision codes. Sepsis was classified as implicit (known infection site plus organ dysfunction), explicit (unknown infection site), or COVID-19-related sepsis. The outcome was all-cause mortality. We describe age-standardized 30-day, 90-day, 1-, 5- and 10-year mortality for each admission year and estimated the annual percentage change with 95% confidence interval (CI). The association between clinical characteristics and all-cause mortality is reported as hazard ratios (HRs) adjusted for age, sex and calendar year in Cox regression. RESULTS The study included 222,832 patients, of whom 127,059 (57.1%) had implicit, 92,928 (41.7%) had explicit, and 2,845 (1.3%) had COVID-19-related sepsis (data from 2020 and 2021). Trends in overall age-standardized 30-day, 90-day, 1- and 5-year mortality decreased by 0.29 (95% CI - 0.39 to - 0.19), 0.43 (95% CI - 0.56 to - 0.29), 0.61 (95% CI - 0.73 to - 0.49) and 0.66 (95% CI - 0.84 to - 0.48) percent per year, respectively. The decrease was observed for all infections sites but was largest among patients with respiratory tract infections. Implicit, explicit and COVID-19-related sepsis had largely similar overall mortality, with explicit sepsis having an adjusted HR of 0.980 (95% CI 0.969 to 0.991) and COVID-19-related sepsis an adjusted HR of 0.916 (95% CI 0.836 to 1.003) compared to implicit sepsis. Patients with respiratory tract infections have somewhat higher mortality than those with other infection sites. Number of comorbidities was positively associated with mortality, but mortality varied considerably between different comorbidities. Similarly, number of acute organ dysfunctions was strongly associated with mortality, whereas the risk varied for each type of organ dysfunction. CONCLUSION Overall mortality has declined over the past 14 years among patients with a first sepsis admission. Comorbidity, site of infection, and acute organ dysfunction are patient characteristics that are associated with mortality. This could inform health care workers and raise the awareness toward subgroups of patients that needs particular attention to improve long-term mortality.
Collapse
Affiliation(s)
- Nina Vibeche Skei
- Department of Anesthesia and Intensive Care, Nord-Trondelag Hospital Trust, Levanger, Norway.
- Department of Circulation and Medical Imaging, Mid Norway Sepsis Research Center, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Tom Ivar Lund Nilsen
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Randi Marie Mohus
- Department of Circulation and Medical Imaging, Mid Norway Sepsis Research Center, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Anesthesia and Intensive Care, St. Olav's University Hospital, Trondheim, Norway
| | - Hallie C Prescott
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- VA Center for Clinical Management Research, Ann Arbor, MI, USA
| | - Stian Lydersen
- Department of Mental Health, Faculty of Medicine and Health Sciences, Regional Centre for Child and Youth Mental Health and Child Welfare, Norwegian University of Science and Technology, (NTNU), Trondheim, Norway
| | - Erik Solligård
- Department of Circulation and Medical Imaging, Mid Norway Sepsis Research Center, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jan Kristian Damås
- Department of Circulation and Medical Imaging, Mid Norway Sepsis Research Center, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Centre of Molecular Inflammation Research, Institute for Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Infectious Diseases, St. Olav's University Hospital, Trondheim, Norway
| | - Lise Tuset Gustad
- Department of Circulation and Medical Imaging, Mid Norway Sepsis Research Center, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Faculty of Nursing and Health Sciences, Nord University, Levanger, Norway
- Department of Medicine and Rehabilitation, Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| |
Collapse
|
10
|
Qian G, Fang H, Chen A, Sun Z, Huang M, Luo M, Cheng E, Zhang S, Wang X, Fang H. A hub gene signature as a therapeutic target and biomarker for sepsis and geriatric sepsis-induced ARDS concomitant with COVID-19 infection. Front Immunol 2023; 14:1257834. [PMID: 37822934 PMCID: PMC10562607 DOI: 10.3389/fimmu.2023.1257834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/08/2023] [Indexed: 10/13/2023] Open
Abstract
Background COVID-19 and sepsis represent formidable public health challenges, characterized by incompletely elucidated molecular mechanisms. Elucidating the interplay between COVID-19 and sepsis, particularly in geriatric patients suffering from sepsis-induced acute respiratory distress syndrome (ARDS), is of paramount importance for identifying potential therapeutic interventions to mitigate hospitalization and mortality risks. Methods We employed bioinformatics and systems biology approaches to identify hub genes, shared pathways, molecular biomarkers, and candidate therapeutics for managing sepsis and sepsis-induced ARDS in the context of COVID-19 infection, as well as co-existing or sequentially occurring infections. We corroborated these hub genes utilizing murine sepsis-ARDS models and blood samples derived from geriatric patients afflicted by sepsis-induced ARDS. Results Our investigation revealed 189 differentially expressed genes (DEGs) shared among COVID-19 and sepsis datasets. We constructed a protein-protein interaction network, unearthing pivotal hub genes and modules. Notably, nine hub genes displayed significant alterations and correlations with critical inflammatory mediators of pulmonary injury in murine septic lungs. Simultaneously, 12 displayed significant changes and correlations with a neutrophil-recruiting chemokine in geriatric patients with sepsis-induced ARDS. Of these, six hub genes (CD247, CD2, CD40LG, KLRB1, LCN2, RETN) showed significant alterations across COVID-19, sepsis, and geriatric sepsis-induced ARDS. Our single-cell RNA sequencing analysis of hub genes across diverse immune cell types furnished insights into disease pathogenesis. Functional analysis underscored the interconnection between sepsis/sepsis-ARDS and COVID-19, enabling us to pinpoint potential therapeutic targets, transcription factor-gene interactions, DEG-microRNA co-regulatory networks, and prospective drug and chemical compound interactions involving hub genes. Conclusion Our investigation offers potential therapeutic targets/biomarkers, sheds light on the immune response in geriatric patients with sepsis-induced ARDS, emphasizes the association between sepsis/sepsis-ARDS and COVID-19, and proposes prospective alternative pathways for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Guojun Qian
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Hongwei Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Anning Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhun Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Meiying Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Mengyuan Luo
- Department of Anesthesiology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Erdeng Cheng
- Department of Anesthesiology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shengyi Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaokai Wang
- Department of Interventional and Vascular Surgery, Xuzhou First People's Hospital, Xuzhou, China
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Anesthesiology, Minhang Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Fudan Zhangjiang Institute, Shanghai, China
- Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai, China
| |
Collapse
|
11
|
Skei NV, Nilsen TIL, Knoop ST, Prescott H, Lydersen S, Mohus RM, Brkic A, Liyanarachi KV, Solligård E, Damås JK, Gustad LT. Long-term temporal trends in incidence rate and case fatality of sepsis and COVID-19-related sepsis in Norwegian hospitals, 2008-2021: a nationwide registry study. BMJ Open 2023; 13:e071846. [PMID: 37532480 PMCID: PMC10401253 DOI: 10.1136/bmjopen-2023-071846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 07/18/2023] [Indexed: 08/04/2023] Open
Abstract
OBJECTIVES To estimate temporal trends in incidence rate (IR) and case fatality during a 14-year period from 2008 to 2021, and to assess possible shifts in these trends during the COVID-19 pandemic. SETTING All Norwegian hospitals 2008-2021. PARTICIPANTS 317 705 patients ≥18 year with a sepsis International Classification of Diseases 10th revision code retrieved from The Norwegian Patient Registry. PRIMARY AND SECONDARY MEASURES Annual age-standardised IRs with 95% CIs. Poisson regression was used to estimate changes in IRs across time, and logistic regression was used to estimate ORs for in-hospital death. RESULTS Among 12 619 803 adult hospitalisations, a total of 317 705 (2.5%) hospitalisations in 222 832 (70.0%) unique patients met the sepsis criteria. The overall age-standardised IR of a first sepsis admission was 246/100 000 (95% CI 245 to 247), whereas the age-standardised IR of all sepsis admissions was 352/100 000 (95% CI 351 to 354). In the period 2009-2019, the annual IR for a first sepsis episode was stable (IR ratio (IRR) per year, 0.999; 95% CI 0.994 to 1.004), whereas for recurrent sepsis the IR increased (annual IRR, 1.048; 95% CI 1.037 to 1.059). During the COVID-19 pandemic, the IRR for a first sepsis was 0.877 (95% CI 0.829 to 0.927) in 2020 and 0.929 (95% CI 0.870 to 0.992) in 2021, and for all sepsis it was 0.870 (95% CI 0.810 to 0.935) in 2020 and 0.908 (95% CI 0.840 to 0.980) in 2021, compared with the previous 11-year period. Case fatality among first sepsis admissions declined in the period 2009-2019 (annual OR 0.954 (95% CI 0.950 to 0.958)), whereas case fatality increased during the COVID-19 pandemic in 2020 (OR 1.061 (95% CI 1.001 to 1.124) and in 2021 (OR 1.164 (95% CI 1.098 to 1.233)). CONCLUSION The overall IR of sepsis increased from 2009 to 2019, due to an increasing IR of recurrent sepsis, and indicates that sepsis awareness with updated guidelines and education must continue.
Collapse
Affiliation(s)
- Nina Vibeche Skei
- Department of Anesthesia and Intensive Care, Nord-Trøndelag Hospital Trust, Levanger, Norway
- Institute of Circulation and Medical Imaging, Mid-Norway Centre of Sepsis Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tom Ivar Lund Nilsen
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Siri Tandberg Knoop
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hallie Prescott
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- VA Center for Clinical Management Research, HSR&D Center of Innovation, Ann Arbor, Michigan, USA
| | - Stian Lydersen
- Regional Centre for Child and Youth Mental Health and Child Welfare, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Randi Marie Mohus
- Institute of Circulation and Medical Imaging, Mid-Norway Centre of Sepsis Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Anaesthesia and Intensive Care, St Olavs Hospital Trondheim University Hospital, Trondheim, Norway
| | - Alen Brkic
- Research Department, Sørlandet Sykehus HF, Kristiansand, Norway
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristin Vardheim Liyanarachi
- Institute of Circulation and Medical Imaging, Mid-Norway Centre of Sepsis Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Infectious Diseases, St. Olav's University Hospital, Trondheim, Norway
| | - Erik Solligård
- Institute of Circulation and Medical Imaging, Mid-Norway Centre of Sepsis Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jan Kristian Damås
- Institute of Circulation and Medical Imaging, Mid-Norway Centre of Sepsis Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Infectious Diseases, St. Olav's University Hospital, Trondheim, Norway
- Institute of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Lise Tuset Gustad
- Institute of Circulation and Medical Imaging, Mid-Norway Centre of Sepsis Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Medicine and Rehabilitation, Nord-Trondelag Hospital Trust, Levanger, Norway
- Faculty of Nursing and Health Sciences, Nord University, Levanger, Norway
| |
Collapse
|
12
|
Zingaropoli MA, Latronico T, Pasculli P, Masci GM, Merz R, Ciccone F, Dominelli F, Del Borgo C, Lichtner M, Iafrate F, Galardo G, Pugliese F, Panebianco V, Ricci P, Catalano C, Ciardi MR, Liuzzi GM, Mastroianni CM. Tissue Inhibitor of Matrix Metalloproteinases-1 (TIMP-1) and Pulmonary Involvement in COVID-19 Pneumonia. Biomolecules 2023; 13:1040. [PMID: 37509076 PMCID: PMC10377146 DOI: 10.3390/biom13071040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Background: The aim of the study was to longitudinally evaluate the association between MMP-2, MMP-9, TIMP-1 and chest radiological findings in COVID-19 patients. Methods: COVID-19 patients were evaluated based on their hospital admission (baseline) and three months after hospital discharge (T post) and were stratified into ARDS and non-ARDS groups. As a control group, healthy donors (HD) were enrolled. Results: At the baseline, compared to HD (n = 53), COVID-19 patients (n = 129) showed higher plasma levels of MMP-9 (p < 0.0001) and TIMP-1 (p < 0.0001) and the higher plasma activity of MMP-2 (p < 0.0001) and MMP-9 (p < 0.0001). In the ARDS group, higher plasma levels of MMP-9 (p = 0.0339) and TIMP-1 (p = 0.0044) and the plasma activity of MMP-2 (p = 0.0258) and MMP-9 (p = 0.0021) compared to non-ARDS was observed. A positive correlation between the plasma levels of TIMP-1 and chest computed tomography (CT) score (ρ = 0.2302, p = 0.0160) was observed. At the T post, a reduction in plasma levels of TIMP-1 (p < 0.0001), whereas an increase in the plasma levels of MMP-9 was observed (p = 0.0088). Conclusions: The positive correlation between TIMP-1 with chest CT scores highlights its potential use as a marker of fibrotic burden. At T post, the increase in plasma levels of MMP-9 and the reduction in plasma levels of TIMP-1 suggested that inflammation and fibrosis resolution were still ongoing.
