1
|
Arumugam P, Saha K, Nighot P. Intestinal Epithelial Tight Junction Barrier Regulation by Novel Pathways. Inflamm Bowel Dis 2025; 31:259-271. [PMID: 39321109 DOI: 10.1093/ibd/izae232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Indexed: 09/27/2024]
Abstract
Intestinal epithelial tight junctions (TJs), a dynamically regulated barrier structure composed of occludin and claudin family of proteins, mediate the interaction between the host and the external environment by allowing selective paracellular permeability between the luminal and serosal compartments of the intestine. TJs are highly dynamic structures and can undergo constant architectural remodeling in response to various external stimuli. This is mediated by an array of intracellular signaling pathways that alters TJ protein expression and localization. Dysfunctional regulation of TJ components compromising the barrier homeostasis is an important pathogenic factor for pathological conditions including inflammatory bowel disease (IBD). Previous studies have elucidated the significance of TJ barrier integrity and key regulatory mechanisms through various in vitro and in vivo models. In recent years, considerable efforts have been made to understand the crosstalk between various signaling pathways that regulate formation and disassembly of TJs. This review provides a comprehensive view on the novel mechanisms that regulate the TJ barrier and permeability. We discuss the latest evidence on how ion transport, cytoskeleton and extracellular matrix proteins, signaling pathways, and cell survival mechanism of autophagy regulate intestinal TJ barrier function. We also provide a perspective on the context-specific outcomes of the TJ barrier modulation. The knowledge on the diverse TJ barrier regulatory mechanisms will provide further insights on the relevance of the TJ barrier defects and potential target molecules/pathways for IBD.
Collapse
Affiliation(s)
- Priya Arumugam
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Kushal Saha
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA, USA
| |
Collapse
|
2
|
An Y, He L, Xu X, Piao M, Wang B, Liu T, Cao H. Gut microbiota in post-acute COVID-19 syndrome: not the end of the story. Front Microbiol 2024; 15:1500890. [PMID: 39777148 PMCID: PMC11703812 DOI: 10.3389/fmicb.2024.1500890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has led to major global health concern. However, the focus on immediate effects was assumed as the tip of iceberg due to the symptoms following acute infection, which was defined as post-acute COVID-19 syndrome (PACS). Gut microbiota alterations even after disease resolution and the gastrointestinal symptoms are the key features of PACS. Gut microbiota and derived metabolites disorders may play a crucial role in inflammatory and immune response after SARS-CoV-2 infection through the gut-lung axis. Diet is one of the modifiable factors closely related to gut microbiota and COVID-19. In this review, we described the reciprocal crosstalk between gut and lung, highlighting the participation of diet and gut microbiota in and after COVID-19 by destroying the gut barrier, perturbing the metabolism and regulating the immune system. Therefore, bolstering beneficial species by dietary supplements, probiotics or prebiotics and fecal microbiota transplantation (FMT) may be a novel avenue for COVID-19 and PACS prevention. This review provides a better understanding of the association between gut microbiota and the long-term consequences of COVID-19, which indicates modulating gut dysbiosis may be a potentiality for addressing this multifaceted condition.
Collapse
Affiliation(s)
| | | | | | | | | | - Tianyu Liu
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
3
|
Pellicano C, Oliva A, Colalillo A, Gigante A, D'Aliesio E, Al Ismail D, Miele MC, Cianci R, Mastroianni CM, Rosato E. Serum markers of microbial translocation and intestinal damage in assessment of gastrointestinal tract involvement in systemic sclerosis. Clin Exp Med 2024; 24:225. [PMID: 39294494 PMCID: PMC11410972 DOI: 10.1007/s10238-024-01466-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/14/2024] [Indexed: 09/20/2024]
Abstract
Gastrointestinal (GI) tract involvement affects up to 90% of Systemic sclerosis (SSc) patients. The presence of GI symptoms is assessed by the University of California, Los Angeles, and Scleroderma Clinical Trials Consortium Gastrointestinal Scale (UCLA SCTC GIT 2.0). Microbial translocation (MT) is reported in SSc patients consequently to increased intestinal permeability due to intestinal damage (ID) and dysbiosis. Aim of this study was to assess circulating levels of LBP and EndoCab IgM (markers of MT), IL-6 (marker of inflammation), I-FABP and Zonulin (markers of ID) in a cohort of SSc patients and healthy controls (HC). Moreover, we aimed to correlate these parameters with severity of GI symptoms. UCLA SCTC GIT 2.0 questionnaire was administered to 60 consecutive SSc patients. Markers of MT, inflammation and ID were evaluated in SSc patients and HC. SSc patients had higher median value of markers of MT, inflammation and ID than HC. The logistic regression analysis showed LBP as the only variable associated with an UCLA total score "moderate-to-very severe" [OR 1.001 (CI 95%: 1.001-1.002), p < 0.001]. The logistic regression analysis showed LBP [OR 1.002 (CI 95%: 1.001-1.003), p < 0.01] and disease duration [OR 1.242 (CI 95%: 1.023-1.506), p < 0.05] as variables associated with UCLA distension/bloating "moderate-to-very severe". The logistic regression analysis showed LBP as the only variable associated with UCLA diarrhea "moderate-to-very severe" [OR 1.002 (CI 95%: 1.001-1.003), p < 0.01]. SSc patients with dysregulation gut mucosal integrity expressed by high levels of MT and ID biomarkers had more severe GI symptoms.
Collapse
Affiliation(s)
- Chiara Pellicano
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185, Rome, Italy
| | - Amalia Colalillo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Antonietta Gigante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Elisa D'Aliesio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185, Rome, Italy
| | - Dania Al Ismail
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185, Rome, Italy
| | - Maria Claudia Miele
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185, Rome, Italy
| | - Rosario Cianci
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185, Rome, Italy
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy.
| |
Collapse
|
4
|
Liu J, Huang Y, Liu N, Qiu H, Zhang X, Liu X, He M, Chen M, Huang S. The imbalance of pulmonary Th17/Treg cells in BALB/c suckling mice infected with respiratory syncytial virus-mediated intestinal immune damage and gut microbiota changes. Microbiol Spectr 2024; 12:e0328323. [PMID: 38727214 PMCID: PMC11237571 DOI: 10.1128/spectrum.03283-23] [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: 10/10/2023] [Accepted: 04/02/2024] [Indexed: 06/06/2024] Open
Abstract
The immune response induced by respiratory syncytial virus (RSV) infection is closely related to changes in the composition and function of gastrointestinal microorganisms. However, the specific mechanism remains unknown and the pulmonary-intestinal axis deserves further study. In this study, the mRNA levels of ROR-γt and Foxp3 in the lung and intestine increased first and then decreased. IL-17 and IL-22 reached the maximum on the third day after infection in the lung, and on the second day after infection in the small intestine and colon, respectively. RegⅢγ in intestinal tissue reached the maximum on the third day after RSV infection. Moreover, the genus enriched in the RSV group was Aggregatibacter, and Proteus was reduced. RSV infection not only causes Th17/Treg cell imbalance in the lungs of mice but also leads to the release of excessive IL-22 from the lungs through blood circulation which binds to IL-22 receptors on the intestinal surface, inducing RegⅢγ overexpression, impaired intestinal Th17/Treg development, and altered gut microbiota composition. Our research reveals a significant link between the pulmonary and intestinal axis after RSV infection. IMPORTANCE RSV is the most common pathogen causing acute lower respiratory tract infections in infants and young children, but the complex interactions between the immune system and gut microbiota induced by RSV infection still requires further research. In this study, it was suggested that RSV infection in 7-day-old BALB/c suckling mice caused lung inflammation and disruption of Th17/Treg cells development, and altered the composition of gut microbiota through IL-22 induced overexpression of RegⅢγ, leading to intestinal immune injury and disruption of gut microbiota. This research reveals that IL-22 may be the link between the lung and gut. This study may provide a new insight into the intestinal symptoms caused by RSV and other respiratory viruses and the connection between the lung and gut axis, as well as new therapeutic ideas for the treatment of RSV-infected children.
Collapse
Affiliation(s)
- Jiling Liu
- Department of Microbiology, The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- College of Life Science, Hebei University, Baoding, Hebei, China
| | - Yixuan Huang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Nian Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Huan Qiu
- School of Nursing, Anhui Medical University, Hefei, Anhui, China
| | - Xiaoyan Zhang
- Department of Microbiology, The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Xiaojie Liu
- Department of Microbiology, The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Maozhang He
- Department of Microbiology, The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Mingwei Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shenghai Huang
- Department of Microbiology, The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
5
|
Boncheva I, Poudrier J, Falcone EL. Role of the intestinal microbiota in host defense against respiratory viral infections. Curr Opin Virol 2024; 66:101410. [PMID: 38718575 DOI: 10.1016/j.coviro.2024.101410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 06/07/2024]
Abstract
Viral infections, including those affecting the respiratory tract, can alter the composition of the intestinal microbiota, which, in turn, can significantly influence both innate and adaptive immune responses, resulting in either enhanced pathogen clearance or exacerbation of the infection, possibly leading to inflammatory complications. A deeper understanding of the interplay between the intestinal microbiota and host immune responses in the context of respiratory viral infections (i.e. the gut-lung axis) is necessary to develop new treatments. This review highlights key mechanisms by which the intestinal microbiota, including its metabolites, can act locally or at distant organs to combat respiratory viruses. Therapeutics aimed at harnessing the microbiota to prevent and/or help treat respiratory viral infections represent a promising avenue for future investigation.