Collapse
Affiliation(s)
- Maria Antonella Zingaropoli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Tiziana Latronico
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Patrizia Pasculli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Giorgio Maria Masci
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Roberta Merz
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Federica Ciccone
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Federica Dominelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Cosmo Del Borgo
- Infectious Diseases Unit, Santa Maria Goretti Hospital, Sapienza, University of Rome, 04100 Latina, Italy
| | - Miriam Lichtner
- Infectious Diseases Unit, Santa Maria Goretti Hospital, Sapienza, University of Rome, 04100 Latina, Italy
- Department of Neurosciences Mental Health and Sensory Organs, Sapienza University of Rome, 00161 Rome, Italy
| | - Franco Iafrate
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Gioacchino Galardo
- Medical Emergency Unit, Sapienza University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Francesco Pugliese
- Department of Specialist Surgery and Organ Transplantation "Paride Stefanini", Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy
| | - Valeria Panebianco
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Paolo Ricci
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Unit of Emergency Radiology, Policlinico Umberto I, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Carlo Catalano
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Grazia Maria Liuzzi
- Department of Biosciences, Biotechnologies and Environment, University of Bari "Aldo Moro", 70121 Bari, Italy
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
13
|
Salomão R, Assis V, de Sousa Neto IV, Petriz B, Babault N, Durigan JLQ, de Cássia Marqueti R. Involvement of Matrix Metalloproteinases in COVID-19: Molecular Targets, Mechanisms, and Insights for Therapeutic Interventions. BIOLOGY 2023; 12:843. [PMID: 37372128 PMCID: PMC10295079 DOI: 10.3390/biology12060843] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023]
Abstract
MMPs are enzymes involved in SARS-CoV-2 pathogenesis. Notably, the proteolytic activation of MMPs can occur through angiotensin II, immune cells, cytokines, and pro-oxidant agents. However, comprehensive information regarding the impact of MMPs in the different physiological systems with disease progression is not fully understood. In the current study, we review the recent biological advances in understanding the function of MMPs and examine time-course changes in MMPs during COVID-19. In addition, we explore the interplay between pre-existing comorbidities, disease severity, and MMPs. The reviewed studies showed increases in different MMP classes in the cerebrospinal fluid, lung, myocardium, peripheral blood cells, serum, and plasma in patients with COVID-19 compared to non-infected individuals. Individuals with arthritis, obesity, diabetes, hypertension, autoimmune diseases, and cancer had higher MMP levels when infected. Furthermore, this up-regulation may be associated with disease severity and the hospitalization period. Clarifying the molecular pathways and specific mechanisms that mediate MMP activity is important in developing optimized interventions to improve health and clinical outcomes during COVID-19. Furthermore, better knowledge of MMPs will likely provide possible pharmacological and non-pharmacological interventions. This relevant topic might add new concepts and implications for public health in the near future.
Collapse
Affiliation(s)
- Rebecca Salomão
- Laboratory of Molecular Analysis, Postgraduate Program in Health and Sciences and Technology, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil
| | - Victoria Assis
- Laboratory of Molecular Analysis, Postgraduate Program in Rehabilitation Sciences, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil; (V.A.); (J.L.Q.D.)
| | - Ivo Vieira de Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-907, SP, Brazil;
| | - Bernardo Petriz
- Graduate Program in Genomic Sciences and Biotechnology, Catholic University of Brasilia, Brasilia 71966-700, DF, Brazil;
- Laboratory of Exercise Molecular Physiology, University Center UDF, Brasília 71966-900, DF, Brazil
| | - Nicolas Babault
- INSERM UMR1093-CAPS, UFR des Sciences du Sport, Université de Bourgogne, F-21000 Dijon, France;
- Centre d’Expertise de la Performance, UFR des Sciences du Sport, Université de Bourgogne, F-21000 Dijon, France
| | - João Luiz Quaglioti Durigan
- Laboratory of Molecular Analysis, Postgraduate Program in Rehabilitation Sciences, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil; (V.A.); (J.L.Q.D.)
| | - Rita de Cássia Marqueti
- Laboratory of Molecular Analysis, Postgraduate Program in Health and Sciences and Technology, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil
- Laboratory of Molecular Analysis, Postgraduate Program in Rehabilitation Sciences, Faculty of Ceilândia, University of Brasilia, Brasilia 72220-275, DF, Brazil; (V.A.); (J.L.Q.D.)
| |
Collapse
|
14
|
Polushin YS, Sokolov DV, Dreval RO, Zabotina AN. Clinical and Economic Evaluation of the Blood Purification with Selective Sorption Techniques in ICU Patients. MESSENGER OF ANESTHESIOLOGY AND RESUSCITATION 2023. [DOI: 10.24884/2078-5658-2023-20-1-6-16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
The objective was to analyze the clinical and economic feasibility of using selective methods of lipopolysaccharide (LPS) sorption for sepsis treatment.Materials and methods. A clinical and economic model was used to assess the feasibility of using some technologies of selective lipopolysaccharide sorption. The model was developed in accordance with the current industry standard «Clinical and Economic Research» used in Russia. The target population was patients of 18 years old and older with sepsis. The methodology involved the evaluation of available randomized clinical trials, as well as studies involving data from network meta-analyses and systematic reviews of the use of the studied technologies. The calculation of direct medical and indirect non-medical costs was made with an analysis of the impact on the budget during the first year and on the time horizon for five years.Results. The value of direct and indirect costs was determined when using sorption devices based on Efferon LPS, Toraymyxin, Alteco LPS Adsorber, Toxipak columns. It was shown that among the therapeutic alternatives, the use of the Efferon LPS column had the least burden on the budget.Conclusion. The use of selective lipopolysaccharide sorption technologies in the intensive care program for sepsis patients, although accompanied by an increase in direct and indirect financial costs, leads to budget savings in the medium term. Improving the mechanism for compensating the costs of using this technology is a promising way to improve the outcomes of sepsis treatment.
Collapse
Affiliation(s)
| | | | - R. O. Dreval
- Nonprofit Partnership «Center for Social Economics»
| | | |
Collapse
|
15
|
Gravrand V, Mellot F, Ackermann F, Ballester MC, Zuber B, Kirk JT, Navalkar K, Yager TD, Petit F, Pascreau T, Farfour E, Vasse M. Stratification of COVID-19 Severity Using SeptiCyte RAPID, a Novel Host Immune Response Test. Viruses 2023; 15:419. [PMID: 36851633 PMCID: PMC9960895 DOI: 10.3390/v15020419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
SeptiCyte® RAPID is a gene expression assay measuring the relative expression levels of host response genes PLA2G7 and PLAC8, indicative of a dysregulated immune response during sepsis. As severe forms of COVID-19 may be considered viral sepsis, we evaluated SeptiCyte RAPID in a series of 94 patients admitted to Foch Hospital (Suresnes, France) with proven SARS-CoV-2 infection. EDTA blood was collected in the emergency department (ED) in 67 cases, in the intensive care unit (ICU) in 23 cases and in conventional units in 4 cases. SeptiScore (0-15 scale) increased with COVID-19 severity. Patients in ICU had the highest SeptiScores, producing values comparable to 8 patients with culture-confirmed bacterial sepsis. Receiver operating characteristic (ROC) curve analysis had an area under the curve (AUC) of 0.81 for discriminating patients requiring ICU admission from patients who were immediately discharged or from patients requiring hospitalization in conventional units. SeptiScores increased with the extent of the lung injury. For 68 patients, a chest computed tomography (CT) scan was performed within 24 h of COVID-19 diagnosis. SeptiScore >7 suggested lung injury ≥50% (AUC = 0.86). SeptiCyte RAPID was compared to other biomarkers for discriminating Critical + Severe COVID-19 in ICU, versus Moderate + Mild COVID-19 not in ICU. The mean AUC for SeptiCyte RAPID was superior to that of any individual biomarker or combination thereof. In contrast to C-reactive protein (CRP), correlation of SeptiScore with lung injury was not impacted by treatment with anti-inflammatory agents. SeptiCyte RAPID can be a useful tool to identify patients with severe forms of COVID-19 in ED, as well as during follow-up.
Collapse
Affiliation(s)
| | | | - Felix Ackermann
- Internal Medicine Department, Foch Hospital, 92150 Suresnes, France
| | | | - Benjamin Zuber
- Intensive Care Unit, Foch Hospital, 92150 Suresnes, France
| | | | | | | | - Fabien Petit
- Biology Department, Foch Hospital, 92150 Suresnes, France
| | - Tiffany Pascreau
- Biology Department, Foch Hospital, 92150 Suresnes, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S1176, 94270 Le Kremlin-Bicêtre, France
| | - Eric Farfour
- Biology Department, Foch Hospital, 92150 Suresnes, France
| | - Marc Vasse
- Biology Department, Foch Hospital, 92150 Suresnes, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-S1176, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
16
|
Cafferkey J, Ferguson A, Grahamslaw J, Oatey K, Norrie J, Lone N, Walsh T, Horner D, Appelboam A, Hall P, Skipworth R, Bell D, Rooney K, Shankar-Hari M, Corfield A, Gray A. Albumin versus balanced crystalloid for resuscitation in the treatment of sepsis: A protocol for a randomised controlled feasibility study, "ABC-Sepsis". J Intensive Care Soc 2023; 24:78-84. [PMID: 36860553 PMCID: PMC9157259 DOI: 10.1177/17511437221103692] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Patients presenting with suspected sepsis to secondary care often require fluid resuscitation to correct hypovolaemia and/or septic shock. Existing evidence signals, but does not demonstrate, a benefit for regimes including albumin over balanced crystalloid alone. However, interventions may be started too late, missing a critical resuscitation window. Methods ABC Sepsis is a currently recruiting randomised controlled feasibility trial comparing 5% human albumin solution (HAS) with balanced crystalloid for fluid resuscitation in patients with suspected sepsis. This multicentre trial is recruiting adult patients within 12 hours of presentation to secondary care with suspected community acquired sepsis, with a National Early Warning Score ≥5, who require intravenous fluid resuscitation. Participants are randomised to 5% HAS or balanced crystalloid as the sole resuscitation fluid for the first 6 hours. Objectives Primary objectives are feasibility of recruitment to the study and 30-day mortality between groups. Secondary objectives include in-hospital and 90-day mortality, adherence to trial protocol, quality of life measurement and secondary care costs. Discussion This trial aims to determine the feasibility of conducting a trial to address the current uncertainty around optimal fluid resuscitation of patients with suspected sepsis. Understanding the feasibility of delivering a definitive study will be dependent on how the study team are able to negotiate clinician choice, Emergency Department pressures and participant acceptability, as well as whether any clinical signal of benefit is detected.
Collapse
Affiliation(s)
- John Cafferkey
- Emergency Medicine Research Group
Edinburgh (EMERGE), Department of Emergency Medicine, Royal Infirmary of
Edinburgh, Edinburgh, UK
| | - Andrew Ferguson
- Emergency Medicine Research Group
Edinburgh (EMERGE), Department of Emergency Medicine, Royal Infirmary of
Edinburgh, Edinburgh, UK
| | - Julia Grahamslaw
- Emergency Medicine Research Group
Edinburgh (EMERGE), Department of Emergency Medicine, Royal Infirmary of
Edinburgh, Edinburgh, UK
| | - Katherine Oatey
- Edinburgh Clinical Trials Unit,
Usher Institute, University of Edinburgh, Edinburgh, UK
| | - John Norrie
- Department of Critical Care, Royal
Infirmary of Edinburgh, Usher Institute, University of
Edinburgh, Edinburgh, UK
| | - Nazir Lone
- Department of Critical Care, Royal
Infirmary of Edinburgh, Usher Institute, University of
Edinburgh, Edinburgh, UK
| | - Timothy Walsh
- Department of Critical Care, Royal
Infirmary of Edinburgh, Usher Institute, University of
Edinburgh, Edinburgh, UK
| | - Daniel Horner
- Emergency Department, Salford Royal NHS Foundation
Trust, Salford, UK
- Division of Infection, Immunity and
Respiratory Medicine, University of
Manchester, Manchester, UK
| | - Andy Appelboam
- Academic Department of Emergency
Medicine Exeter (ACADEMEx), Royal Devon and Exeter Hospital NHS
Foundation Trust, Exeter Devon
| | - Peter Hall
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, UK
| | | | - Derek Bell
- Department of Acute Medicine, Chelsea and Westminster
Campus, Imperial College London, London
| | | | - Manu Shankar-Hari
- The Queen’s Medical Research
Institute, Edinburgh BioQuarter, Centre for
Inflammation Research, University of Edinburgh, UK
| | - Alasdair Corfield
- Emergency Department, Royal Alexandra
Hospital, NHS Greater Glasgow and Clyde, UK
| | - Alasdair Gray
- Emergency Medicine Research Group
Edinburgh (EMERGE), Department of Emergency Medicine, Royal Infirmary of
Edinburgh, Edinburgh, UK
- Acute Care Edinburgh, Centre for
Population and Health Sciences, Usher Institute, University of
Edinburgh, Edinburgh UK
| |
Collapse
|
17
|
Baghela A, An A, Zhang P, Acton E, Gauthier J, Brunet-Ratnasingham E, Blimkie T, Freue GC, Kaufmann D, Lee AHY, Levesque RC, Hancock REW. Predicting severity in COVID-19 disease using sepsis blood gene expression signatures. Sci Rep 2023; 13:1247. [PMID: 36690713 PMCID: PMC9868505 DOI: 10.1038/s41598-023-28259-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Severely-afflicted COVID-19 patients can exhibit disease manifestations representative of sepsis, including acute respiratory distress syndrome and multiple organ failure. We hypothesized that diagnostic tools used in managing all-cause sepsis, such as clinical criteria, biomarkers, and gene expression signatures, should extend to COVID-19 patients. Here we analyzed the whole blood transcriptome of 124 early (1-5 days post-hospital admission) and late (6-20 days post-admission) sampled patients with confirmed COVID-19 infections from hospitals in Quebec, Canada. Mechanisms associated with COVID-19 severity were identified between severity groups (ranging from mild disease to the requirement for mechanical ventilation and mortality), and established sepsis signatures were assessed for dysregulation. Specifically, gene expression signatures representing pathophysiological events, namely cellular reprogramming, organ dysfunction, and mortality, were significantly enriched and predictive of severity and lethality in COVID-19 patients. Mechanistic endotypes reflective of distinct sepsis aetiologies and therapeutic opportunities were also identified in subsets of patients, enabling prediction of potentially-effective repurposed drugs. The expression of sepsis gene expression signatures in severely-afflicted COVID-19 patients indicates that these patients should be classified as having severe sepsis. Accordingly, in severe COVID-19 patients, these signatures should be strongly considered for the mechanistic characterization, diagnosis, and guidance of treatment using repurposed drugs.
Collapse
Affiliation(s)
- Arjun Baghela
- Department of Microbiology and Immunology, University of British Columbia (UBC), Vancouver, Canada
| | - Andy An
- Department of Microbiology and Immunology, University of British Columbia (UBC), Vancouver, Canada
| | | | - Erica Acton
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Jeff Gauthier
- Institut de Biologie Intégrative et des Systèmes (IBIS), Département de Microbiologie-Infectiologie et d'immunologie, Université Laval, Quebec, QC, Canada
| | - Elsa Brunet-Ratnasingham
- Département de Microbiologie, Infectiologie Et Immunologie, Université de Montréal, Montreal, Canada
- Centre de Recherche du CHUM, Montreal, QC, Canada
| | - Travis Blimkie
- Department of Microbiology and Immunology, University of British Columbia (UBC), Vancouver, Canada
| | | | - Daniel Kaufmann
- Centre de Recherche du CHUM, Montreal, QC, Canada
- Département de Médecine, Université de Montréal, Montreal, Canada
| | - Amy H Y Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Roger C Levesque
- Institut de Biologie Intégrative et des Systèmes (IBIS), Département de Microbiologie-Infectiologie et d'immunologie, Université Laval, Quebec, QC, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia (UBC), Vancouver, Canada.
- Asep Medical, Vancouver, Canada.
| |
Collapse
|
18
|
Montero MM, Hardy-Werbin M, Gonzalez-Gallardo S, Torres E, Rueda R, Hannet I, Kirk JT, Yager TD, Navalkar K, Arenas MDM, Arietta-Aldea I, Castañeda S, Gómez-Junyent J, Gómez-Zorrilla S, Guerri-Fernandez R, Sanchez-Martinez F, López-Montesinos I, Pelegrín I, Sendra E, Sorlí L, Villar-García J, Bellosillo B, Horcajada JP. Evaluation of the host immune response assay SeptiCyte RAPID for potential triage of COVID-19 patients. Sci Rep 2023; 13:944. [PMID: 36653401 PMCID: PMC9845827 DOI: 10.1038/s41598-023-28178-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Tools for the evaluation of COVID-19 severity would help clinicians with triage decisions, especially the decision whether to admit to ICU. The aim of this study was to evaluate SeptiCyte RAPID, a host immune response assay (Immunexpress, Seattle USA) as a triaging tool for COVID-19 patients requiring hospitalization and potentially ICU care. SeptiCyte RAPID employs a host gene expression signature consisting of the ratio of expression levels of two immune related mRNAs, PLA2G7 and PLAC8, measured from whole blood samples. Blood samples from 146 adult SARS-CoV-2 (+) patients were collected within 48 h of hospital admission in PAXgene blood RNA tubes at Hospital del Mar, Barcelona, Spain, between July 28th and December 1st, 2020. Data on demographics, vital signs, clinical chemistry parameters, radiology, interventions, and SeptiCyte RAPID were collected and analyzed with bioinformatics methods. The performance of SeptiCyte RAPID for COVID-19 severity assessment and ICU admission was evaluated, relative to the comparator of retrospective clinical assessment by the Hospital del Mar clinical care team. In conclusion, SeptiCyte RAPID was able to stratify COVID-19 cases according to clinical severity: critical vs. mild (AUC = 0.93, p < 0.0001), critical vs. moderate (AUC = 0.77, p = 0.002), severe vs. mild (AUC = 0.85, p = 0.0003), severe vs. moderate (AUC = 0.63, p = 0.05). This discrimination was significantly better (by AUC or p-value) than could be achieved by CRP, lactate, creatine, IL-6, or D-dimer. Some of the critical or severe cases had "early" blood draws (before ICU admission; n = 33). For these cases, when compared to moderate and mild cases not in ICU (n = 37), SeptiCyte RAPID had AUC = 0.78 (p = 0.00012). In conclusion, SeptiCyte RAPID was able to stratify COVID-19 cases according to clinical severity as defined by the WHO COVID-19 Clinical Management Living Guidance of January 25th, 2021. Measurements taken early (before a patient is considered for ICU admission) suggest that high SeptiScores could aid in predicting the need for later ICU admission.
Collapse
Affiliation(s)
- Maria Milagro Montero
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, 08002, Barcelona, Spain.,CIBER of Infectious Diseases, Institute of Health Carlos III, Madrid, Spain
| | - Max Hardy-Werbin
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Emergency Department, Hospital del Mar, Barcelona, Spain
| | | | - Erica Torres
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | - Rebeca Rueda
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | | | | | | | | | - Maria Del Mar Arenas
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Itziar Arietta-Aldea
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Silvia Castañeda
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Joan Gómez-Junyent
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Silvia Gómez-Zorrilla
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, 08002, Barcelona, Spain.,CIBER of Infectious Diseases, Institute of Health Carlos III, Madrid, Spain
| | - Roberto Guerri-Fernandez
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, 08002, Barcelona, Spain.,CIBER of Infectious Diseases, Institute of Health Carlos III, Madrid, Spain
| | - Francisca Sanchez-Martinez
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, 08002, Barcelona, Spain
| | - Immaculada López-Montesinos
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Ivan Pelegrín
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Elena Sendra
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Luisa Sorlí
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, 08002, Barcelona, Spain.,CIBER of Infectious Diseases, Institute of Health Carlos III, Madrid, Spain
| | - Judith Villar-García
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, 08002, Barcelona, Spain
| | - Beatriz Bellosillo
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain. .,Pathology Department, Hospital del Mar, Barcelona, Spain.
| | - Juan Pablo Horcajada
- Infectious Disease Department, Hospital del Mar, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra Barcelona, 08002, Barcelona, Spain.,CIBER of Infectious Diseases, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Yih WK, Daley MF, Duffy J, Fireman B, McClure D, Nelson J, Qian L, Smith N, Vazquez-Benitez G, Weintraub E, Williams JTB, Xu S, Maro JC. A broad assessment of covid-19 vaccine safety using tree-based data-mining in the vaccine safety datalink. Vaccine 2023; 41:826-835. [PMID: 36535825 PMCID: PMC9755007 DOI: 10.1016/j.vaccine.2022.12.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/18/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Except for spontaneous reporting systems, vaccine safety monitoring generally involves pre-specifying health outcomes and post-vaccination risk windows of concern. Instead, we used tree-based data-mining to look more broadly for possible adverse events after Pfizer-BioNTech, Moderna, and Janssen COVID-19 vaccination. METHODS Vaccine Safety Datalink enrollees receiving ≥1 dose of COVID-19 vaccine in 2020-2021 were followed for 70 days after Pfizer-BioNTech or Moderna and 56 days after Janssen vaccination. Incident diagnoses in inpatient or emergency department settings were analyzed for clustering within both the hierarchical ICD-10-CM code structure and the post-vaccination follow-up period. We used the self-controlled tree-temporal scan statistic and TreeScan software. Monte Carlo simulation was used to estimate p-values; p = 0.01 was the pre-specified cut-off for statistical significance of a cluster. RESULTS There were 4.1, 2.6, and 0.4 million Pfizer-BioNTech, Moderna, and Janssen vaccinees, respectively. Clusters after Pfizer-BioNTech vaccination included: (1) unspecified adverse effects, (2) common vaccine reactions, such as fever, myalgia, and headache, (3) myocarditis/pericarditis, and (4) less specific cardiac or respiratory symptoms, all with the strongest clusters generally after Dose 2; and (5) COVID-19/viral pneumonia/sepsis/respiratory failure in the first 3 weeks after Dose 1. Moderna results were similar but without a significant myocarditis/pericarditis cluster. Further investigation suggested the fifth signal group was a manifestation of mRNA vaccine effectiveness after the first 3 weeks. Janssen vaccinees had clusters of unspecified or common vaccine reactions, gait/mobility abnormalities, and muscle weakness. The latter two were deemed to have arisen from confounding related to practices at one site. CONCLUSIONS We detected post-vaccination clusters of unspecified adverse effects, common vaccine reactions, and, for the mRNA vaccines, chest pain and palpitations, as well as myocarditis/pericarditis after Pfizer-BioNTech Dose 2. Unique advantages of this data mining are its untargeted nature and its inherent adjustment for the multiplicity of diagnoses and risk intervals scanned.
Collapse
Affiliation(s)
- W Katherine Yih
- Harvard Pilgrim Health Care Institute and Department of Population Medicine, Harvard Medical School, Boston, MA, United States
| | | | - Jonathan Duffy
- Immunization Safety Office, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Bruce Fireman
- Kaiser Permanente Northern California, Oakland, CA, United States
| | - David McClure
- Marshfield Clinic Research Institute, Marshfield, WI, United States
| | | | - Lei Qian
- Kaiser Permanente Southern California, Pasadena, CA, United States
| | - Ning Smith
- Kaiser Permanente Northwest, Portland, OR, United States
| | | | - Eric Weintraub
- Immunization Safety Office, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | | | - Stanley Xu
- Kaiser Permanente Southern California, Pasadena, CA, United States
| | - Judith C Maro
- Harvard Pilgrim Health Care Institute and Department of Population Medicine, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
20
|
An AY, Baghela A, Zhang P, Falsafi R, Lee AH, Trahtemberg U, Baker AJ, dos Santos CC, Hancock REW. Severe COVID-19 and non-COVID-19 severe sepsis converge transcriptionally after a week in the intensive care unit, indicating common disease mechanisms. Front Immunol 2023; 14:1167917. [PMID: 37090709 PMCID: PMC10115984 DOI: 10.3389/fimmu.2023.1167917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
Introduction Severe COVID-19 and non-COVID-19 pulmonary sepsis share pathophysiological, immunological, and clinical features. To what extent they share mechanistically-based gene expression trajectories throughout hospitalization was unknown. Our objective was to compare gene expression trajectories between severe COVID-19 patients and contemporaneous non-COVID-19 severe sepsis patients in the intensive care unit (ICU). Methods In this prospective single-center observational cohort study, whole blood was drawn from 20 COVID-19 patients and 22 non-COVID-19 adult sepsis patients at two timepoints: ICU admission and approximately a week later. RNA-Seq was performed on whole blood to identify differentially expressed genes and significantly enriched pathways. Results At ICU admission, despite COVID-19 patients being almost clinically indistinguishable from non-COVID-19 sepsis patients, COVID-19 patients had 1,215 differentially expressed genes compared to non-COVID-19 sepsis patients. After one week in the ICU, the number of differentially expressed genes dropped to just 9 genes. This drop coincided with decreased expression of antiviral genes and relatively increased expression of heme metabolism genes over time in COVID-19 patients, eventually reaching expression levels seen in non-COVID-19 sepsis patients. Both groups also had similar underlying immune dysfunction, with upregulation of immune processes such as "Interleukin-1 signaling" and "Interleukin-6/JAK/STAT3 signaling" throughout disease compared to healthy controls. Discussion Early on, COVID-19 patients had elevated antiviral responses and suppressed heme metabolism processes compared to non-COVID-19 severe sepsis patients, although both had similar underlying immune dysfunction. However, after one week in the ICU, these diseases became indistinguishable on a gene expression level. These findings highlight the importance of early antiviral treatment for COVID-19, the potential for heme-related therapeutics, and consideration of immunomodulatory therapies for both diseases to treat shared immune dysfunction.