Collapse
Affiliation(s)
- Idia Boncheva
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Johanne Poudrier
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada; Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Emilia L Falcone
- Center for Immunity, Inflammation and Infectious Diseases, Montreal Clinical Research Institute/Institut de recherches cliniques de Montréal (IRCM), Montreal, QC, Canada; Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada; Department of Medicine, Université de Montréal, Montreal, QC, Canada; Department of Microbiology and Infectious Diseases, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.
| |
Collapse
|
6
|
Galeana-Cadena D, Ramirez-Martínez G, Alberto Choreño-Parra J, Silva-Herzog E, Margarita Hernández-Cárdenas C, Soberón X, Zúñiga J. Microbiome in the nasopharynx: Insights into the impact of COVID-19 severity. Heliyon 2024; 10:e31562. [PMID: 38826746 PMCID: PMC11141365 DOI: 10.1016/j.heliyon.2024.e31562] [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: 07/12/2023] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/04/2024] Open
Abstract
Background The respiratory tract harbors a variety of microbiota, whose composition and abundance depend on specific site factors, interaction with external factors, and disease. The aim of this study was to investigate the relationship between COVID-19 severity and the nasopharyngeal microbiome. Methods We conducted a prospective cohort study in Mexico City, collecting nasopharyngeal swabs from 30 COVID-19 patients and 14 healthy volunteers. Microbiome profiling was performed using 16S rRNA gene analysis. Taxonomic assignment, classification, diversity analysis, core microbiome analysis, and statistical analysis were conducted using R packages. Results The microbiome data analysis revealed taxonomic shifts within the nasopharyngeal microbiome in severe COVID-19. Particularly, we observed a significant reduction in the relative abundance of Lawsonella and Cutibacterium genera in critically ill COVID-19 patients (p < 0.001). In contrast, these patients exhibited a marked enrichment of Streptococcus, Actinomyces, Peptostreptococcus, Atopobium, Granulicatella, Mogibacterium, Veillonella, Prevotella_7, Rothia, Gemella, Alloprevotella, and Solobacterium genera (p < 0.01). Analysis of the core microbiome across all samples consistently identified the presence of Staphylococcus, Corynebacterium, and Streptococcus. Conclusions Our study suggests that the disruption of physicochemical conditions and barriers resulting from inflammatory processes and the intubation procedure in critically ill COVID-19 patients may facilitate the colonization and invasion of the nasopharynx by oral microorganisms.
Collapse
Affiliation(s)
- David Galeana-Cadena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Gustavo Ramirez-Martínez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - José Alberto Choreño-Parra
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Eugenia Silva-Herzog
- Unidad de Vinculación Científica Facultad de Medicina UNAM-INMEGEN, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Carmen Margarita Hernández-Cárdenas
- Unidad de Cuidados Intensivos y Dirección General, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Ciudad de México, Mexico
| | - Xavier Soberón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Joaquín Zúñiga
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| |
Collapse
|
7
|
Mouchati C, Durieux JC, Zisis SN, Tribout H, Scott S, Smith B, Labbato D, McComsey GA. Zinc Deficiency And sTNF-RII Are Associated With Worse COVID-19 Outcomes. J Nutr 2024; 154:1588-1595. [PMID: 38043624 PMCID: PMC11347801 DOI: 10.1016/j.tjnut.2023.11.026] [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: 09/28/2023] [Revised: 11/07/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Zinc (Zn) is known for its substantial involvement in the immune response as an antioxidant and anti-inflammatory agent. Zn plasma levels' clinical significance in coronavirus disease (COVID) diagnosis is not yet fully established. OBJECTIVE We assessed the association between Zn deficiency, gut integrity, inflammation, and COVID-19 outcomes. METHODS A prospective observational cohort in which plasma Zn, soluble tumor necrosis factor alpha receptor II (sTNF-RII) intestinal fatty-acid binding protein (IFABP; marker of intestinal integrity), and zonulin levels (intestinal permeability) were collected from participants during the acute phase of a confirmed COVID-19 diagnosis. Zn was modeled as continuous and binary, categorized as Zn deficiency (Zn < 75 μg/dL) and Zn sufficiency (Zn ≥ 75 μg/dL). COVID-19 outcomes were classified according to the World Health Organization clinical progression scale. We used cumulative probit regression to assess if suboptimal Zn levels, gut, and inflammatory markers increase the likelihood of worse COVID-19 outcomes. RESULTS Zn deficiency was independently associated with 63% higher predicted odds of worse COVID outcomes. Increases in sTNF-RII {unadjusted odds ratio (uOR): 3.43 [95% confidence interval (CI): 2.02, 5.82]} and zonulin [uOR: 1.83 (95% CI: 1.21, 2.76)] levels were associated with greater odds of worse COVID outcomes. IFABP was not associated with worse COVID outcomes [uOR: 1.12 (95% CI: 0.82, 1.53)] or acute Zn deficiency [uOR: 1.35 (95% CI: 0.79, 2.35)]. The adjusted predicted odds of worse COVID outcomes are 3-fold higher (P = 0.04) for every one-unit decrease in Zn and is more than 2 times greater odds of COVID severity (P = 0.01) for every 1-unit increase in sTNF-RII. CONCLUSION Zn deficiency and inflammation were independently associated with greater odds of worse COVID outcomes.
Collapse
Affiliation(s)
- Christian Mouchati
- School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jared C Durieux
- Center for Clinical Research, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Sokratis N Zisis
- School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Heather Tribout
- Center for Clinical Research, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Sarah Scott
- Center for Clinical Research, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Beth Smith
- Center for Clinical Research, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Danielle Labbato
- Center for Clinical Research, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Grace A McComsey
- School of Medicine, Case Western Reserve University, Cleveland, OH, United States; Center for Clinical Research, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.
| |
Collapse
|
8
|
Bohmwald K, Diethelm-Varela B, Rodríguez-Guilarte L, Rivera T, Riedel CA, González PA, Kalergis AM. Pathophysiological, immunological, and inflammatory features of long COVID. Front Immunol 2024; 15:1341600. [PMID: 38482000 PMCID: PMC10932978 DOI: 10.3389/fimmu.2024.1341600] [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: 11/20/2023] [Accepted: 02/09/2024] [Indexed: 04/12/2024] Open
Abstract
The COVID-19 pandemic continues to cause severe global disruption, resulting in significant excess mortality, overwhelming healthcare systems, and imposing substantial social and economic burdens on nations. While most of the attention and therapeutic efforts have concentrated on the acute phase of the disease, a notable proportion of survivors experience persistent symptoms post-infection clearance. This diverse set of symptoms, loosely categorized as long COVID, presents a potential additional public health crisis. It is estimated that 1 in 5 COVID-19 survivors exhibit clinical manifestations consistent with long COVID. Despite this prevalence, the mechanisms and pathophysiology of long COVID remain poorly understood. Alarmingly, evidence suggests that a significant proportion of cases within this clinical condition develop debilitating or disabling symptoms. Hence, urgent priority should be given to further studies on this condition to equip global public health systems for its management. This review provides an overview of available information on this emerging clinical condition, focusing on the affected individuals' epidemiology, pathophysiological mechanisms, and immunological and inflammatory profiles.
Collapse
Affiliation(s)
- Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Benjamín Diethelm-Varela
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Linmar Rodríguez-Guilarte
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Thomas Rivera
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
9
|
Ghoshal UC, Ghoshal U. Gastrointestinal involvement in post-acute Coronavirus disease (COVID)-19 syndrome. Curr Opin Infect Dis 2023; 36:366-370. [PMID: 37606895 DOI: 10.1097/qco.0000000000000959] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
PURPOSE OF REVIEW Ten percentage of patients with coronavirus disease (COVID)-19 report having gastrointestinal (GI) symptoms as severe acute respiratory syndrome coronavirus-2 (SARS-CoV2) not only infects the pulmonary but also the GI tract. GI infections including that due to viral infection is known to cause postinfection disorders of gut-brain interaction (DGBI); hence, we wish to review the long-term GI consequences following COVID-19, particularly post-COVID-19 DGBI. RECENT FINDINGS At least 12 cohort studies, four of which also included controls documented the occurrence of post-COVID-19 DGBI, particularly IBS following COVID-19. The risk factors for post-COVID-19 DGBI included female gender, symptomatic COVID-19, particularly GI symptoms, the severity of COVID-19, the occurrence of anosmia and ageusia, use of antibiotics and hospitalization during the acute illness, persistent GI symptoms beyond 1 month after recovery, presence of mental health factors, The putative mechanisms for post-COVID-19 DGBI include altered gut motility, visceral hypersensitivity, gut microbiota dysbiosis, GI inflammation, and immune activation, changes in intestinal permeability, and alterations in the enteroendocrine system and serotonin metabolism. SUMMARY Long-term sequelae of SARS-CoV2 infection may persist even after recovery from COVID-19. Patients with COVID-19 are more likely to develop post-COVID-19 IBS than healthy controls. Post-COVID-19 IBS may pose a substantial healthcare burden to society.
Collapse
Affiliation(s)
- Uday C Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow
| | - Ujjala Ghoshal
- Department of Microbiology, All India Institute of Medical Science, Kalyani, West Bengal, India
| |
Collapse
|
10
|
Sfera A, Rahman L, Zapata-Martín Del Campo CM, Kozlakidis Z. Long COVID as a Tauopathy: Of "Brain Fog" and "Fusogen Storms". Int J Mol Sci 2023; 24:12648. [PMID: 37628830 PMCID: PMC10454863 DOI: 10.3390/ijms241612648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Long COVID, also called post-acute sequelae of SARS-CoV-2, is characterized by a multitude of lingering symptoms, including impaired cognition, that can last for many months. This symptom, often called "brain fog", affects the life quality of numerous individuals, increasing medical complications as well as healthcare expenditures. The etiopathogenesis of SARS-CoV-2-induced cognitive deficit is unclear, but the most likely cause is chronic inflammation maintained by a viral remnant thriving in select body reservoirs. These viral sanctuaries are likely comprised of fused, senescent cells, including microglia and astrocytes, that the pathogen can convert into neurotoxic phenotypes. Moreover, as the enteric nervous system contains neurons and glia, the virus likely lingers in the gastrointestinal tract as well, accounting for the intestinal symptoms of long COVID. Fusogens are proteins that can overcome the repulsive forces between cell membranes, allowing the virus to coalesce with host cells and enter the cytoplasm. In the intracellular compartment, the pathogen hijacks the actin cytoskeleton, fusing host cells with each other and engendering pathological syncytia. Cell-cell fusion enables the virus to infect the healthy neighboring cells. We surmise that syncytia formation drives cognitive impairment by facilitating the "seeding" of hyperphosphorylated Tau, documented in COVID-19. In our previous work, we hypothesized that the SARS-CoV-2 virus induces premature endothelial senescence, increasing the permeability of the intestinal and blood-brain barrier. This enables the migration of gastrointestinal tract microbes and/or their components into the host circulation, eventually reaching the brain where they may induce cognitive dysfunction. For example, translocated lipopolysaccharides or microbial DNA can induce Tau hyperphosphorylation, likely accounting for memory problems. In this perspective article, we examine the pathogenetic mechanisms and potential biomarkers of long COVID, including microbial cell-free DNA, interleukin 22, and phosphorylated Tau, as well as the beneficial effect of transcutaneous vagal nerve stimulation.