Collapse
Affiliation(s)
- Andy Y. An
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Arjun Baghela
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Peter Zhang
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Reza Falsafi
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Amy H. Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Uriel Trahtemberg
- The Department of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
- Department of Critical Care, Galilee Medical Center, Nahariya, Israel
| | - Andrew J. Baker
- The Department of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Claudia C. dos Santos
- The Department of Critical Care, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, University of Toronto, Toronto, ON, Canada
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Robert E. W. Hancock,
| |
Collapse
|
21
|
Zhang B, Yang Q, Wang X, Jia B, Cheng L, Zeng HL. Association of whole blood metals/metalloids with severity in sepsis patients: A prospective, single-center, pilot study. J Trace Elem Med Biol 2023; 75:127098. [PMID: 36335678 DOI: 10.1016/j.jtemb.2022.127098] [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: 06/09/2022] [Revised: 09/26/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Trace metals/metalloids were important for the biological functions of both the eukaryotic host and the microorganism. Their concentrations and variations may associate with the critical illness in sepsis, which still needs to be investigated. METHODS We performed a prospective cohort study on the patients with sepsis admitted to Tongji hospital (Wuhan, China) from Jul 01 to Dec 31, 2021. Sepsis was diagnosed in accordance with the third international consensus definitions for sepsis and septic shock (Sepsis-3). The concentrations of metals/metalloids including magnesium (Mg), calcium (Ca), chromium (Cr), manganese (Mn), iron (Fe), copper (Cu), zinc (Zn), arsenic (As), cadmium (Cd), mercury (Hg), thallium (Tl) and lead (Pb) in whole blood were analyzed by ICP-MS based methods. RESULTS Compared to the healthy controls, patients with sepsis showed higher levels of Ca, Cr and Cu, and lower levels of Mg, Mn, Fe, Zn, As, Hg and Pb. Further analysis between the critical illness and noncritical illness, revealed the Mn, Fe were significantly lower in the critically-ill sepsis. The longitudinal profile of the two metals show the differences appeared to exist almost throughout the clinical course. By performing the binary regression logistic analysis, we determined the Fe, Mn as independently risk factors for critical illness in sepsis, with effect sizes (β) of 17.14 (95%CI: 1.79-163.81) and 10.83 (1.96-59.83), respectively, which collectively discriminated 83.3% of all cases between critical-illness and non-critically illness. CONCLUSIONS The variations of whole blood metals/metalloids were associated with the critically-ill sepsis.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Yang
- Institute of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Xu Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingmei Jia
- Institute of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Liming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hao-Long Zeng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
22
|
Krawczyk N, Rivera BD, Basaraba C, Corbeil T, Allen B, Schultebraucks K, Henry BF, Pincus HA, Levin FR, Martinez D. COVID-19 complications among patients with opioid use disorder: a retrospective cohort study across five major NYC hospital systems. Addiction 2022; 118:857-869. [PMID: 36459420 PMCID: PMC9878119 DOI: 10.1111/add.16105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND AND AIMS Individuals with opioid use disorder (OUD) suffer disproportionately from COVID-19. To inform clinical management of OUD patients, research is needed to identify characteristics associated with COVID-19 progression and death among this population. We aimed to investigate the role of OUD and specific comorbidities on COVID-19 progression among hospitalized OUD patients. DESIGN Retrospective cohort study of merged electronic health records (EHR) from five large private health systems. SETTING New York City, New York, USA, 2011-21. PARTICIPANTS Adults with a COVID-19 encounter and OUD or opioid overdose diagnosis between March 2020 and February 2021. MEASUREMENTS Primary exposure included diagnosis of OUD/opioid overdose. Risk factors included age, sex, race/ethnicity and common medical, substance use and psychiatric comorbidities known to be associated with COVID-19 severity. Outcomes included COVID-19 hospitalization and subsequent intubation, acute kidney failure, severe sepsis and death. FINDINGS Of 110 917 COVID-19+ adults, 1.17% were ever diagnosed with OUD/opioid overdose. OUD patients had higher risk of COVID-19 hospitalization [adjusted risk ratio (aRR) = 1.40, 95% confidence interval (CI) = 1.33, 1.47], intubation [adjusted odds ratio (aOR) = 2.05, 95% CI = 1.74, 2.42], kidney failure (aRR = 1.51, 95% CI = 1.34, 1.70), sepsis (aRR = 2.30, 95% CI = 1.88, 2.81) and death (aRR = 2.10, 95% CI = 1.84, 2.40). Among hospitalized OUD patients, risks for worse COVID-19 outcomes included being male; older; of a race/ethnicity other than white, black or Hispanic; and having comorbid chronic kidney disease, diabetes, obesity or cancer. Protective factors included having asthma, hepatitis-C and chronic pain. CONCLUSIONS Opioid use disorder patients appear to have a substantial risk for COVID-19-associated morbidity and mortality, with particular comorbidities and treatments moderating this risk.
Collapse
Affiliation(s)
- Noa Krawczyk
- Center for Opioid Epidemiology and Policy, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Bianca D Rivera
- Center for Opioid Epidemiology and Policy, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Cale Basaraba
- Area Mental Health Data Science, New York State Psychiatric Institute, New York, NY, USA
| | - Thomas Corbeil
- Area Mental Health Data Science, New York State Psychiatric Institute, New York, NY, USA
| | - Bennett Allen
- Center for Opioid Epidemiology and Policy, Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Brandy F Henry
- College of Education, The Pennsylvania State University, University Park, PA, USA
| | - Harold A Pincus
- Department of Psychiatry and Irving Institute for Clinical and Translational Research, Columbia University and New York State Psychiatric Institute, New York, NY, USA
| | - Frances R Levin
- Department of Psychiatry and Irving Institute for Clinical and Translational Research, Columbia University and New York State Psychiatric Institute, New York, NY, USA
| | - Diana Martinez
- Department of Psychiatry and Irving Institute for Clinical and Translational Research, Columbia University and New York State Psychiatric Institute, New York, NY, USA
| |
Collapse
|
23
|
Walsh TJ, Bright RA, Ahuja A, McCarthy MW, Marfuggi RA, Simpson SQ. Meeting the Challenges of Sepsis in Severe Coronavirus Disease 2019: A Call to Arms. Open Forum Infect Dis 2022; 10:ofac645. [PMID: 36686626 PMCID: PMC9850274 DOI: 10.1093/ofid/ofac645] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Sepsis is a life-threatening organ dysfunction that is caused by a dysregulated host response to infection. Sepsis may be caused by bacterial, fungal, or viral pathogens. The clinical manifestations exhibited by patients with severe coronavirus disease 2019 (COVID-19)-related sepsis overlap with those exhibited by patients with sepsis from secondary bacterial or fungal infections and can include an altered mental status, dyspnea, reduced urine output, tachycardia, and hypotension. Critically ill patients hospitalized with severe acute respiratory syndrome coronavirus 2 infections have increased risk for secondary bacterial and fungal infections. The same risk factors that may predispose to sepsis and poor outcome from bloodstream infections (BSIs) converge in patients with severe COVID-19. Current diagnostic standards for distinguishing between (1) patients who are critically ill, septic, and have COVID-19 and (2) patients with sepsis from other causes leave healthcare providers with 2 suboptimal choices. The first choice is to empirically administer broad-spectrum, antimicrobial therapy for what may or may not be sepsis. Such treatment may not only be ineffective and inappropriate, but it also has the potential to cause harm. The development of better methods to identify and characterize antimicrobial susceptibility will guide more accurate therapeutic interventions and reduce the evolution of new antibiotic-resistant strains. The ideal diagnostic test should (1) be rapid and reliable, (2) have a lower limit of detection than blood culture, and (3) be able to detect a specific organism and drug sensitivity directly from a clinical specimen. Rapid direct detection of antimicrobial-resistant pathogens would allow targeted therapy and result in improved outcomes in patients with severe COVID-19 and sepsis.
Collapse
Affiliation(s)
- Thomas J Walsh
- Correspondence: Thomas J. Walsh MD, PhD, Center for Innovative Therapeutics and Diagnostics, 6641 West Broad Street, Room 100, Richmond, Virginia 23220 ()
| | - Rick A Bright
- The Rockefeller Foundation, Pandemic Prevention Institute, New York, New York, USA
| | | | - Matthew W McCarthy
- Weill Cornell Medicine and New York Presbyterian Hospital, New York, New York, USA
| | - Richard A Marfuggi
- American Medical Association Foundation, Chicago, Illinois, USA,WBB Research Institute, Cranford, New Jersey, USA
| | | |
Collapse
|
24
|
Brusa S, Terracciano D, Bruzzese D, Fiorenza M, Stanziola L, Pinchera B, Valente V, Gentile I, Cittadini A, Mormile I, Mormile M, Portella G. Circulating tissue inhibitor of metalloproteinases 1 (TIMP-1) at COVID-19 onset predicts severity status. Front Med (Lausanne) 2022; 9:1034288. [PMID: 36523781 PMCID: PMC9744795 DOI: 10.3389/fmed.2022.1034288] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/15/2022] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Systemic biomarkers for severity of SARS-CoV-2 infection are of great interest. In this study, we evaluated a set of collagen metabolites and extracellular matrix remodeling biomarkers including procollagen type III amino terminal propeptide (PIIINP), tissue inhibitor of metalloproteinases 1 (TIMP-1) and hyaluronic acid (HA) as prognostic indicators in COVID-19 patients. METHODS Ninety COVID-19 patients with the absence of chronic liver diseases were enrolled. Serum PIIINP, TIMP-1, and HA were measured and correlated with inflammatory indices and clinical variables. Patients were stratified for disease severity according to WHO criteria in two groups, based on the requirement of oxygen support. RESULTS Serum TIMP-1, but not PIIINP and HA was significantly higher in patients with WHO score ≥5 compared to patients with WHO score <5 [PIIINP: 7.2 (5.4-9.5) vs. 7.1 (4.5-9.9), p = 0.782; TIMP-1: 298.1 (20.5-460) vs. 222.2 (28.5-452.8), p = 0.01; HA: 117.1 (55.4-193.7) vs. 75.1 (36.9-141.8), p = 0.258]. TIMP-1 showed moderate correlation with CRP (r = 0.312, p = 0.003) and with LDH (r = 0.263, p = 0.009). CRP and serum LDH levels were significantly higher in COVID-19 patients with WHO score ≥5 compared to the group of patients with WHO score < 5 [15.8 (9-44.5) vs. 9.3 (3.4-33.8), p = 0.039 and 373 (282-465) vs. 289 (218-383), p = 0.013, respectively]. CONCLUSION In patients with COVID-19, circulating TIMP-1 was associated with disease severity and with systemic inflammatory index, suggesting that TIMP-1 could represent a promising non-invasive prognostic biomarker in COVID-19 patients. Interestingly, our results prompted that serum TIMP-1 level may potentially be used to select the patients for therapeutic approaches targeting matrix metalloproteases pathway.
Collapse
Affiliation(s)
- Stefano Brusa
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Daniela Terracciano
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Dario Bruzzese
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mariano Fiorenza
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Lucia Stanziola
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Biagio Pinchera
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Valeria Valente
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Ivan Gentile
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Antonio Cittadini
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Ilaria Mormile
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Mauro Mormile
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Giuseppe Portella
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| |
Collapse
|
25
|
Labarrere CA, Kassab GS. Glutathione: A Samsonian life-sustaining small molecule that protects against oxidative stress, ageing and damaging inflammation. Front Nutr 2022; 9:1007816. [PMID: 36386929 PMCID: PMC9664149 DOI: 10.3389/fnut.2022.1007816] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/12/2022] [Indexed: 11/26/2022] Open
Abstract
Many local and systemic diseases especially diseases that are leading causes of death globally like chronic obstructive pulmonary disease, atherosclerosis with ischemic heart disease and stroke, cancer and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causing coronavirus disease 19 (COVID-19), involve both, (1) oxidative stress with excessive production of reactive oxygen species (ROS) that lower glutathione (GSH) levels, and (2) inflammation. The GSH tripeptide (γ- L-glutamyl-L-cysteinyl-glycine), the most abundant water-soluble non-protein thiol in the cell (1-10 mM) is fundamental for life by (a) sustaining the adequate redox cell signaling needed to maintain physiologic levels of oxidative stress fundamental to control life processes, and (b) limiting excessive oxidative stress that causes cell and tissue damage. GSH activity is facilitated by activation of the Kelch-like ECH-associated protein 1 (Keap1)-Nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) redox regulator pathway, releasing Nrf2 that regulates expression of genes controlling antioxidant, inflammatory and immune system responses. GSH exists in the thiol-reduced (>98% of total GSH) and disulfide-oxidized (GSSG) forms, and the concentrations of GSH and GSSG and their molar ratio are indicators of the functionality of the cell. GSH depletion may play a central role in inflammatory diseases and COVID-19 pathophysiology, host immune response and disease severity and mortality. Therapies enhancing GSH could become a cornerstone to reduce severity and fatal outcomes of inflammatory diseases and COVID-19 and increasing GSH levels may prevent and subdue these diseases. The life value of GSH makes for a paramount research field in biology and medicine and may be key against systemic inflammation and SARS-CoV-2 infection and COVID-19 disease. In this review, we emphasize on (1) GSH depletion as a fundamental risk factor for diseases like chronic obstructive pulmonary disease and atherosclerosis (ischemic heart disease and stroke), (2) importance of oxidative stress and antioxidants in SARS-CoV-2 infection and COVID-19 disease, (3) significance of GSH to counteract persistent damaging inflammation, inflammaging and early (premature) inflammaging associated with cell and tissue damage caused by excessive oxidative stress and lack of adequate antioxidant defenses in younger individuals, and (4) new therapies that include antioxidant defenses restoration.
Collapse
|
26
|
Reprograming of Gene Expression of Key Inflammatory Signaling Pathways in Human Peripheral Blood Mononuclear Cells by Soybean Lectin and Resveratrol. Int J Mol Sci 2022; 23:ijms232112946. [DOI: 10.3390/ijms232112946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammation is linked to several human diseases like microbial infections, cancer, heart disease, asthma, diabetes, and neurological disorders. We have shown that the prototype inflammatory agonist LPS modulates the activity of Ubiquitin-Proteasome System (UPS) and regulates transcription factors such as NF-κB, leading to inflammation, tolerance, hypoxia, autophagy, and apoptosis of cells. We hypothesized that proteasome modulators resveratrol and soybean lectin would alter the gene expression of mediators involved in inflammation-induced signaling pathways, when administered ex vivo to human peripheral blood mononuclear blood cells (PBMCs) obtained from normal healthy controls. To test this hypothesis, analysis of RNA derived from LPS-treated human PBMCs, with or without resveratrol and soybean lectin, was carried out using Next Generation Sequencing (NGS). Collectively, the findings described herein suggest that proteasome modulators, resveratrol (proteasome inhibitor) and lectins (proteasome activator), have a profound capacity to modulate cytokine expression in response to proteasome modulators, as well as expression of mediators in multiple signaling pathways in PBMCs of control subjects. We show for the first-time that resveratrol downregulates expression of mediators involved in several key signaling pathways IFN-γ, IL-4, PSMB8 (LMP7), and a subset of LPS-induced genes, while lectins induced IFN-γ, IL-4, PSMB8, and many of the same genes as LPS that are important for innate and adaptive immunity. These findings suggest that inflammation may be influenced by common dietary components and this knowledge may be used to prevent or reverse inflammation-based diseases.
Collapse
|
27
|
Labarrere CA, Kassab GS. Glutathione deficiency in the pathogenesis of SARS-CoV-2 infection and its effects upon the host immune response in severe COVID-19 disease. Front Microbiol 2022; 13:979719. [PMID: 36274722 PMCID: PMC9582773 DOI: 10.3389/fmicb.2022.979719] [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: 06/27/2022] [Accepted: 09/14/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes coronavirus disease 19 (COVID-19) has numerous risk factors leading to severe disease with high mortality rate. Oxidative stress with excessive production of reactive oxygen species (ROS) that lower glutathione (GSH) levels seems to be a common pathway associated with the high COVID-19 mortality. GSH is a unique small but powerful molecule paramount for life. It sustains adequate redox cell signaling since a physiologic level of oxidative stress is fundamental for controlling life processes via redox signaling, but excessive oxidation causes cell and tissue damage. The water-soluble GSH tripeptide (γ-L-glutamyl-L-cysteinyl-glycine) is present in the cytoplasm of all cells. GSH is at 1-10 mM concentrations in all mammalian tissues (highest concentration in liver) as the most abundant non-protein thiol that protects against excessive oxidative stress. Oxidative stress also activates the Kelch-like ECH-associated protein 1 (Keap1)-Nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) redox regulator pathway, releasing Nrf2 to regulate the expression of genes that control antioxidant, inflammatory and immune system responses, facilitating GSH activity. GSH exists in the thiol-reduced and disulfide-oxidized (GSSG) forms. Reduced GSH is the prevailing form accounting for >98% of total GSH. The concentrations of GSH and GSSG and their molar ratio are indicators of the functionality of the cell and its alteration is related to various human pathological processes including COVID-19. Oxidative stress plays a prominent role in SARS-CoV-2 infection following recognition of the viral S-protein by angiotensin converting enzyme-2 receptor and pattern recognition receptors like toll-like receptors 2 and 4, and activation of transcription factors like nuclear factor kappa B, that subsequently activate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) expression succeeded by ROS production. GSH depletion may have a fundamental role in COVID-19 pathophysiology, host immune response and disease severity and mortality. Therapies enhancing GSH could become a cornerstone to reduce severity and fatal outcomes of COVID-19 disease and increasing GSH levels may prevent and subdue the disease. The life value of GSH makes for a paramount research field in biology and medicine and may be key against SARS-CoV-2 infection and COVID-19 disease.
Collapse
|
28
|
Smeller L, Toth-Molnar E, Sohar N. White Dot Syndrome Report in a SARS-CoV-2 Patient. Case Rep Ophthalmol 2022; 13:744-750. [PMID: 36845457 PMCID: PMC9944209 DOI: 10.1159/000526090] [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: 02/22/2022] [Accepted: 06/22/2022] [Indexed: 11/19/2022] Open
Abstract
Our purpose was to report clinical features in bilateral white dot syndrome in a 47-year-old female patient who was tested positive for the SARS-CoV-2. A 47-year-old female visited our department with complaints of bilateral photophobia and blurred vision in both her eyes. She visited our department during the pandemic period after her PCR-proven SARS-CoV-2 positivity. Her symptoms were chills and fever with a temperature of 40.0°C, associated with fatigue, sweat, and complete loss of taste. Besides basic ophthalmological examinations, ocular diagnostic testing were made to differentiate between specific white dot syndromes with suggestive features of fluorescein angiography, optical coherence tomography, and fundus autofluorescence. Laboratory tests were ordered, including immunserological and haematological ones. Eye examination revealed mild bilateral vitritis and white dots in the fundus of both eyes, including the macula explaining the blurred vision. Herpes simplex virus reactivation was proved, after the SARS-CoV-2 infection. Local corticosteroids were given according to the European Reference Network's recommendations for patients with uveitis during the COVID-19 pandemic. Our report demonstrates that white dot syndrome with blurred vision could be associated with SARS-CoV-2 infection, being potentially sight-threatening because of macular involvement. Ophthalmological examinations found posterior uveitis white dot syndrome, and this should call attention to the risk of acute 2019-CoV infection or occurred 2019-CoV infection. Immunodeficiency favours the occurrence of other viral infections, such as herpes virus infections. Everybody should be aware of the risk of 2019-CoV infection, especially professionals, social workers, and those who work or live with elder people and people with immunodeficiency.
Collapse
Affiliation(s)
- Lilla Smeller
- Department of Ophthalmology, University of Szeged, Szeged, Hungary
| | - Edit Toth-Molnar
- Department of Ophthalmology, University of Szeged, Szeged, Hungary
| | | |
Collapse
|
29
|
Wu Q, Pennini ME, Bergmann JN, Kozak ML, Herring K, Sciarretta KL, Armstrong KL. Applying lessons learned from COVID-19 therapeutic trials to improve future ALI/ARDS trials. Open Forum Infect Dis 2022; 9:ofac381. [PMID: 35983268 PMCID: PMC9379817 DOI: 10.1093/ofid/ofac381] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Host-directed therapeutics targeting immune dysregulation are considered the most promising approach to address the unmet clinical need for acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) related to coronavirus disease 2019 (COVID-19). To better understand the current clinical study landscape and gaps in treating hospitalized patients with severe or critical COVID-19, we identified COVID-19 trials developing host-directed therapies registered at ClinicalTrials.gov and discussed the factors contributing to the success vs failure of these studies. We have learned, instead of the one-size-fits-all approach, future clinical trials evaluating a targeted immunomodulatory agent in heterogeneous patients with ALI/ARDS due to COVID-19 or other infectious diseases can use immune-based biomarkers in addition to clinical and demographic characteristics to improve patient stratification and inform clinical decision-making. Identifying distinct patient subgroups based on immune profiles across the disease trajectory, regardless of the causative pathogen, may accelerate evaluating host-directed therapeutics in trials of ALI/ARDS and related conditions (eg, sepsis).
Collapse
Affiliation(s)
- Qun Wu
- Influenza and Emerging Infectious Diseases Division (IEIDD), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Meghan E Pennini
- Division of Research Innovation and Ventures (DRIVe), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Julie N Bergmann
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Marina L Kozak
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kristen Herring
- Division of Chemical Biological Radiological Nuclear (CBRN), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kimberly L Sciarretta
- Division of Research Innovation and Ventures (DRIVe), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| | - Kimberly L Armstrong
- Influenza and Emerging Infectious Diseases Division (IEIDD), Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS) , Washington, DC , United States of America
| |
Collapse
|
30
|
Beraud M, Hashami SA, Lozano M, Bah A, Keith P. Role of therapeutic plasma exchange in the management of COVID-19-induced cytokine storm syndrome. Transfus Apher Sci 2022; 61:103433. [PMID: 35341691 PMCID: PMC8942460 DOI: 10.1016/j.transci.2022.103433] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/14/2022] [Indexed: 12/23/2022]
Abstract
The risk of mortality in patients with coronavirus disease 2019 (COVID-19) is largely related to an excessive immune response, resulting in a hyperinflammatory and hypercoagulable condition collectively referred to as cytokine storm syndrome (CSS). Management of critically ill patients with COVID-19 has included attempts to abate this process, prevent disease progression, and reduce mortality. In this context, therapeutic plasma exchange (TPE) offers an approach to eliminate inflammatory factors and cytokines, offset the pathologic coagulopathy, and reduce the CSS effects. The aim of this review is to analyze available data on the use of TPE for the treatment of CSS in patients with COVID-19. Systematic searches of PubMed, Scopus and COVID-19 Research were conducted to identify articles published between March 1, 2020 and May 26, 2021 reporting the use of TPE for the treatment of COVID-19-induced CSS. A total of 34 peer-reviewed articles (1 randomized controlled trial, 4 matched case-control series, 15 single-group case series, and 14 case reports), including 267 patients, were selected. Despite the low evidence level of the available data, TPE appeared to be a safe intervention for critically ill patients with COVID-19-induced CSS. Although inconsistencies exist between studies, they showed a general trend for decreased interleukin-6, C-reactive protein, ferritin, D-dimer, and fibrinogen levels and increased lymphocyte counts following TPE, supporting the immunomodulatory effect of this treatment. Moreover, TPE was associated with improvements in clinical outcomes in critically ill patients with COVID-19. While TPE may offer a valuable option to treat patients with COVID-19-induced CSS, high-quality randomized controlled clinical trials are needed to confirm its potential clinical benefits, feasibility, and safety. Moreover, clear criteria should be established to identify patients with CSS who might benefit from TPE.