Collapse
Affiliation(s)
- Adonis Sfera
- Paton State Hospital, 3102 Highland Ave, Patton, CA 92369, USA
- School of Behavioral Health, Loma Linda University, 11139 Anderson St., Loma Linda, CA 92350, USA
- Department of Psychiatry, University of California, Riverside 900 University Ave, Riverside, CA 92521, USA
| | - Leah Rahman
- Department of Neuroscience, University of Oregon, 222 Huestis Hall, Eugene, OR 97401, USA
| | | | - Zisis Kozlakidis
- International Agency for Research on Cancer, World Health Organization, 69000 Lyon, France
| |
Collapse
|
11
|
Cogliati Dezza F, Covino S, Petrucci F, Sacco F, Viscido A, Gavaruzzi F, Ceccarelli G, Raponi G, Borrazzo C, Alessandri F, Mastroianni CM, Venditti M, Oliva A. Risk factors for carbapenem-resistant Acinetobacter baumannii (CRAB) bloodstream infections and related mortality in critically ill patients with CRAB colonization. JAC Antimicrob Resist 2023; 5:dlad096. [PMID: 37577156 PMCID: PMC10412853 DOI: 10.1093/jacamr/dlad096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023] Open
Abstract
Background Among MDR bacteria, carbapenem-resistant Acinetobacter baumannii (CRAB) is a major concern due to the limited therapeutic options. During the COVID-19 pandemic, a worrying increase in the spread of CRAB infections was reported. Objectives The study assessed the risk factors for CRAB bloodstream infection (BSI) in patients admitted to the ICU with CRAB colonization, and the related mortality risk factors. Methods We conducted a single-centre, observational, prospective study; all consecutive patients with CRAB colonization admitted to the ICU of a tertiary hospital in Rome from January 2021 to September 2022 were included in the study. Univariate and multivariate analyses were performed to investigate BSI and mortality risk factors. Results Overall, 129 patients were included in the study; 57 (44%) out of these developed BSI. In our study population, at the multivariable analysis the Charlson comorbidity index (CCI) (P = 0.026), COVID-19 (P < 0.001), multisite colonization (P = 0.016) and the need for mechanical ventilation (P = 0.024) were risk factors independently associated with BSI development. Furthermore, age (P = 0.026), CCI (P < 0.001), septic shock (P = 0.001) and Pitt score (P < 0.001) were independently associated with mortality in the BSI patients. Instead, early appropriate therapy (P = 0.002) and clinical improvement within 72 h (P = 0.011) were shown to be protective factors. Conclusions In critically ill patients colonized by CRAB, higher CCI, multisite colonization and the need for mechanical ventilation were identified as risk factors for BSI onset. These predictors could be useful to identify patients at highest risk of BSI.
Collapse
Affiliation(s)
| | - Sara Covino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Flavia Petrucci
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Federica Sacco
- Microbiology and Virology Laboratory, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Agnese Viscido
- Microbiology and Virology Laboratory, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Gavaruzzi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Gianmarco Raponi
- Microbiology and Virology Laboratory, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cristian Borrazzo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Francesco Alessandri
- Department of General and Specialistic Surgery, Sapienza University of Rome, Rome, Italy
| | | | - Mario Venditti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
12
|
Frattari A, Polilli E, Rapacchiale G, Coladonato S, Ianniruberto S, Mazzotta E, Patarchi A, Battilana M, Ciulli R, Moretta A, Visocchi L, Savini V, Spacone A, Zocaro R, Carinci F, Parruti G. Predictors of bacteremia and death, including immune status, in a large single-center cohort of unvaccinated ICU patients with COVID-19 pneumonia. Eur J Med Res 2023; 28:219. [PMID: 37400898 DOI: 10.1186/s40001-023-01166-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 06/11/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND We investigated the possible role of the immune profile at ICU admission, among other well characterized clinical and laboratory predictors of unfavorable outcome in COVID-19 patients assisted in ICU. METHODS Retrospective analysis of clinical and laboratory data collected for all consecutive patients admitted to the ICUs of the General Hospital of Pescara (Abruzzo, Italy), between 1st March 2020 and 30th April 2021, with a confirmed diagnosis of COVID-19 respiratory failure. Logistic regressions were used to identify independent predictors of bacteremia and mortality. RESULTS Out of 431 patients included in the study, bacteremia was present in N = 191 (44.3%) and death occurred in N = 210 (48.7%). After multivariate analysis, increased risk of bacteremia was found for viral reactivation (OR = 3.28; 95% CI:1.83-6.08), pronation (3.36; 2.12-5.37) and orotracheal intubation (2.51; 1.58-4.02). Increased mortality was found for bacteremia (2.05; 1.31-3.22), viral reactivation (2.29; 1.29-4.19) and lymphocytes < 0.6 × 103c/µL (2.32; 1.49-3.64). CONCLUSIONS We found that viral reactivation, mostly due to Herpesviridae, was associated with increased risk of both bacteremia and mortality. In addition, pronation and intubation are strong predictors of bacteremia, which in turn together with severe lymphocytopenia due to SARS-CoV2 was associated with increased mortality. Most episodes of bacteremia, even due to Acinetobacter spp, were not predicted by microbiological evidence of colonization.
Collapse
Affiliation(s)
| | - Ennio Polilli
- Clinical Pathology Unit, Pescara General Hospital, Pescara, Italy
| | | | | | | | - Elena Mazzotta
- Infectious Diseases Unit, Pescara General Hospital, Pescara, Italy
| | | | | | - Raffaella Ciulli
- Unit of Intensive Care, Pescara General Hospital, Pescara, Italy
| | - Angelo Moretta
- Unit of Intensive Care, Pescara General Hospital, Pescara, Italy
| | - Lina Visocchi
- Unit of Intensive Care, Pescara General Hospital, Pescara, Italy
| | - Vincenzo Savini
- Microbiology and Virology Unit, Pescara General Hospital, Pescara, Italy
| | | | - Rosamaria Zocaro
- Unit of Intensive Care, Pescara General Hospital, Pescara, Italy
| | - Fabrizio Carinci
- Department of Statistical Sciences, Università Di Bologna, Bologna, Italy
| | - Giustino Parruti
- Infectious Diseases Unit, Pescara General Hospital, Pescara, Italy.
| |
Collapse
|
13
|
Singh R, Malik P, Kumar M, Kumar R, Alam MS, Mukherjee TK. Secondary fungal infections in SARS-CoV-2 patients: pathological whereabouts, cautionary measures, and steadfast treatments. Pharmacol Rep 2023:10.1007/s43440-023-00506-z. [PMID: 37354313 DOI: 10.1007/s43440-023-00506-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
The earliest documented COVID-19 case caused by the SARS-CoV-2 coronavirus occurred in Wuhan, China, in December 2019. Since then, several SARS-CoV-2 mutants have rapidly disseminated as exemplified by the community spread of the recent omicron variant. The disease already attained a pandemic status with ever-dwindling mortality even after two and half years of identification and considerable vaccination. Aspergillosis, candidiasis, cryptococcosis and mucormycosis are the prominent fungal infections experienced by the majority of SARS-CoV-2 high-risk patients. In its entirety, COVID-19's nexus with these fungal infections may worsen the intricacies in the already beleaguered high-risk patients, making this a topic of substantial clinical concern. Thus, thorough knowledge of the subject is necessary. This article focuses on the concomitant fungal infection(s) in COVID-19 patients, taking into account their underlying causes, the screening methods, manifested drug resistance, and long-term effects. The information and knowledge shared herein could be crucial for the management of critically ill, aged, and immunocompromised SARS-CoV-2 patients who have had secondary fungal infections (SFIs).
Collapse
Affiliation(s)
- Raj Singh
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| | - Mukesh Kumar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Raman Kumar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Md Shamshir Alam
- Department of Pharmacy Practice, College of Pharmacy, National University of Science and Technology, PO Box 620, 130, Bosher-Muscat, Sultanate of Oman
| | - Tapan Kumar Mukherjee
- Amity Institute of Biotechnology, Amity University, Sector-125, Noida, UP, India.
- Department of Biotechnology, Amity University, Major Arterial Road, Action Area II, Rajarhat, New Town, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
14
|
Ciacci P, Paraninfi A, Orlando F, Rella S, Maggio E, Oliva A, Cangemi R, Carnevale R, Bartimoccia S, Cammisotto V, D'Amico A, Magna A, Nocella C, Mastroianni CM, Pignatelli P, Violi F, Loffredo L. Endothelial dysfunction, oxidative stress and low-grade endotoxemia in COVID-19 patients hospitalised in medical wards. Microvasc Res 2023:104557. [PMID: 37268038 DOI: 10.1016/j.mvr.2023.104557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Endothelial dysfunction, assessed by flow-mediated dilation (FMD), is related to poor prognosis in patients with COVID-19 pneumonia (CP). In this study, we explored the interplay among FMD, NADPH oxidase type 2 (NOX-2) and lipopolysaccharides (LPS) in hospitalised patients with CP, community acquired pneumonia (CAP) and controls (CT). METHODS We enrolled 20 consecutive patients with CP, 20 hospitalised patients with CAP and 20 CT matched for sex, age, and main cardiovascular risk factors. In all subjects we performed FMD and collected blood samples to analyse markers of oxidative stress (soluble Nox2-derived peptide (sNOX2-dp), hydrogen peroxide breakdown activity (HBA), nitric oxide (NO), hydrogen peroxide (H2O2)), inflammation (TNF-α and IL-6), LPS and zonulin levels. RESULTS Compared with controls, CP had significant higher values of LPS, sNOX-2-dp, H2O2,TNF-α, IL-6 and zonulin; conversely FMD, HBA and NO bioavailability were significantly lower in CP. Compared to CAP patients, CP had significantly higher levels of sNOX2-dp, H2O2, TNF-α, IL-6, LPS, zonulin and lower HBA. Simple linear regression analysis showed that FMD inversely correlated with sNOX2-dp, H2O2, TNF-α, IL-6, LPS and zonulin; conversely FMD was directly correlated with NO bioavailability and HBA. Multiple linear regression analysis highlighted LPS as the only predictor of FMD. CONCLUSION This study shows that patients with COVID-19 have low-grade endotoxemia that could activate NOX-2, generating increased oxidative stress and endothelial dysfunction.