Collapse
Affiliation(s)
- Mickael Beraud
- Terumo Blood and Cell Technologies Europe NV, Zaventem, Belgium.
| | | | - Miquel Lozano
- Department of Hemotherapy and Hemostasis, ICMHO, University Clinic Hospital, IDIBAPS, University of Barcelona, Barcelona, Catalonia, Spain.
| | - Aicha Bah
- Terumo Blood and Cell Technologies Europe NV, Zaventem, Belgium.
| | - Philip Keith
- Critical Care Medicine, Lexington Medical Center, West Columbia, SC 29169, USA.
| |
Collapse
|
31
|
Ahern DJ, Ai Z, Ainsworth M, Allan C, Allcock A, Angus B, Ansari MA, Arancibia-Cárcamo CV, Aschenbrenner D, Attar M, Baillie JK, Barnes E, Bashford-Rogers R, Bashyal A, Beer S, Berridge G, Beveridge A, Bibi S, Bicanic T, Blackwell L, Bowness P, Brent A, Brown A, Broxholme J, Buck D, Burnham KL, Byrne H, Camara S, Candido Ferreira I, Charles P, Chen W, Chen YL, Chong A, Clutterbuck EA, Coles M, Conlon CP, Cornall R, Cribbs AP, Curion F, Davenport EE, Davidson N, Davis S, Dendrou CA, Dequaire J, Dib L, Docker J, Dold C, Dong T, Downes D, Drakesmith H, Dunachie SJ, Duncan DA, Eijsbouts C, Esnouf R, Espinosa A, Etherington R, Fairfax B, Fairhead R, Fang H, Fassih S, Felle S, Fernandez Mendoza M, Ferreira R, Fischer R, Foord T, Forrow A, Frater J, Fries A, Gallardo Sanchez V, Garner LC, Geeves C, Georgiou D, Godfrey L, Golubchik T, Gomez Vazquez M, Green A, Harper H, Harrington HA, Heilig R, Hester S, Hill J, Hinds C, Hird C, Ho LP, Hoekzema R, Hollis B, Hughes J, Hutton P, Jackson-Wood MA, Jainarayanan A, James-Bott A, Jansen K, Jeffery K, Jones E, Jostins L, Kerr G, Kim D, Klenerman P, Knight JC, Kumar V, Kumar Sharma P, Kurupati P, Kwok A, Lee A, Linder A, Lockett T, Lonie L, Lopopolo M, Lukoseviciute M, Luo J, Marinou S, Marsden B, Martinez J, Matthews PC, Mazurczyk M, McGowan S, McKechnie S, Mead A, Mentzer AJ, Mi Y, Monaco C, Montadon R, Napolitani G, Nassiri I, Novak A, O'Brien DP, O'Connor D, O'Donnell D, Ogg G, Overend L, Park I, Pavord I, Peng Y, Penkava F, Pereira Pinho M, Perez E, Pollard AJ, Powrie F, Psaila B, Quan TP, Repapi E, Revale S, Silva-Reyes L, Richard JB, Rich-Griffin C, Ritter T, Rollier CS, Rowland M, Ruehle F, Salio M, Sansom SN, Sanches Peres R, Santos Delgado A, Sauka-Spengler T, Schwessinger R, Scozzafava G, Screaton G, Seigal A, Semple MG, Sergeant M, Simoglou Karali C, Sims D, Skelly D, Slawinski H, Sobrinodiaz A, Sousos N, Stafford L, Stockdale L, Strickland M, Sumray O, Sun B, Taylor C, Taylor S, Taylor A, Thongjuea S, Thraves H, Todd JA, Tomic A, Tong O, Trebes A, Trzupek D, Tucci FA, Turtle L, Udalova I, Uhlig H, van Grinsven E, Vendrell I, Verheul M, Voda A, Wang G, Wang L, Wang D, Watkinson P, Watson R, Weinberger M, Whalley J, Witty L, Wray K, Xue L, Yeung HY, Yin Z, Young RK, Youngs J, Zhang P, Zurke YX. A blood atlas of COVID-19 defines hallmarks of disease severity and specificity. Cell 2022; 185:916-938.e58. [PMID: 35216673 PMCID: PMC8776501 DOI: 10.1016/j.cell.2022.01.012] [Citation(s) in RCA: 150] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/16/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023]
Abstract
Treatment of severe COVID-19 is currently limited by clinical heterogeneity and incomplete description of specific immune biomarkers. We present here a comprehensive multi-omic blood atlas for patients with varying COVID-19 severity in an integrated comparison with influenza and sepsis patients versus healthy volunteers. We identify immune signatures and correlates of host response. Hallmarks of disease severity involved cells, their inflammatory mediators and networks, including progenitor cells and specific myeloid and lymphocyte subsets, features of the immune repertoire, acute phase response, metabolism, and coagulation. Persisting immune activation involving AP-1/p38MAPK was a specific feature of COVID-19. The plasma proteome enabled sub-phenotyping into patient clusters, predictive of severity and outcome. Systems-based integrative analyses including tensor and matrix decomposition of all modalities revealed feature groupings linked with severity and specificity compared to influenza and sepsis. Our approach and blood atlas will support future drug development, clinical trial design, and personalized medicine approaches for COVID-19.
Collapse
|
32
|
Basra R, Whyte M, Karalliedde J, Vas P. What is the impact of microvascular complications of diabetes on severe COVID-19? Microvasc Res 2022; 140:104310. [PMID: 34979154 PMCID: PMC8719364 DOI: 10.1016/j.mvr.2021.104310] [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: 09/06/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023]
Abstract
Evidence suggests severe coronavirus disease-19 (COVID-19) infection is characterised by pulmonary and systemic microvasculature dysfunction, specifically, acute endothelial injury, hypercoagulation and increased capillary permeability. Diabetes, which is also characterised by vascular injury in itself, confers an increased risk of adverse COVID-19 outcomes. It has been suggested that pre-existing endothelial dysfunction and microvascular disease in diabetes will exacerbate the vascular insults associated with COVID-19 and thus lead to increased severity of COVID-19 infection. In this article, we evaluate the current evidence exploring the impact of microvascular complications, in the form of diabetic retinopathy and nephropathy, in individuals with COVID-19 and diabetes. Future insights gained from exploring the microvascular injury patterns and clinical outcomes may come to influence care delivery algorithms for either of these conditions.
Collapse
Affiliation(s)
- Ruman Basra
- School of Cardiovascular Medicine & Sciences, King's College London, London, UK
| | - Martin Whyte
- Department of Clinical and Experimental Medicine, University of Surrey, Guildford, UK,Department of Diabetes, King's College NHS Foundation Trust, London, UK,King's Health Partners' Institute of Diabetes, Endocrinology and Obesity, London, UK
| | - Janaka Karalliedde
- School of Cardiovascular Medicine & Sciences, King's College London, London, UK,King's Health Partners' Institute of Diabetes, Endocrinology and Obesity, London, UK
| | - Prashanth Vas
- Department of Diabetes, King's College NHS Foundation Trust, London, UK,King's Health Partners' Institute of Diabetes, Endocrinology and Obesity, London, UK,Corresponding author at: Department of Diabetes, King's College Hospital, London SE5 9RS, UK
| |
Collapse
|
33
|
Sun S, Annadi RR, Chaudhri I, Munir K, Hajagos J, Saltz J, Hoai M, Mallipattu SK, Moffitt R, Koraishy FM. Short- and Long-Term Recovery after Moderate/Severe AKI in Patients with and without COVID-19. KIDNEY360 2022; 3:242-257. [PMID: 35373118 PMCID: PMC8967640 DOI: 10.34067/kid.0005342021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/24/2021] [Indexed: 01/10/2023]
Abstract
Background Severe AKI is strongly associated with poor outcomes in coronavirus disease 2019 (COVID-19), but data on renal recovery are lacking. Methods We retrospectively analyzed these associations in 3299 hospitalized patients (1338 with COVID-19 and 1961 with acute respiratory illness but who tested negative for COVID-19). Uni- and multivariable analyses were used to study mortality and recovery after Kidney Disease Improving Global Outcomes Stages 2 and 3 AKI (AKI-2/3), and Machine Learning was used to predict AKI and recovery using admission data. Long-term renal function and other outcomes were studied in a subgroup of AKI-2/3 survivors. Results Among the 172 COVID-19-negative patients with AKI-2/3, 74% had partial and 44% complete renal recovery, whereas 12% died. Among 255 COVID-19 positive patients with AKI-2/3, lower recovery and higher mortality were noted (51% partial renal recovery, 25% complete renal recovery, 24% died). On multivariable analysis, intensive care unit admission and acute respiratory distress syndrome were associated with nonrecovery, and recovery was significantly associated with survival in COVID-19-positive patients. With Machine Learning, we were able to predict recovery from COVID-19-associated AKI-2/3 with an average precision of 0.62, and the strongest predictors of recovery were initial arterial partial pressure of oxygen and carbon dioxide, serum creatinine, potassium, lymphocyte count, and creatine phosphokinase. At 12-month follow-up, among 52 survivors with AKI-2/3, 26% COVID-19-positive and 24% COVID-19-negative patients had incident or progressive CKD. Conclusions Recovery from COVID-19-associated moderate/severe AKI can be predicted using admission data and is associated with severity of respiratory disease and in-hospital death. The risk of CKD might be similar between COVID-19-positive and -negative patients.
Collapse
Affiliation(s)
- Siao Sun
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York
| | - Raji R. Annadi
- Department of Computer Science, Stony Brook University, Stony Brook, New York
| | - Imran Chaudhri
- Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Kiran Munir
- Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Janos Hajagos
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York
| | - Joel Saltz
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York
| | - Minh Hoai
- Department of Computer Science, Stony Brook University, Stony Brook, New York
| | - Sandeep K. Mallipattu
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Richard Moffitt
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York
| | - Farrukh M. Koraishy
- Division of Nephrology and Hypertension, Department of Medicine, Stony Brook University, Stony Brook, New York
| |
Collapse
|
34
|
Jain K, Sharma D, Patidar M, Nandedkar S, Pathak A, Purohit M. Red Cell Distribution Width as a Predictor of Mortality in Patients With Clinical Sepsis: Experience From a Single Rural Center in Central India. CLINICAL PATHOLOGY (THOUSAND OAKS, VENTURA COUNTY, CALIF.) 2022; 15:2632010X221075592. [PMID: 35141523 PMCID: PMC8819754 DOI: 10.1177/2632010x221075592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022]
Abstract
Introduction: Early diagnosis of sepsis and its severity is essential for appropriate
treatment to improve patient survival, especially in resource-limited
settings. The aim of the present study was to study the role of red blood
cell distribution (RDW) as a biomarker for the early detection of severe
sepsis defined clinically and also in the prediction of mortality from
sepsis. Methods: The cross-sectional study included a total of 175 subjects who met the
inclusion criteria for the diagnosis of severe sepsis. After a thorough
clinical examination, blood samples were taken from all patients within
3 hours of presenting the disease. The RDW values and other investigations
were studied on the day of admission compared to other severity markers with
the mortality index of 30 days. Result: The RDW value was significantly higher in patients with severe sepsis and in
non-survivor patients than in survivors (P < .0001).
There was a strong correlation between the SOFA score and RDW in predicting
the disease outcome with the Pearson correlation coefficient of
r = .46. The area under the receiver operating
characteristic curve was found to be 0.852 at a CI of 95% (0.796-0.909) with
RDW 17.15, sensitivity was 88.6% and specificity was 63.5%. There was a
positive correlation with Pearson’s correlation coefficient of
r = .46 between RDW and the SOFA score. Conclusions: RDW can be used as a potential marker for the early detection of severe
sepsis and in the prediction of the outcome. Large multicenter prospective
studies can confirm the utility of this routinely available marker for
patients with sepsis.
Collapse
Affiliation(s)
- Kavita Jain
- Department of Pathology, R.D. Gardi Medical College, Ujjain, India
| | - Darshita Sharma
- Department of Pathology, R.D. Gardi Medical College, Ujjain, India
| | - Mala Patidar
- Department of Pathology, R.D. Gardi Medical College, Ujjain, India
| | | | - Ashish Pathak
- Department of Paediatrics, R.D. Gardi Medical College, Ujjain, India.,Department of Public Health Sciences, Karolinska Institutet, Stockholm, Sweden
| | - Manju Purohit
- Department of Pathology, R.D. Gardi Medical College, Ujjain, India.,Department of Public Health Sciences, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
Chiang KC, Rizk JG, Nelson DJ, Krishnamurti L, Subbian S, Imig JD, Khan I, Reddy ST, Gupta A. Ramatroban for chemoprophylaxis and treatment of COVID-19: David takes on Goliath. Expert Opin Ther Targets 2022; 26:13-28. [PMID: 35068281 PMCID: PMC10119876 DOI: 10.1080/14728222.2022.2031975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/17/2022] [Indexed: 01/08/2023]
Abstract
INTRODUCTION In COVID-19 pneumonia, there is a massive increase in fatty acid levels and lipid mediators with a predominance of cyclooxygenase metabolites, notably TxB2 ≫ PGE2 > PGD2 in the lungs, and 11-dehydro-TxB2, a TxA2 metabolite, in the systemic circulation. While TxA2 stimulates thromboxane prostanoid (TP) receptors, 11-dehydro-TxB2 is a full agonist of DP2 (formerly known as the CRTh2) receptors for PGD2. Anecdotal experience of using ramatroban, a dual receptor antagonist of the TxA2/TP and PGD2/DP2 receptors, demonstrated rapid symptomatic relief from acute respiratory distress and hypoxemia while avoiding hospitalization. AREAS COVERED Evidence supporting the role of TxA2/TP receptors and PGD2/DP2 receptors in causing rapidly progressive lung injury associated with hypoxemia, a maladaptive immune response and thromboinflammation is discussed. An innovative perspective on the dual antagonism of TxA2/TP and PGD2/DP2 receptor signaling as a therapeutic approach in COVID-19 is presented. This paper examines ramatroban an anti-platelet, immunomodulator, and antifibrotic agent for acute and long-haul COVID-19. EXPERT OPINION Ramatroban, a dual blocker of TP and DP2 receptors, has demonstrated efficacy in animal models of respiratory dysfunction, atherosclerosis, thrombosis, and sepsis, as well as preliminary evidence for rapid relief from dyspnea and hypoxemia in COVID-19 pneumonia. Ramatroban merits investigation as a promising antithrombotic and immunomodulatory agent for chemoprophylaxis and treatment.
Collapse
Affiliation(s)
| | - John G. Rizk
- Department of Pharmaceutical Health Services Research, University of Maryland School of Pharmacy, Baltimore, MD, USA
- Arizona State University, Edson College, Phoenix, AZ, USA
| | | | - Lakshmanan Krishnamurti
- Department of Pediatric Hematology and Oncology, Yale School of Medicine, New Haven, CT, USA
| | - Selvakumar Subbian
- Rutgers University, New Jersey Medical School and Public Health Research Institute, Newark, NJ, USA
| | - John D. Imig
- Drug Discovery Center and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Imran Khan
- Department of Pathology and Laboratory Medicine, the University of California at Davis, Sacramento, CA, USA
| | - Srinivasa T. Reddy
- Departments of Medicine, and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Molecular Toxicology Interdepartmental Degree Program, UCLA, Los Angeles, CA, USA
| | - Ajay Gupta
- Charak Foundation, Orange, CA
- Division of Nephrology, Hypertension and Kidney Transplantation, University of California Irvine, Orange, CA, USA
| |
Collapse
|
36
|
Udovicic I, Stanojevic I, Djordjevic D, Zeba S, Rondovic G, Abazovic T, Lazic S, Vojvodic D, To K, Abazovic D, Khan W, Surbatovic M. Immunomonitoring of Monocyte and Neutrophil Function in Critically Ill Patients: From Sepsis and/or Trauma to COVID-19. J Clin Med 2021; 10:jcm10245815. [PMID: 34945111 PMCID: PMC8706110 DOI: 10.3390/jcm10245815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/27/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Immune cells and mediators play a crucial role in the critical care setting but are understudied. This review explores the concept of sepsis and/or injury-induced immunosuppression and immuno-inflammatory response in COVID-19 and reiterates the need for more accurate functional immunomonitoring of monocyte and neutrophil function in these critically ill patients. in addition, the feasibility of circulating and cell-surface immune biomarkers as predictors of infection and/or outcome in critically ill patients is explored. It is clear that, for critically ill, one size does not fit all and that immune phenotyping of critically ill patients may allow the development of a more personalized approach with tailored immunotherapy for the specific patient. In addition, at this point in time, caution is advised regarding the quality of evidence of some COVID-19 studies in the literature.
Collapse
Affiliation(s)
- Ivo Udovicic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Ivan Stanojevic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Dragan Djordjevic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Snjezana Zeba
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Goran Rondovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Tanja Abazovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
| | - Srdjan Lazic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute of Epidemiology, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Danilo Vojvodic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Kendrick To
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 2QQ, UK; (K.T.); (W.K.)
| | - Dzihan Abazovic
- Emergency Medical Centar of Montenegro, Vaka Djurovica bb, 81000 Podgorica, Montenegro;
| | - Wasim Khan
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 2QQ, UK; (K.T.); (W.K.)
| | - Maja Surbatovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Correspondence: ; Tel.: +381-11-2665-125
| |
Collapse
|
37
|
da Silva Ramos FJ, de Freitas FGR, Machado FR. Sepsis in patients hospitalized with coronavirus disease 2019: how often and how severe? Curr Opin Crit Care 2021; 27:474-479. [PMID: 34292175 PMCID: PMC8452249 DOI: 10.1097/mcc.0000000000000861] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW To discuss why severe COVID-19 should be considered sepsis and how co-infection and secondary infection can aggravate this condition and perpetuate organ dysfunction leading to high mortality rates. RECENT FINDINGS In severe COVID-19, there is both direct viral toxicity and dysregulated host response to infection. Although both coinfection and/or secondary infection are present, the latest is of greater concern mainly in resource-poor settings. Patients with severe COVID-19 present a phenotype of multiorgan dysfunction that leads to death in an unacceptable high percentage of the patients, with wide variability around the world. Similarly to endemic sepsis, the mortality of COVID-19 critically ill patients is higher in low-income and middle-income countries as compared with high-income countries. Disparities, including hospital strain, resources limitations, higher incidence of healthcare-associated infections (HAI), and staffing issues could in part explain this variability. SUMMARY The high mortality rates of critically ill patients with severe COVID-19 disease are not only related to the severity of patient disease but also to modifiable factors, such as the ICU strain, HAI incidence, and organizational aspects. Therefore, HAI prevention and the delivery of best evidence-based care for these patients to avoid additional damage is important. Quality improvement interventions might help in improving outcomes mainly in resource-limited settings.
Collapse
|
38
|
Erber J, Wiessner JR, Huberle C, Schneider J, Mijočević H, von Bomhard D, Luppa P, Schmid RM, Rasch S, Lahmer T. Convalescent plasma therapy in B-cell-depleted and B-cell sufficient patients with life-threatening COVID-19 - A case series. Transfus Apher Sci 2021; 60:103278. [PMID: 34548246 PMCID: PMC8447834 DOI: 10.1016/j.transci.2021.103278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/14/2021] [Indexed: 12/30/2022]
Abstract
Objective To investigate the effect of convalescent plasma therapy (CPT) on clinical courses of B-cell-sufficient and B-cell-depleted patients with life-threatening COVID-19. Patients and Methods In this case series, we retrospectively analysed clinical, laboratory and cardiopulmonary parameters of six patients with life-threatening COVID-19 receiving convalescent plasma (CP) as rescue therapy between April 11, 2020 to October 10, 2020. Clinical and laboratory parameters before and after transfusion were compared in two B-cell-depleted patients and four B-cell sufficient patients (control group). Results Both B-cell-depleted patients cleared SARS-CoV-2 virus and survived, while all other patients died within 14 days from intervention despite maximal therapeutic efforts. D-dimer levels increased in both cohorts subsequent to CPT. In control patients, mean Interleukin-6 increased and platelet levels decreased as opposed to decreasing and stable levels in B-cell-depleted patients, respectively. Control patients required increased doses of vasopressor compared to decreasing doses in B-cell depleted patients subsequent to CPT. PO2/FiO2 decrease was more pronounced and respiratory deterioration required postinterventional extracorporeal membrane oxygenation in two control patients. Transpulmonary thermodilution revealed a further increase of the Extravascular Lung Water Index upon CPT in control patients. Conclusion Use of CP in late stages of life-threatening COVID-19 should be used with caution but may be beneficial in B-cell-depleted patients. Further studies are necessary to assess factors predicting potential therapeutic benefits as well as possible hazards.
Collapse
Affiliation(s)
- Johanna Erber
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, 81675 Munich, Germany
| | - Johannes R Wiessner
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, 81675 Munich, Germany
| | - Christina Huberle
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, 81675 Munich, Germany
| | - Jochen Schneider
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, 81675 Munich, Germany
| | - Hrvoje Mijočević
- Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany
| | - Doris von Bomhard
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Institute of Clinical Chemistry and Pathobiochemistry, 81675, Munich, Germany
| | - Peter Luppa
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Institute of Clinical Chemistry and Pathobiochemistry, 81675, Munich, Germany
| | - Roland M Schmid
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, 81675 Munich, Germany
| | - Sebastian Rasch
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, 81675 Munich, Germany
| | - Tobias Lahmer
- Technical University of Munich, School of Medicine, University Hospital rechts der Isar, Department of Internal Medicine II, 81675 Munich, Germany.
| |
Collapse
|
39
|
D Avila-Mesquita C, Couto AES, Campos LCB, Vasconcelos TF, Michelon-Barbosa J, Corsi CAC, Mestriner F, Petroski-Moraes BC, Garbellini-Diab MJ, Couto DMS, Jordani MC, Ferro D, Sbragia L, Joviliano EE, Evora PR, Carvalho Santana RD, Martins-Filho OA, Polonis K, Menegueti MG, Ribeiro MS, Auxiliadora-Martins M, Becari C. MMP-2 and MMP-9 levels in plasma are altered and associated with mortality in COVID-19 patients. Biomed Pharmacother 2021; 142:112067. [PMID: 34449310 PMCID: PMC8376652 DOI: 10.1016/j.biopha.2021.112067] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 01/08/2023] Open
Abstract
Respiratory symptoms are one of COVID-19 manifestations, and the metalloproteinases (MMPs) have essential roles in the lung physiology. We sought to characterize the plasmatic levels of matrix metalloproteinase-2 and 9 (MMP-2 and MMP-9) in patients with severe COVID-19 and to investigate an association between plasma MMP-2 and MMP-9 levels and clinical outcomes and mortality. MMP-2 and MMP-9 levels in plasma from patients with COVID-19 treated in the ICU (COVID-19 group) and Control patients were measured with the zymography. The study groups were matched for age, sex, hypertension, diabetes, BMI, and obesity profile. MMP-2 levels were lower and MMP-9 levels were higher in a COVID-19 group (p < 0.0001) compared to Controls. MMP-9 levels in COVID-19 patients were not affected by comorbidity such as hypertension or obesity. MMP-2 levels were affected by hypertension (p < 0.05), but unaffected by obesity status. Notably, hypertensive COVID-19 patients had higher MMP-2 levels compared to the non-hypertensive COVID-19 group, albeit still lower than Controls (p < 0.05). No association between MMP-2 and MMP-9 plasmatic levels and corticosteroid treatment or acute kidney injury was found in COVID-19 patients. The survival analysis showed that COVID-19 mortality was associated with increased MMP-2 and MMP-9 levels. Age, hypertension, BMI, and MMP-2 and MMP-9 were better predictors of mortality during hospitalization than SAPS3 and SOFA scores at hospital admission. In conclusion, a significant association between MMP-2 and MMP-9 levels and COVID-19 was found. Notably, MMP-2 and MMP-9 levels predicted the risk of in-hospital death suggesting possible pathophysiologic and prognostic roles.
Collapse
Affiliation(s)
- Carolina D Avila-Mesquita
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ariel E S Couto
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ligia C B Campos
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Tauana F Vasconcelos
- Division of Intensive Care, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jessyca Michelon-Barbosa
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carlos A C Corsi
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fabiola Mestriner
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno C Petroski-Moraes
- Division of Intensive Care, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria J Garbellini-Diab
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniel M S Couto
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria C Jordani
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Denise Ferro
- Division of Cardiac Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Lourenço Sbragia
- Division of Pediatrics Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Edwaldo E Joviliano
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Paulo R Evora
- Division of Cardiac Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rodrigo de Carvalho Santana
- Division of Infectious Diseases, Department of Clinical Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Katarzyna Polonis
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Mayra G Menegueti
- Ribeirão Preto Nurse Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mauricio S Ribeiro
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Auxiliadora-Martins
- Division of Intensive Care, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Christiane Becari
- Division of Vascular and Endovascular Surgery, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| |
Collapse
|
40
|
Dejani NN, Elshabrawy HA, Bezerra Filho CDSM, de Sousa DP. Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives. Biomolecules 2021; 11:biom11081254. [PMID: 34439920 PMCID: PMC8394099 DOI: 10.3390/biom11081254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/14/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023] Open
Abstract
In 2019, COVID-19 emerged as a severe respiratory disease that is caused by the novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). The disease has been associated with high mortality rate, especially in patients with comorbidities such as diabetes, cardiovascular and kidney diseases. This could be attributed to dysregulated immune responses and severe systemic inflammation in COVID-19 patients. The use of effective antiviral drugs against SARS-CoV-2 and modulation of the immune responses could be a potential therapeutic strategy for COVID-19. Studies have shown that natural phenolic compounds have several pharmacological properties, including anticoronavirus and immunomodulatory activities. Therefore, this review discusses the dual action of these natural products from the perspective of applicability at COVID-19.