Collapse
Affiliation(s)
- Paolo Ciacci
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Aurora Paraninfi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Federica Orlando
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Silvia Rella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Enrico Maggio
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Roberto Cangemi
- Department of Translational and Precision Medicine, Sapienza-University of Rome, Viale del Policlinico 155, 00162 Rome, Italy
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; Mediterranea Cardiocentro, Naples, Italy
| | - Simona Bartimoccia
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Vittoria Cammisotto
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Alessandra D'Amico
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", 00135 Rome, Italy
| | - Arianna Magna
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Cristina Nocella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy
| | - Francesco Violi
- Mediterranea Cardiocentro, Naples, Italy; Sapienza University of Rome, Rome, Italy
| | - Lorenzo Loffredo
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155, 00161 Rome, Italy.
| |
Collapse
|
15
|
Tsounis EP, Triantos C, Konstantakis C, Marangos M, Assimakopoulos SF. Intestinal barrier dysfunction as a key driver of severe COVID-19. World J Virol 2023; 12:68-90. [PMID: 37033148 PMCID: PMC10075050 DOI: 10.5501/wjv.v12.i2.68] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/08/2022] [Accepted: 01/16/2023] [Indexed: 03/21/2023] Open
Abstract
The intestinal lumen harbors a diverse consortium of microorganisms that participate in reciprocal crosstalk with intestinal immune cells and with epithelial and endothelial cells, forming a multi-layered barrier that enables the efficient absorption of nutrients without an excessive influx of pathogens. Despite being a lung-centered disease, severe coronavirus disease 2019 (COVID-19) affects multiple systems, including the gastrointestinal tract and the pertinent gut barrier function. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can inflict either direct cytopathic injury to intestinal epithelial and endothelial cells or indirect immune-mediated damage. Alternatively, SARS-CoV-2 undermines the structural integrity of the barrier by modifying the expression of tight junction proteins. In addition, SARS-CoV-2 induces profound alterations to the intestinal microflora at phylogenetic and metabolomic levels (dysbiosis) that are accompanied by disruption of local immune responses. The ensuing dysregulation of the gut-lung axis impairs the ability of the respiratory immune system to elicit robust and timely responses to restrict viral infection. The intestinal vasculature is vulnerable to SARS-CoV-2-induced endothelial injury, which simultaneously triggers the activation of the innate immune and coagulation systems, a condition referred to as “immunothrombosis” that drives severe thrombotic complications. Finally, increased intestinal permeability allows an aberrant dissemination of bacteria, fungi, and endotoxin into the systemic circulation and contributes, to a certain degree, to the over-exuberant immune responses and hyper-inflammation that dictate the severe form of COVID-19. In this review, we aim to elucidate SARS-CoV-2-mediated effects on gut barrier homeostasis and their implications on the progression of the disease.
Collapse
Affiliation(s)
- Efthymios P Tsounis
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Christos Konstantakis
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Markos Marangos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Patras 26504, Greece
| | - Stelios F Assimakopoulos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Patras 26504, Greece
| |
Collapse
|
16
|
Lugonja SI, Pantic IL, Milovanovic TM, Grbovic VM, Djokovic BM, Todorovic ŽD, Simovic SM, Medovic RH, Zdravkovic ND, Zdravkovic ND. Atherosclerotic Cardiovascular Disease in Inflammatory Bowel Disease: The Role of Chronic Inflammation and Platelet Aggregation. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:554. [PMID: 36984554 PMCID: PMC10059701 DOI: 10.3390/medicina59030554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
Background and Objectives: Atherosclerosis is one of inflammatory bowel disease's most significant cardiovascular manifestations. This research aimed to examine the relationship between biochemical, haemostatic, and immune parameters of atherosclerosis and ulcerative colitis patients and its relationship to platelet aggregation. Materials and Methods: A clinical, observational cross-sectional study was performed, during which the tested parameters were compared in the experimental and control groups. The patients were divided into four groups. The first group had 25 patients who had ulcerative colitis and atherosclerosis. The second group included 39 patients with ulcerative colitis without atherosclerosis. The third group comprised 31 patients suffering from atherosclerosis without ulcerative colitis, and the fourth group comprised 25 healthy subjects. Results: In our study, we registered statistically higher levels of inflammatory markers like SE, CRP, Le, fecal calprotectin, TNF-α, and IL-6, as well as the higher value of thrombocytes and thrombocyte aggregation in the group of patients with ulcerative colitis compared to the control group. Lower levels of total cholesterol and LDL were also recorded in patients with ulcerative colitis and atherosclerosis and ulcerative colitis without atherosclerosis compared to healthy control. Triglyceride and remnant cholesterol were higher in patients with ulcerative colitis and atherosclerosis when compared to patients with ulcerative colitis and healthy control but lower than in patients with atherosclerosis only. Conclusions: Several inflammatory markers and platelet aggregation could be good discrimination markers for subjects with ulcerative colitis with the highest risk of atherosclerosis.
Collapse
Affiliation(s)
- Sofija I. Lugonja
- Division of Gastroenterology, Department of Internal Medicine, General Hospital “Djordje Joanovic”, 5 Dr. Vase Savica Street, 23000 Zrenjanin, Serbia
| | - Ivana L. Pantic
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, 2 Dr. Koste Todorovica Street, 11000 Belgrade, Serbia
| | - Tamara M. Milovanovic
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, 2 Dr. Koste Todorovica Street, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 8 Dr. Subotica Starijeg Street, 11000 Belgrade, Serbia
| | - Vesna M. Grbovic
- Department of Physical Medicine and Rehabilitation, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia
- Center for Physical Medicine and Rehabilitation, University Clinical Center Kragujevac, 30 Zmaj Jovina Street, 34000 Kragujevac, Serbia
| | - Bojana M. Djokovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia
- Clinic for Cardiology, University Clinical Center Kragujevac, 30 Zmaj Jovina Street, 34000 Kragujevac, Serbia
| | - Željko D. Todorovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia
- Clinic for Hematology, University Clinical Center Kragujevac, 30 Zmaj Jovina Street, 34000 Kragujevac, Serbia
| | - Stefan M. Simovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia
- Clinic for Cardiology, University Clinical Center Kragujevac, 30 Zmaj Jovina Street, 34000 Kragujevac, Serbia
| | - Raša H. Medovic
- Department of Pediatrics, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia
- Pediatric Clinic, University Clinical Center Kragujevac, 30 Zmaj Jovina Street, 34000 Kragujevac, Serbia
| | - Nebojsa D. Zdravkovic
- Department of Medical Statistics and Informatics, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia
| | - Natasa D. Zdravkovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, 34000 Kragujevac, Serbia
- Clinic for Gastroenterology and Hepatology, University Clinical Center Kragujevac, 30 Zmaj Jovina Street, 34000 Kragujevac, Serbia
| |
Collapse
|
17
|
Islam MS, Wang Z, Abdel-Mohsen M, Chen X, Montaner LJ. Tissue injury and leukocyte changes in post-acute sequelae of SARS-CoV-2: review of 2833 post-acute patient outcomes per immune dysregulation and microbial translocation in long COVID. J Leukoc Biol 2023; 113:236-254. [PMID: 36807444 DOI: 10.1093/jleuko/qiac001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Indexed: 01/18/2023] Open
Abstract
A significant number of persons with coronavirus disease 2019 (COVID-19) experience persistent, recurrent, or new symptoms several months after the acute stage of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. This phenomenon, termed post-acute sequelae of SARS-CoV-2 (PASC) or long COVID, is associated with high viral titers during acute infection, a persistently hyperactivated immune system, tissue injury by NETosis-induced micro-thrombofibrosis (NETinjury), microbial translocation, complement deposition, fibrotic macrophages, the presence of autoantibodies, and lymphopenic immune environments. Here, we review the current literature on the immunological imbalances that occur during PASC. Specifically, we focus on data supporting common immunopathogenesis and tissue injury mechanisms shared across this highly heterogenous disorder, including NETosis, coagulopathy, and fibrosis. Mechanisms include changes in leukocyte subsets/functions, fibroblast activation, cytokine imbalances, lower cortisol, autoantibodies, co-pathogen reactivation, and residual immune activation driven by persistent viral antigens and/or microbial translocation. Taken together, we develop the premise that SARS-CoV-2 infection results in PASC as a consequence of acute and/or persistent single or multiple organ injury mediated by PASC determinants to include the degree of host responses (inflammation, NETinjury), residual viral antigen (persistent antigen), and exogenous factors (microbial translocation). Determinants of PASC may be amplified by comorbidities, age, and sex.
Collapse
Affiliation(s)
- Md Sahidul Islam
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China
| | - Zhaoxiong Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China
| | - Mohamed Abdel-Mohsen
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, United States
| | - Xin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, Avenida da Universidade, Taipa 999078, University of Macau, Macau S.A.R., China.,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Research Building N22, University of Macau, Avenida da Universidade, Taipa 999078, Macau S.A.R., China
| | - Luis J Montaner
- Vaccine and Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, United States
| |
Collapse
|
18
|
High Incidence of Candidemia in Critically Ill COVID-19 Patients Supported by Veno-Venous Extracorporeal Membrane Oxygenation: A Retrospective Study. J Fungi (Basel) 2023; 9:jof9010119. [PMID: 36675940 PMCID: PMC9861971 DOI: 10.3390/jof9010119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The incidence of candidemia in severe COVID-19 patients (0.8-14%) is two- to ten-fold higher than in non-COVID-19 patients. METHODS This retrospective analysis aimed to analyse the incidence of bloodstream infections (BSI) due to Candida in a cohort of COVID-19 patients supported with ECMO. RESULTS Among 138 intubated and ventilated patients hospitalized for ≥10 days in the intensive care unit of a teaching hospital, 45 (32.6%) patients received ECMO support, while 93 patients (67.4%) did not meet ECMO criteria and were considered the control group. In the ECMO group, 16 episodes of candidaemia were observed, while only 13 in patients of the control group (36.0% vs. 14.0%, p-value 0.004). It was confirmed at the survival analysis (SHR: 2.86, 95% CI: 1.39-5.88) and at the multivariable analyses (aSHR: 3.91, 95% CI: 1.73-8.86). A higher candida score seemed to increase the hazard for candidemia occurrence (aSHR: 3.04, 95% CI: 2.09-4.42), while vasopressor therapy was negatively associated with the outcome (aSHR: 0.15, 95% CI: 0.05-0.43). CONCLUSIONS This study confirms that the incidence of candidemia was significantly higher in critically ill COVID-19 patients supported with VV-ECMO than in critically ill COVID patients who did not meet criteria for VV-ECMO.
Collapse
|
19
|
Duck plague virus infection alter the microbiota composition and intestinal functional activity in Muscovy ducks. Poult Sci 2022; 102:102365. [PMID: 36610104 PMCID: PMC9829712 DOI: 10.1016/j.psj.2022.102365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/24/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022] Open
Abstract
Intestinal damage from the duck plague virus (DPV) infection affects intestinal inflammation factors expression and barrier dysfunction. Here we report findings from the pathogenicity of the intestinal tract, intestinal morphological, intestinal permeability, inflammatory cytokines, and tight junction gene expression in 72 two-wk-old Muscovy ducks exposed to DPV. The characterization of intestinal metabolites and their classification were examined using 16-sequencing technology. The primary outcomes of the study evaluated the correlation between intestinal microbiota characteristics and the degree of infected tissue. The secondary outcomes were to determine whether the biosignatures that defined the microbiota were positively or negatively correlated with viral infection. The tissue was infected accompanied a mild damage of liver and spleen, and severe intestinal bleeding. Two inoculation routes were constructed with susceptible animals to assess the pathogenicity of the DPV in order to enrich the status of infection in Muscovy ducks. High levels of virus titer from Muscovy ducks were found being in the intestine. The expression of INF-α and IL-β with viral infection increased at 4, and 6 dpi, respectively, after detecting of the inflammatory factor and barrier function genes. At 4 and 6 dpi, barrier function gene of ZO-1 and Occludin reduced. The severity of viral infection was significantly correlated with the characteristics of the intestinal microbiota. Ducks infected with the DPV had an increase in the phylum Firmicutes, a decrease in the phylum Actinobacteriota, and differential enrichment with the genus Bacteroides, Tyzzerella, Enterococcus, and Escherchia-Shigella, while the genus Rothia, Streptococcus, and Ralstonia were differentially enriched in the control group. The findings from the current study demonstrated that DPV infection leads to an imbalance of the intestinal microbiota and disruption of the microbial homeostasis in the intestinal tissue in ducks, which might be one of the mechanisms whereby DPV infection might be established in Muscovy ducks. Na+/K+-ATPase and Ca2+/Mg2+-ATPase activity monitoring also showed that viral infection reduced these activities. These findings imply that changes in intestinal microbiota, intestinal barrier gene expression, and inflammatory factor are related to viral infection. When taken as a whole, this work provides fresh perspectives on the characteristics of intestinal microbiota and the infection damage caused by the DPV.
Collapse
|
20
|
Floridia M, Giuliano M, Monaco M, Palmieri L, Lo Noce C, Palamara AT, Pantosti A, Brusaferro S, Onder G, Palmieri L, Agazio E, Barbariol P, Bella A, Benelli E, Bertinato L, Bocci M, Boros S, Bressi M, Calcagnini G, Canevelli M, Censi F, Ciervo A, Colaizzo E, Da Cas R, Del Manso M, Di Benedetto C, Donfrancesco C, Fabiani M, Facchiano F, Floridia M, Galati F, Giuliano M, Grisetti T, Guastadisegni C, Lega I, Lo Noce C, Maiozzi P, Manno V, Martini M, Massari M, Urdiales AM, Mattei E, Meduri C, Meli P, Menniti Ippolito F, Minelli G, Onder G, Petrone D, Pezzotti P, Pricci F, Punzo O, Quarata F, Raparelli V, Riccardo F, Rocchetto S, Sacco C, Salerno P, Sarti G, Serra D, Spila Alegiani S, Spuri M, Tallon M, Tamburo De Bella M, Tiple D, Toccaceli Blasi M, Trentin F, Unim B, Vaianella L, Vanacore N, Vescio MF, Villani ER, Weimer LE, Brusaferro S. Microbiologically confirmed infections and antibiotic-resistance in a national surveillance study of hospitalised patients who died with COVID-19, Italy 2020–2021. Antimicrob Resist Infect Control 2022; 11:74. [PMID: 35598032 PMCID: PMC9123740 DOI: 10.1186/s13756-022-01113-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 05/07/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Patients hospitalised for COVID-19 may present with or acquire bacterial or fungal infections that can affect the course of the disease. The aim of this study was to describe the microbiological characteristics of laboratory-confirmed infections in hospitalised patients with severe COVID-19.
Methods
We reviewed the hospital charts of a sample of patients deceased with COVID-19 from the Italian National COVID-19 Surveillance, who had laboratory-confirmed bacterial or fungal bloodstream infections (BSI) or lower respiratory tract infections (LRTI), evaluating the pathogens responsible for the infections and their antimicrobial susceptibility.
Results
Among 157 patients with infections hospitalised from February 2020 to April 2021, 28 (17.8%) had co-infections (≤ 48 h from admission) and 138 (87.9%) had secondary infections (> 48 h). Most infections were bacterial; LRTI were more frequent than BSI. The most common co-infection was pneumococcal LRTI. In secondary infections, Enterococci were the most frequently recovered pathogens in BSI (21.7% of patients), followed by Enterobacterales, mainly K. pneumoniae, while LRTI were mostly associated with Gram-negative bacteria, firstly Enterobacterales (27.4% of patients, K. pneumoniae 15.3%), followed by A. baumannii (19.1%). Fungal infections, both BSI and LRTI, were mostly due to C. albicans. Antibiotic resistance rates were extremely high in Gram-negative bacteria, with almost all A. baumannii isolates resistant to carbapenems (95.5%), and K. pneumoniae and P. aeruginosa showing carbapenem resistance rates of 59.5% and 34.6%, respectively.
Conclusions
In hospitalised patients with severe COVID-19, secondary infections are considerably more common than co-infections, and are mostly due to Gram-negative bacterial pathogens showing a very high rate of antibiotic resistance.
Collapse
|
21
|
Monaco M, Floridia M, Giuliano M, Palmieri L, Lo Noce C, Pantosti A, Palamara AT, Brusaferro S, Onder G. Hospital-acquired bloodstream infections in patients deceased with COVID-19 in Italy (2020-2021). Front Med (Lausanne) 2022; 9:1041668. [PMID: 36465906 PMCID: PMC9713028 DOI: 10.3389/fmed.2022.1041668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/31/2022] [Indexed: 10/06/2023] Open
Abstract
INTRODUCTION In hospitalized patients with COVID-19, bloodstream infections (BSI) are associated with high mortality and high antibiotic resistance rates. The aim of this study was to describe BSI etiology, antimicrobial resistance profile and risk factors in a sample of patients deceased with COVID-19 from the Italian National COVID-19 surveillance. METHODS Hospital charts of patients who developed BSI during hospitalization were reviewed to describe the causative microorganisms and their antimicrobial susceptibility profiles. Risk factors were analyzed in univariate and multivariate analyses. RESULTS The study included 73 patients (71.2% male, median age 70): 40 of them (54.8%) received antibiotics and 30 (41.1%) systemic steroids within 48 h after admission; 53 (72.6%) were admitted to intensive care unit. Early steroid use was associated with a significantly shorter interval between admission and BSI occurrence. Among 107 isolated microorganisms, the most frequent were Enterococcus spp., Candida spp., Acinetobacter baumannii, and Klebsiella pneumoniae. Median time from admission to BSI was shorter for Staphylococcus aureus compared to all other bacteria (8 vs. 24 days, p = 0.003), and longer for Enterococcus spp., compared to all other bacteria (26 vs. 18 days, p = 0.009). Susceptibility tests showed a high rate of resistance, with 37.6% of the bacterial isolates resistant to key antibiotics. Resistance was associated with geographical area [adjusted odds ratio (AOR) for Central/South Italy compared to North Italy: 6.775, p = 0.002], and with early use of systemic steroids (AOR 6.971, p = 0.018). CONCLUSIONS In patients deceased with COVID-19, a large proportion of BSI are caused by antibiotic-resistant bacteria. Early steroid use may facilitate this occurrence.
Collapse
Affiliation(s)
- Monica Monaco
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Floridia
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Giuliano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Luigi Palmieri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Cinzia Lo Noce
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Annalisa Pantosti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Graziano Onder
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
22
|
Cheng X, Zhang Y, Li Y, Wu Q, Wu J, Park SK, Guo C, Lu J. Meta-analysis of 16S rRNA microbial data identified alterations of the gut microbiota in COVID-19 patients during the acute and recovery phases. BMC Microbiol 2022; 22:274. [PMID: 36376804 PMCID: PMC9662111 DOI: 10.1186/s12866-022-02686-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Dozens of studies have demonstrated gut dysbiosis in COVID-19 patients during the acute and recovery phases. However, a consensus on the specific COVID-19 associated bacteria is missing. In this study, we performed a meta-analysis to explore whether robust and reproducible alterations in the gut microbiota of COVID-19 patients exist across different populations. METHODS A systematic review was conducted for studies published prior to May 2022 in electronic databases. After review, we included 16 studies that comparing the gut microbiota in COVID-19 patients to those of controls. The 16S rRNA sequence data of these studies were then re-analyzed using a standardized workflow and synthesized by meta-analysis. RESULTS We found that gut bacterial diversity of COVID-19 patients in both the acute and recovery phases was consistently lower than non-COVID-19 individuals. Microbial differential abundance analysis showed depletion of anti-inflammatory butyrate-producing bacteria and enrichment of taxa with pro-inflammatory properties in COVID-19 patients during the acute phase compared to non-COVID-19 individuals. Analysis of microbial communities showed that the gut microbiota of COVID-19 recovered patients were still in unhealthy ecostates. CONCLUSIONS Our results provided a comprehensive synthesis to better understand gut microbial perturbations associated with COVID-19 and identified underlying biomarkers for microbiome-based diagnostics and therapeutics.