Collapse
Affiliation(s)
- Naiara Naiana Dejani
- Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA;
| | - Carlos da Silva Maia Bezerra Filho
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
| | - Damião Pergentino de Sousa
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
- Correspondence: ; Tel.: +55-83-3216-7347
| |
Collapse
|
41
|
Non-acidic bifunctional benzothiazole-based thiazolidinones with antimicrobial and aldose reductase inhibitory activity as a promising therapeutic strategy for sepsis. Med Chem Res 2021; 30:1837-1848. [PMID: 34366640 PMCID: PMC8335715 DOI: 10.1007/s00044-021-02778-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/27/2021] [Indexed: 12/24/2022]
Abstract
Sepsis is a life-threatening disease that affects millions of people worldwide. Microbial infections that lead to sepsis syndrome are associated with an increased production of inflammatory molecules. Aldose reductase has recently emerged as a molecular target that is involved in various inflammatory diseases, including sepsis. Herein, a series of previously synthesized benzothiazole-based thiazolidinones that exhibited strong antibacterial and antifungal activities has been evaluated for inhibition efficacy against aldose reductase and selectivity toward aldehyde reductase under in vitro conditions. The most promising inhibitor 5 was characterized with IC50 value of 3.99 μM and a moderate selectivity. Molecular docking simulations revealed the binding mode of compounds at the active site of human aldose reductase. Moreover, owning to the absence of an acidic pharmacophore, good membrane permeation of the novel aldose reductase inhibitors was predicted. Excellent “drug-likeness” was assessed for most of the compounds by applying the criteria of Lipinski’s “rule of five”. ![]()
Collapse
|
42
|
Lim J, Lee YY, Choy YB, Park W, Park CG. Sepsis diagnosis and treatment using nanomaterials. Biomed Eng Lett 2021; 11:197-210. [PMID: 34277115 PMCID: PMC8274966 DOI: 10.1007/s13534-021-00200-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/10/2021] [Accepted: 07/04/2021] [Indexed: 12/13/2022] Open
Abstract
Sepsis is a life-threatening reaction that occurs when the body's severe response to an infection damages the host's own tissues. Sepsis has been globally recognized as a fatal disease. Rapid treatment of sepsis requires prompt identification, administering antibiotics, careful hemodynamic support, and treating the cause of the infection. Clinical outcomes of sepsis depend on early diagnosis and appropriate treatment. Unfortunately, current sepsis diagnosis and treatment, such as polymerase chain reaction-based assay, blood culture assay, and antibiotic therapy, are ineffective; consequently, sepsis-related mortality remains high and increases antimicrobial resistance. To overcome this challenge, nanotechnology, which involves engineering at a nanoscale, is used for diagnosing and treating sepsis. Preclinical models have shown protective effects and potential utility in managing septic shock. Furthermore, nanotechnology treatments based on diverse materials result in the effective treatment of sepsis, improving the survival rate. In this review, we present an overview of the recent research advancements in nanotechnology to diagnose and treat sepsis with a brief introduction to sepsis.
Collapse
Affiliation(s)
- Jaesung Lim
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Yun Young Lee
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Young Bin Choy
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Wooram Park
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, Bucheon, Gyeonggi 14662 Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Gyeonggi 16419 Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Gyeonggi 16419 Republic of Korea
| |
Collapse
|
43
|
Quantifying the Burden of Viral Sepsis During the Coronavirus Disease 2019 Pandemic and Beyond. Crit Care Med 2021; 49:2140-2143. [PMID: 34259668 PMCID: PMC8594516 DOI: 10.1097/ccm.0000000000005207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Osca-Verdegal R, Beltrán-García J, Pallardó FV, García-Giménez JL. Role of microRNAs As Biomarkers in Sepsis-Associated Encephalopathy. Mol Neurobiol 2021; 58:4682-4693. [PMID: 34160774 PMCID: PMC8220114 DOI: 10.1007/s12035-021-02445-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/06/2021] [Indexed: 12/29/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is a neurological complication of sepsis, characterized by brain dysfunction without any direct central nervous system infection. The diagnosis of SAE is currently a challenge. In fact, problems in making a diagnosis of SAE cause a great variability of incidence that can reach up to 70% of all septic patients. Even more, despite SAE is the most frequent type of encephalopathy occurring in critically ill patients, the molecular mechanisms that guide its progression have not been completely elucidated. On the other hand, miRNAs have proven to be excellent biomarkers for both diagnosis and prognosis, especially in brain pathologies because of their small size they can cross the blood–brain barrier easier than other biomolecules. The identification of new miRNAs as biomarkers may help to improve SAE diagnosis and prognosis and also to design new therapies for this clinical manifestation that produces diffuse cerebral dysfunction. This review is focused on SAE physiopathology and the need to have clear criteria for its diagnosis; thus, this work postulates some miRNA candidates to be used for SAE biomarkers because of their role in both, neurological damage and sepsis.
Collapse
Affiliation(s)
- Rebeca Osca-Verdegal
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina Y Odontología, Universitat de València, València, Spain
| | - Jesús Beltrán-García
- Departamento de Fisiología, Facultad de Medicina Y Odontología, Universitat de València, València, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - Federico V. Pallardó
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina Y Odontología, Universitat de València, València, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| | - José Luis García-Giménez
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Valencia, Spain
- Departamento de Fisiología, Facultad de Medicina Y Odontología, Universitat de València, València, Spain
- Instituto de Investigación Sanitaria INCLIVA, Valencia, Spain
| |
Collapse
|
45
|
Roach M, Munshi S. P.150 SARS-CoV-2 causing septic shock in pregnancy. Int J Obstet Anesth 2021. [PMCID: PMC8186984 DOI: 10.1016/j.ijoa.2021.103148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
46
|
Mazzoni A, Salvati L, Maggi L, Annunziato F, Cosmi L. Hallmarks of immune response in COVID-19: Exploring dysregulation and exhaustion. Semin Immunol 2021; 55:101508. [PMID: 34728121 PMCID: PMC8547971 DOI: 10.1016/j.smim.2021.101508] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023]
Abstract
One and half year following the occurrence of COVID-19 pandemic, significant efforts from laboratories all over the world generated a huge amount of data describing the prototypical features of immunity in the course of SARS-CoV-2 infection. In this Review, we rationalize and organize the main observations, trying to define a "core" signature of immunity in COVID-19. We identified six hallmarks describing the main alterations occurring in the early infection phase and in the course of the disease, which predispose to severe illness. The six hallmarks are dysregulated type I IFN activity, hyperinflammation, lymphopenia, lymphocyte impairment, dysregulated myeloid response, and heterogeneous adaptive immunity to SARS-CoV-2. Dysregulation and exhaustion came out as the trait d'union, connecting abnormalities affecting both innate and adaptive immunity, humoral and cellular responses.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
47
|
Goyal M, Murthy SI, Annum S. Retinal manifestations in patients following COVID-19 infection: A consecutive case series. Indian J Ophthalmol 2021; 69:1275-1282. [PMID: 33913876 PMCID: PMC8186578 DOI: 10.4103/ijo.ijo_403_21] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose To describe retinal manifestations seen in patients associated with COVID-19 infection at a multi-specialty tertiary care hospital in Southern India. Methods In this retrospective chart review, all consecutive cases presenting to the Retina-Uveitis service from May 2020 to January 2021 with retinal manifestations associated with COVID-19 infection or its sequelae or as a result of treatment given for COVID-19 were included. Results : Of the 7 patients, 3 were female, and 4 were male. Four patients had onset of symptoms during the active phase of COVID-19 infection. Four had bilateral and three had unilateral involvement. The manifestations ranged from mild to vision threatening. Vision threatening manifestations included infections: endogenous endophthalmitis, candida retinitis and tubercular choroidal abscess and bilateral pre-foveal hemorrhages. Milder manifestations included paracentral acute middle maculopathy, central serous chorio-retinopathy and voriconazole induced visual symptoms. Final visual acuity was 6/36 or better in the four severe cases and 6/9 or better in the mild cases. Conclusion This study highlights the retinal manifestations associated with COVID-19 infection and its sequelae. As these patients presented with an association with COVID-19 (either during or after recovery), ophthalmologists should be vigilant and screen for such entities in case of complaints of visual symptoms or in the presence of systemic sepsis. The outcomes can be good with prompt and aggressive management.
Collapse
Affiliation(s)
- Mallika Goyal
- Retina-Uveitis Service, Department of Ophthalmology, Apollo Health City, Hyderabad, Telangana, India
| | | | - Sridhar Annum
- Retina-Uveitis Service, Department of Ophthalmology, Apollo Health City, Hyderabad, Telangana, India
| |
Collapse
|
48
|
Barh D, Tiwari S, Andrade BS, Weener ME, Góes-Neto A, Azevedo V, Ghosh P, Blum K, Ganguly NK. A novel multi-omics-based highly accurate prediction of symptoms, comorbid conditions, and possible long-term complications of COVID-19. Mol Omics 2021; 17:317-337. [PMID: 33683246 DOI: 10.1039/d0mo00189a] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Comprehensive clinical pictures, comorbid conditions, and long-term complications of COVID-19 are still unknown. Recently, using a multi-omics-based strategy, we predicted potential drugs for COVID-19 with ∼70% accuracy. Herein, using a novel multi-omics-based bioinformatic approach and three ways of analysis, we identified the symptoms, comorbid conditions, and short-, mid-, and possible long-term complications of COVID-19 with >90% precision including 27 parent, 170 child, and 403 specific conditions. Among the specific conditions, 36 viral, 53 short-term, 62 short-mid-long-term, 194 mid-long-term, and 57 congenital conditions are identified. At a threshold "count of occurrence" of 4, we found that 83-100% (average 92.67%) of enriched conditions are associated with COVID-19. Except for dry cough and loss of taste, all the other COVID-19-associated mild and severe symptoms are enriched. CVDs, and pulmonary, metabolic, musculoskeletal, neuropsychiatric, kidney, liver, and immune system disorders are top comorbid conditions. Specific diseases like myocardial infarction, hypertension, COPD, lung injury, diabetes, cirrhosis, mood disorders, dementia, macular degeneration, chronic kidney disease, lupus, arthritis, etc. along with several other NCDs were found to be top candidates. Interestingly, many cancers and congenital disorders associated with COVID-19 severity are also identified. Arthritis, gliomas, diabetes, psychiatric disorders, and CVDs having a bidirectional relationship with COVID-19 are also identified as top conditions. Based on our accuracy (>90%), the long-term presence of SARS-CoV-2 RNA in human, and our "genetic remittance" assumption, we hypothesize that all the identified top-ranked conditions could be potential long-term consequences in COVID-19 survivors, warranting long-term observational studies.
Collapse
Affiliation(s)
- Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, WB, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Digala LP, Prasanna S, Rao P, Govindarajan R, Qureshi AI. Impact of COVID- 19 Infection Among Hospitalized Amyotrophic Lateral Sclerosis Patients. J Clin Neuromuscul Dis 2021; 22:180-181. [PMID: 33596004 PMCID: PMC7892201 DOI: 10.1097/cnd.0000000000000335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Lakshmi P. Digala
- Department of Neurology, University of Missouri Health Care, Columbia, MO
| | - Shivika Prasanna
- Departments of Health Management & Informatics, Center for Biomedical Informatics, Electrical Engineering & Computer Science, University of Missouri, Columbia, MO
| | - Praveen Rao
- Departments of Health Management & Informatics, Center for Biomedical Informatics, Electrical Engineering & Computer Science, University of Missouri, Columbia, MO
| | | | - Adnan I. Qureshi
- Department of Neurology, University of Missouri Health Care, Columbia, MO
| |
Collapse
|
50
|
Lo SY, Teah MK, Ho YZ, Yeap TB. Perioperative challenges in managing a patient with COVID-19 undergoing debridement for massive scalp myiasis. BMJ Case Rep 2021; 14:e241189. [PMID: 33547110 PMCID: PMC10577744 DOI: 10.1136/bcr-2020-241189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2021] [Indexed: 12/25/2022] Open
Abstract
A young man presented to our centre needing an urgent debridement of his postcraniotomy wound due to massive myiasis during the COVID-19 pandemic in October 2020. Prior to the surgery, his nasopharyngeal swab real-time PCR test result was unknown. One day later, it returned as SARS-CoV-2 positive. All healthcare workers who were involved in the patient management avoided cross infection as they wore appropriate personal protective equipment. This article depicts the importance of adequate preparations when handling potentially infectious patients and the perioperative issues associated with it.
Collapse
Affiliation(s)
- Shiu Yin Lo
- Department of Anaesthesia and Intensive Care Unit, Hospital Queen Elizabeth 2, Kota Kinabalu, Sabah, Malaysia
| | - Ming Kai Teah
- Department of Anaesthesia and Intensive Care Unit, Hospital Queen Elizabeth, Kota Kinabalu, Sabah, Malaysia
| | - Yan Zheng Ho
- Neurosurgical Department, Hospital Queen Elizabeth, Kota Kinabalu, Sabah, Malaysia
| | - Tat Boon Yeap
- Faculty of Medicine and Health Sciences, Medicine Based Disciplines Department, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| |
Collapse
|