Collapse
Affiliation(s)
- Xiaomin Cheng
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China
- Guangzhou Nansha District Center for Disease Control and Prevention, Guangzhou, China
| | - Yali Zhang
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China
| | - Yifan Li
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China
| | - Qin Wu
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China
| | - Jiani Wu
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China
| | - Soo-Kyung Park
- Division of Gastroenterology, Department of Internal Medicine and Inflammatory Bowel Disease Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Cheng Guo
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA.
| | - Jiahai Lu
- School of Public Health, Sun Yat-sen University, Guangzhou, 510080, Guangdong Province, China.
- One Health Center of Excellence for Research and Training, School of Public Health, Sun Yat-sen University, Guangzhou, China.
- NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou, China.
- Key Laboratory for Tropical Disease Control, Ministry of Education, Guangzhou, China.
- Research Institute of Sun Yat-Sen University in Shenzhen, Shenzhen, China.
- One Health Research Center, Hainan Medical University, Haikou, 571199, China.
| |
Collapse
|
23
|
Long COVID and the Neuroendocrinology of Microbial Translocation Outside the GI Tract: Some Treatment Strategies. ENDOCRINES 2022. [DOI: 10.3390/endocrines3040058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Similar to previous pandemics, COVID-19 has been succeeded by well-documented post-infectious sequelae, including chronic fatigue, cough, shortness of breath, myalgia, and concentration difficulties, which may last 5 to 12 weeks or longer after the acute phase of illness. Both the psychological stress of SARS-CoV-2 infection and being diagnosed with COVID-19 can upregulate cortisol, a stress hormone that disrupts the efferocytosis effectors, macrophages, and natural killer cells, leading to the excessive accumulation of senescent cells and disruption of biological barriers. This has been well-established in cancer patients who often experience unrelenting fatigue as well as gut and blood–brain barrier dysfunction upon treatment with senescence-inducing radiation or chemotherapy. In our previous research from 2020 and 2021, we linked COVID-19 to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) via angiotensin II upregulation, premature endothelial senescence, intestinal barrier dysfunction, and microbial translocation from the gastrointestinal tract into the systemic circulation. In 2021 and 2022, these hypotheses were validated and SARS-CoV-2-induced cellular senescence as well as microbial translocation were documented in both acute SARS-CoV-2 infection, long COVID, and ME/CFS, connecting intestinal barrier dysfunction to disabling fatigue and specific infectious events. The purpose of this narrative review is to summarize what is currently known about host immune responses to translocated gut microbes and how these responses relate to fatiguing illnesses, including long COVID. To accomplish this goal, we examine the role of intestinal and blood–brain barriers in long COVID and other illnesses typified by chronic fatigue, with a special emphasis on commensal microbes functioning as viral reservoirs. Furthermore, we discuss the role of SARS-CoV-2/Mycoplasma coinfection in dysfunctional efferocytosis, emphasizing some potential novel treatment strategies, including the use of senotherapeutic drugs, HMGB1 inhibitors, Toll-like receptor 4 (TLR4) blockers, and membrane lipid replacement.
Collapse
|
24
|
Assante G, Tourna A, Carpani R, Ferrari F, Prati D, Peyvandi F, Blasi F, Bandera A, Le Guennec A, Chokshi S, Patel VC, Cox IJ, Valenti L, Youngson NA. Reduced circulating FABP2 in patients with moderate to severe COVID-19 may indicate enterocyte functional change rather than cell death. Sci Rep 2022; 12:18792. [PMID: 36335131 PMCID: PMC9637119 DOI: 10.1038/s41598-022-23282-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022] Open
Abstract
The gut is of importance in the pathology of COVID-19 both as a route of infection, and gut dysfunction influencing the severity of disease. Systemic changes caused by SARS-CoV-2 gut infection include alterations in circulating levels of metabolites, nutrients and microbial products which alter immune and inflammatory responses. Circulating plasma markers for gut inflammation and damage such as zonulin, lipopolysaccharide and β-glycan increase in plasma along with severity of disease. However, Intestinal Fatty Acid Binding Protein / Fatty Acid Binding Protein 2 (I-FABP/FABP2), a widely used biomarker for gut cell death, has paradoxically been shown to be reduced in moderate to severe COVID-19. We also found this pattern in a pilot cohort of mild (n = 18) and moderately severe (n = 19) COVID-19 patients in Milan from March to June 2020. These patients were part of the first phase of COVID-19 in Europe and were therefore all unvaccinated. After exclusion of outliers, patients with more severe vs milder disease showed reduced FABP2 levels (median [IQR]) (124 [368] vs. 274 [558] pg/mL, P < 0.01). A reduction in NMR measured plasma relative lipid-CH3 levels approached significance (median [IQR]) (0.081 [0.011] vs. 0.073 [0.024], P = 0.06). Changes in circulating lipid levels are another feature commonly observed in severe COVID-19 and a weak positive correlation was observed in the more severe group between reduced FABP2 and reduced relative lipid-CH3 and lipid-CH2 levels. FABP2 is a key regulator of enterocyte lipid import, a process which is inhibited by gut SARS-CoV-2 infection. We propose that the reduced circulating FABP2 in moderate to severe COVID-19 is a marker of infected enterocyte functional change rather than gut damage, which could also contribute to the development of hypolipidemia in patients with more severe disease.
Collapse
Affiliation(s)
- G Assante
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - A Tourna
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - R Carpani
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - F Ferrari
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - D Prati
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - F Peyvandi
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - F Blasi
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - A Bandera
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
| | - A Le Guennec
- Randall Centre for Cell & Molecular Biophysics, King's College, London, UK
| | - S Chokshi
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
| | - V C Patel
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Faculty of Life Sciences & Medicine, King's College, London, UK
- Institute of Liver Studies, King's College Hospital, London, UK
| | - I J Cox
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK.
- Faculty of Life Sciences & Medicine, King's College, London, UK.
| | - L Valenti
- Fondazione IRCSS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy.
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy.
| | - N A Youngson
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK.
- Faculty of Life Sciences & Medicine, King's College, London, UK.
| |
Collapse
|
25
|
Hernández-Solis A, Güemes-González AM, Ruiz-Gómez X, Álvarez-Maldonado P, Castañeda-Casimiro J, Flores-López A, Ramírez-Guerra MA, Muñoz-Miranda O, Madera-Sandoval RL, Arriaga-Pizano LA, Nieto-Patlán A, Estrada-Parra S, Pérez-Tapia SM, Serafín-López J, Chacón-Salinas R, Escobar-Gutiérrez A, Soria-Castro R, Ruiz-Sánchez BP, Wong-Baeza I. IL-6, IL-10, sFas, granulysin and indicators of intestinal permeability as early biomarkers for a fatal outcome in COVID-19. Immunobiology 2022; 227:152288. [PMID: 36209721 PMCID: PMC9527226 DOI: 10.1016/j.imbio.2022.152288] [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: 07/14/2022] [Revised: 09/12/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022]
Abstract
The clinical presentation of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), ranges between mild respiratory symptoms and a severe disease that shares many of the features of sepsis. Sepsis is a deregulated response to infection that causes life-threatening organ failure. During sepsis, the intestinal epithelial cells are affected, causing an increase in intestinal permeability and allowing microbial translocation from the intestine to the circulation, which exacerbates the inflammatory response. Here we studied patients with moderate, severe and critical COVID-19 by measuring a panel of molecules representative of the innate and adaptive immune responses to SARS-CoV-2, which also reflect the presence of systemic inflammation and the state of the intestinal barrier. We found that non-surviving COVID-19 patients had higher levels of low-affinity anti-RBD IgA antibodies than surviving patients, which may be a response to increased microbial translocation. We identified sFas and granulysin, in addition to IL-6 and IL-10, as possible early biomarkers with high sensitivity (>73 %) and specificity (>51 %) to discriminate between surviving and non-surviving COVID-19 patients. Finally, we found that the microbial metabolite d-lactate and the tight junction regulator zonulin were increased in the serum of patients with severe COVID-19 and in COVID-19 patients with secondary infections, suggesting that increased intestinal permeability may be a source of secondary infections in these patients. COVID-19 patients with secondary infections had higher disease severity and mortality than patients without these infections, indicating that intestinal permeability markers could provide complementary information to the serum cytokines for the early identification of COVID-19 patients with a high risk of a fatal outcome.
Collapse
Affiliation(s)
- Alejandro Hernández-Solis
- Servicio de Neumología, Hospital General de México "Dr. Eduardo Liceaga", Secretaría de Salud, Mexico City, Mexico; Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Azmavet M Güemes-González
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ximena Ruiz-Gómez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Pablo Álvarez-Maldonado
- Servicio de Neumología, Hospital General de México "Dr. Eduardo Liceaga", Secretaría de Salud, Mexico City, Mexico
| | - Jessica Castañeda-Casimiro
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico; Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Argelia Flores-López
- Servicio de Neumología, Hospital General de México "Dr. Eduardo Liceaga", Secretaría de Salud, Mexico City, Mexico
| | - Martha Alicia Ramírez-Guerra
- Servicio de Neumología, Hospital General de México "Dr. Eduardo Liceaga", Secretaría de Salud, Mexico City, Mexico
| | - Omar Muñoz-Miranda
- Servicio de Neumología, Hospital General de México "Dr. Eduardo Liceaga", Secretaría de Salud, Mexico City, Mexico
| | - Ruth L Madera-Sandoval
- Unidad de Investigación Médica en Inmunoquímica, Centro Medico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Lourdes A Arriaga-Pizano
- Unidad de Investigación Médica en Inmunoquímica, Centro Medico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Alejandro Nieto-Patlán
- Departamento de Genética, Hospital Infantil de México Federico Gómez, Mexico City, Mexico; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Center for Human Immunobiology, Department of Allergy, Immunology and Rheumatology, Houston, TX, USA.
| | - Sergio Estrada-Parra
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Sonia Mayra Pérez-Tapia
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico; Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Mexico City, Mexico; Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (l+D+i) para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT. Mexico City, Mexico
| | - Jeanet Serafín-López
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rommel Chacón-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Alejandro Escobar-Gutiérrez
- Coordinación de Investigaciones Inmunológicas, Instituto de Diagnóstico y Referencia Epidemiológicos (InDRE), Secretaria de Salud, Mexico City, Mexico
| | - Rodolfo Soria-Castro
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Bibiana Patricia Ruiz-Sánchez
- Facultad de Medicina. Universidad Westhill, Mexico City, Mexico; Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Isabel Wong-Baeza
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico.
| |
Collapse
|
26
|
Lai G, Liu H, Deng J, Li K, Xie B. A Novel 3-Gene Signature for Identifying COVID-19 Patients Based on Bioinformatics and Machine Learning. Genes (Basel) 2022; 13:genes13091602. [PMID: 36140771 PMCID: PMC9498787 DOI: 10.3390/genes13091602] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 12/15/2022] Open
Abstract
Although many biomarkers associated with coronavirus disease 2019 (COVID-19) were found, a novel signature relevant to immune cells has not been developed. In this work, the “CIBERSORT” algorithm was used to assess the fraction of immune infiltrating cells in GSE152641 and GSE171110. Key modules associated with important immune cells were selected by the “WGCNA” package. The “GO” enrichment analysis was used to reveal the biological function associated with COVID-19. The “Boruta” algorithm was used to screen candidate genes, and the “LASSO” algorithm was used for collinearity reduction. A novel gene signature was developed based on multivariate logistic regression analysis. Subsequently, M0 macrophages (PRAUC = 0.948 in GSE152641 and PRAUC = 0.981 in GSE171110) and neutrophils (PRAUC = 0.892 in GSE152641 and PRAUC = 0.960 in GSE171110) were considered as important immune cells. Forty-three intersected genes from two modules were selected, which mainly participated in some immune-related activities. Finally, a three-gene signature comprising CLEC4D, DUSP13, and UNC5A that can accurately distinguish COVID-19 patients and healthy controls in three datasets was constructed. The ROCAUC was 0.974 in the training set, 0.946 in the internal test set, and 0.709 in the external test set. In conclusion, we constructed a three-gene signature to identify COVID-19, and CLEC4D, DUSP13, and UNC5A may be potential biomarkers for COVID-19 patients.
Collapse
|
27
|
Rodrigues PB, Gomes GF, Angelim MKSC, Souza GF, Muraro SP, Toledo-Teixeira DA, Rattis BAC, Passos AS, Pral LP, de Rezende Rodovalho V, dos Santos P. Gomes AB, Matheus VA, Antunes ASLM, Crunfli F, Antunes KH, de Souza APD, Consonni SR, Leiria LO, Alves-Filho JC, Cunha TM, Moraes-Vieira PMM, Proença-Módena JL, R. Vinolo MA. Impact of Microbiota Depletion by Antibiotics on SARS-CoV-2 Infection of K18-hACE2 Mice. Cells 2022; 11:2572. [PMID: 36010648 PMCID: PMC9406363 DOI: 10.3390/cells11162572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/25/2022] Open
Abstract
Clinical and experimental data indicate that severe acute respiratory syndrome coronavirus (SARS-CoV)-2 infection is associated with significant changes in the composition and function of intestinal microbiota. However, the relevance of these effects for SARS-CoV-2 pathophysiology is unknown. In this study, we analyzed the impact of microbiota depletion after antibiotic treatment on the clinical and immunological responses of K18-hACE2 mice to SARS-CoV-2 infection. Mice were treated with a combination of antibiotics (kanamycin, gentamicin, metronidazole, vancomycin, and colistin, Abx) for 3 days, and 24 h later, they were infected with SARS-CoV-2 B lineage. Here, we show that more than 80% of mice succumbed to infection by day 11 post-infection. Treatment with Abx had no impact on mortality. However, Abx-treated mice presented better clinical symptoms, with similar weight loss between infected-treated and non-treated groups. We observed no differences in lung and colon histopathological scores or lung, colon, heart, brain and kidney viral load between groups on day 5 of infection. Despite some minor differences in the expression of antiviral and inflammatory markers in the lungs and colon, no robust change was observed in Abx-treated mice. Together, these findings indicate that microbiota depletion has no impact on SARS-CoV-2 infection in mice.
Collapse
Affiliation(s)
- Patrícia Brito Rodrigues
- Laboratory of Immunoinflammation, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | - Giovanni Freitas Gomes
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
| | - Monara K. S. C. Angelim
- Laboratory of Immunometabolism, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | - Gabriela F. Souza
- Laboratory of Emerging Viruses, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil or
| | - Stefanie Primon Muraro
- Laboratory of Emerging Viruses, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil or
| | - Daniel A. Toledo-Teixeira
- Laboratory of Emerging Viruses, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil or
| | - Bruna Amanda Cruz Rattis
- Department of Pathology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14000-000, Brazil
| | - Amanda Stephane Passos
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
| | - Laís Passarielo Pral
- Laboratory of Immunoinflammation, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | - Vinícius de Rezende Rodovalho
- Laboratory of Immunoinflammation, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | | | - Valquíria Aparecida Matheus
- Laboratory of Immunoinflammation, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | | | - Fernanda Crunfli
- Laboratory of Neuroproteomics, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | - Krist Helen Antunes
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90000-000, Brazil
| | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90000-000, Brazil
| | - Sílvio Roberto Consonni
- Laboratory of Citochemistry and Immunocitochemistry, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | - Luiz Osório Leiria
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
| | - José Carlos Alves-Filho
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
| | - Thiago M. Cunha
- Center of Research in Inflammatory Diseases, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14000-000, Brazil
| | - Pedro M. M. Moraes-Vieira
- Laboratory of Immunometabolism, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas 13000-000, Brazil
- Experimental Medicine Research Cluster, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | - José Luiz Proença-Módena
- Laboratory of Emerging Viruses, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil or
- Experimental Medicine Research Cluster, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | - Marco Aurélio R. Vinolo
- Laboratory of Immunoinflammation, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas 13000-000, Brazil
- Experimental Medicine Research Cluster, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| |
Collapse
|
28
|
Cogliati Dezza F, Arcari G, Alessi F, Valeri S, Curtolo A, Sacco F, Ceccarelli G, Raponi G, Alessandri F, Mastroianni CM, Venditti M, Oliva A. Clinical Impact of COVID-19 on Multi-Drug-Resistant Gram-Negative Bacilli Bloodstream Infections in an Intensive Care Unit Setting: Two Pandemics Compared. Antibiotics (Basel) 2022; 11:926. [PMID: 35884179 PMCID: PMC9312122 DOI: 10.3390/antibiotics11070926] [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: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
Two mutually related pandemics are ongoing worldwide: the COVID-19 and antimicrobial resistance pandemics. This study aims to evaluate the impact of COVID-19 on multi-drug-resistant Gram-negative bacteria (MDR-GN) bloodstream infections (BSIs) in a single intensive care unit (ICU). We conducted a retrospective study including patients admitted to the ICU, reorganized for COVID-19 patients' healthcare, with at least one confirmed MDR-GN BSI during 2019-2020. We compared clinical and microbiological features, incidence density, antibiotic therapy and mortality rate in pre- and during-COVID-19 pandemic periods. We estimated the impact of COVID-19 on mortality by means of univariate Cox regression analyses. A total of 46 patients were included in the study (28 non-COVID-19/18 COVID-19). Overall, 63 BSI episodes occurred (44/19), and non-COVID-19 patients had a higher incidence of MDR-GN BSIs and were more likely to present K. pneumoniae BSIs, while the COVID-19 group showed more A. baumannii BSIs with higher per pathogen incidence. COVID-19 patients presented more critical conditions at the BSI onset, a shorter hospitalization time from BSI to death and higher 30-day mortality rate from BSI onset. COVID-19 and septic shock were associated with 30-day mortality from MDR-GN BSIs, while early active therapy was a protective factor. In conclusion, COVID-19 showed a negative impact on patients with MDR-GN BSIs admitted to the ICU.
Collapse
Affiliation(s)
- Francesco Cogliati Dezza
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.C.D.); (F.A.); (S.V.); (A.C.); (G.C.); (C.M.M.); (M.V.)
| | - Gabriele Arcari
- Microbiology and Virology Laboratory, Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.A.); (F.S.); (G.R.)
| | - Federica Alessi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.C.D.); (F.A.); (S.V.); (A.C.); (G.C.); (C.M.M.); (M.V.)
| | - Serena Valeri
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.C.D.); (F.A.); (S.V.); (A.C.); (G.C.); (C.M.M.); (M.V.)
| | - Ambrogio Curtolo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.C.D.); (F.A.); (S.V.); (A.C.); (G.C.); (C.M.M.); (M.V.)
| | - Federica Sacco
- Microbiology and Virology Laboratory, Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.A.); (F.S.); (G.R.)
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.C.D.); (F.A.); (S.V.); (A.C.); (G.C.); (C.M.M.); (M.V.)
| | - Giammarco Raponi
- Microbiology and Virology Laboratory, Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (G.A.); (F.S.); (G.R.)
| | - Francesco Alessandri
- Department of Anesthesia and Critical Care Medicine, Sapienza University of Rome, Policlinico Umberto I, 00161 Rome, Italy;
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.C.D.); (F.A.); (S.V.); (A.C.); (G.C.); (C.M.M.); (M.V.)
| | - Mario Venditti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.C.D.); (F.A.); (S.V.); (A.C.); (G.C.); (C.M.M.); (M.V.)
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (F.C.D.); (F.A.); (S.V.); (A.C.); (G.C.); (C.M.M.); (M.V.)
| |
Collapse
|
29
|
Rovito R, Augello M, Ben-Haim A, Bono V, d'Arminio Monforte A, Marchetti G. Hallmarks of Severe COVID-19 Pathogenesis: A Pas de Deux Between Viral and Host Factors. Front Immunol 2022; 13:912336. [PMID: 35757770 PMCID: PMC9231592 DOI: 10.3389/fimmu.2022.912336] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Two years into Coronavirus Disease 2019 (COVID-19) pandemic, a comprehensive characterization of the pathogenesis of severe and critical forms of COVID-19 is still missing. While a deep dysregulation of both the magnitude and functionality of innate and adaptive immune responses have been described in severe COVID-19, the mechanisms underlying such dysregulations are still a matter of scientific debate, in turn hampering the identification of new therapies and of subgroups of patients that would most benefit from individual clinical interventions. Here we review the current understanding of viral and host factors that contribute to immune dysregulation associated with COVID-19 severity in the attempt to unfold and broaden the comprehension of COVID-19 pathogenesis and to define correlates of protection to further inform strategies of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Roberta Rovito
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Assaf Ben-Haim
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Valeria Bono
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Antonella d'Arminio Monforte
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Giulia Marchetti
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
30
|
Blaize M, Raoelina A, Kornblum D, Kamus L, Lampros A, Berger M, Demeret S, Constantin JM, Monsel A, Mayaux J, Luyt CE, Piarroux R, Fekkar A. Occurrence of Candidemia in Patients with COVID-19 Admitted to Five ICUs in France. J Fungi (Basel) 2022; 8:jof8070678. [PMID: 35887435 PMCID: PMC9320616 DOI: 10.3390/jof8070678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/16/2022] [Accepted: 06/24/2022] [Indexed: 12/26/2022] Open
Abstract
Whether severe COVID-19 is by itself a significant risk factor for the development of candidemia currently remains an open question as conflicting results have been published. We aim to assess the occurrence of candidemia in patients with severe COVID-19 admitted to the intensive care unit (ICU). We conducted a retrospective study on patients with severe SARS-CoV-2-related pneumonia admitted to 5 ICUs in France who were specifically screened for fungal complications between March 2020 and January 2021. The study population included a total of 264 patients; the median age was 56 years old and most of them were male (n = 186; 70.5%) and immunocompetent (n = 225; 87.5%), and 62.7% (n = 153/244) were on extracorporeal membrane oxygenation support. Microbiological analysis included 4864 blood culture samples and beta-glucan test performed on 975 sera. Candidemia was diagnosed in 13 (4.9%) patients. The species involved were mainly C. albicans (n = 6) and C. parapsilosis (n = 5). Almost all patients (12/13; 92.3%) had a colonization by yeasts. ICU mortality was not significantly impacted by the occurrence of candidemia. Unrelated positive beta-glucan tests were observed in 49 patients (23.4%), including 6 with mold infections and 43 with false positive results. In our series, patients with severe SARS-CoV-2-related pneumonia seemed at low risk of developing invasive candidiasis.
Collapse
Affiliation(s)
- Marion Blaize
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Parasitologie Mycologie, F-75013 Paris, France;
| | - Audrey Raoelina
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Parasitologie Mycologie, F-75013 Paris, France; (A.R.); (D.K.); (L.K.); (A.L.); (M.B.); (R.P.)
| | - Dimitri Kornblum
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Parasitologie Mycologie, F-75013 Paris, France; (A.R.); (D.K.); (L.K.); (A.L.); (M.B.); (R.P.)
| | - Laure Kamus
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Parasitologie Mycologie, F-75013 Paris, France; (A.R.); (D.K.); (L.K.); (A.L.); (M.B.); (R.P.)
| | - Alexandre Lampros
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Parasitologie Mycologie, F-75013 Paris, France; (A.R.); (D.K.); (L.K.); (A.L.); (M.B.); (R.P.)
| | - Marie Berger
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Parasitologie Mycologie, F-75013 Paris, France; (A.R.); (D.K.); (L.K.); (A.L.); (M.B.); (R.P.)
| | - Sophie Demeret
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Réanimation Neurologique, F-75013 Paris, France;
| | - Jean-Michel Constantin
- Sorbonne Université, GRC 29, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Département d’Anesthésie Réanimation, F-75013 Paris, France;
| | - Antoine Monsel
- Sorbonne Université, INSERM, Immunology Immunopathology Immunotherapy (I3), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Département d’Anesthésie Réanimation, F-75013 Paris, France;
| | - Julien Mayaux
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Réanimation Médicale, F-75013 Paris, France;
| | - Charles-Edouard Luyt
- Sorbonne Université, INSERM, Institute of Cardiometabolism and Nutrition, Assistance Publique–Hôpitaux de Paris (APHP), Médecine Intensive Réanimation-Institut de Cardiologie, F-75013 Paris, France;
| | - Renaud Piarroux
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Parasitologie Mycologie, F-75013 Paris, France; (A.R.); (D.K.); (L.K.); (A.L.); (M.B.); (R.P.)
| | - Arnaud Fekkar
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Cimi-Paris, Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Hospitalier La Pitié-Salpêtrière, Parasitologie Mycologie, F-75013 Paris, France;
- Correspondence: ; Tel.: +33-1-42-16-01-84
| |
Collapse
|
31
|
Kılıç AO, Akın F, Yazar A, Metin Akcan Ö, Topcu C, Aydın O. Zonulin and claudin-5 levels in multisystem inflammatory syndrome and SARS-CoV-2 infection in children. J Paediatr Child Health 2022; 58:1561-1565. [PMID: 35638118 PMCID: PMC9347651 DOI: 10.1111/jpc.16033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 12/22/2022]
Abstract
AIM SARS-CoV-2 infection in children is generally asymptomatic or mild; however, it can lead to a life-threatening clinical condition, multisystem inflammatory syndrome in children (MIS-C), days or weeks after the infection. Increased intestinal permeability isa possible triggering factor at the onset of the hyperinflammation associated with MIS-C. Zonulin and claudin-5 are involved in intestinal permeability. In this study, we aimed to investigate serum zonulin and claudin-5 levels in SARS-CoV-2 infection and MIS-C disease. METHODS The study group consisted of children diagnosed with MIS-C or SARS-CoV-2 infection who presented to a university hospital paediatric emergency or infectious diseases departments. The control group included well patients seen at the General Pediatrics units for routine follow-up. Serum zonulin and claudin-5 levels were measured at the time of diagnosis. RESULTS Fifteen patients were included in the MIS-C group, 19 in the SARS-CoV-2 infection group and 21 in the control group. The mean zonulin level in the MIS-C group was significantly higher than in the control group (P < 0.001). Mean Claudin-5 levels were Psignificantly lower in the SARS-CoV-2 infection group than in the control group (P < 0.001). CONCLUSION These results indicate that increased intestinal permeability may be involved in the pathogenesis of SARS-CoV-2 infection and MIS-C disease. Larger clinical trials are needed to clarify the role of serum zonulin and claudin-5 on intestinal permeability in MIS-C and SARS-CoV-2 infection in children.
Collapse
Affiliation(s)
- Ahmet O Kılıç
- Department of Pediatrics, Meram Medical FacultyNecmettin Erbakan UniversityKonyaTurkey
| | - Fatih Akın
- Department of Pediatrics, Meram Medical FacultyNecmettin Erbakan UniversityKonyaTurkey
| | - Abdullah Yazar
- Department of Pediatrics, Meram Medical FacultyNecmettin Erbakan UniversityKonyaTurkey
| | - Özge Metin Akcan
- Department of Pediatrics, Meram Medical FacultyNecmettin Erbakan UniversityKonyaTurkey
| | - Cemile Topcu
- Department of Biochemistry, Meram Medical FacultyNecmettin Erbakan UniversityKonyaTurkey
| | - Orkun Aydın
- Department of PediatricsDr. Sami Ulus Maternity and Children's Health and Diseases Training and Research HospitalAnkaraTurkey
| |
Collapse
|
32
|
Mazer MB, Bulut Y, Brodsky NN, Lam FW, Sturgill JL, Miles SM, Shein SL, Carroll CL, Remy KE. Multisystem Inflammatory Syndrome in Children: Host Immunologic Responses. Pediatr Crit Care Med 2022; 23:315-320. [PMID: 35050932 PMCID: PMC9058188 DOI: 10.1097/pcc.0000000000002897] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Monty B. Mazer
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, UH Rainbow Babies and Children’s Hospital, Cleveland, OH
| | - Yonca Bulut
- Department of Pediatrics, Division of Critical Care Medicine David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Nina N. Brodsky
- Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, P.O.Box 208064, New Haven, CT
- Department of Immunobiology, Yale University School of Medicine, 300 George Street 353G, New Haven, CT
| | - Fong W. Lam
- Department of Pediatrics, Division of Critical Care Medicine, Baylor College of Medicine, Houston, TX
| | - Jamie L. Sturgill
- University of Kentucky College of Medicine, Department of Internal Medicine, Division of Pulmonary Critical Care and Sleep Medicine, Lexington, KY
| | - Sydney M. Miles
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Steven L. Shein
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, UH Rainbow Babies and Children’s Hospital, Cleveland, OH
| | | | - Kenneth E. Remy
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, UH Rainbow Babies and Children’s Hospital, Cleveland, OH
- Department of Internal Medicine, Division of Pulmonary Critical Care Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pediatrics, Division of Critical Care Medicine, Washington University in St. Louis School of Medicine, St. Louis, MO
| |
Collapse
|
33
|
Maes M, Tedesco Junior WLD, Lozovoy MAB, Mori MTE, Danelli T, Almeida ERDD, Tejo AM, Tano ZN, Reiche EMV, Simão ANC. In COVID-19, NLRP3 inflammasome genetic variants are associated with critical disease and these effects are partly mediated by the sickness symptom complex: a nomothetic network approach. Mol Psychiatry 2022; 27:1945-1955. [PMID: 35022530 PMCID: PMC8752583 DOI: 10.1038/s41380-021-01431-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/08/2021] [Accepted: 12/23/2021] [Indexed: 12/21/2022]
Abstract
In coronavirus disease (COVID-19), the nucleotide-binding domain, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome is activated in response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Acute infections are accompanied by a sickness symptom complex (SSC) which is highly conserved and protects against infections and hyperinflammation. The aim of this study is to delineate the associations of COVID-19, SSC and NLPR3 rs10157379 T > C and NLPR3 rs10754558 C > G variants; and the protective role of SSC in SARS-CoV-2 infection. We recruited COVID-19 patients, 308 with critical, 63 with moderate and 157 with mild disease. Increased SSC protects against SARS, critical disease, and death due to COVID-19. Increasing age, male sex and rs10754558 CG significantly reduce SSC protection. The rs10157379 CT and rs10754558 GG genotypes are positively associated with SARS. Partial Least Squares analysis shows that a) 41.8% of the variance in critical COVID-19 symptoms is explained by SSC and oxygen saturation (inversely associated), inflammation, chest computed tomography abnormalities, increased body mass index, SARS and age (positively associated); and b) the effects of the NLRP3 rs10157379 and rs10754558 variants on critical COVID-19 are mediated via SSC (protective) and SARS (detrimental). SSC includes anosmia and dysgeusia, and maybe gastrointestinal symptoms. In conclusion, intersections among the rs10754558 variant, age, and sex increase risk towards critical COVID-19 by attenuating SSC. NLRP3 variants play an important role in SARS, and severe and critical COVID-19 especially in elderly male individuals with reduced SSC and with increased BMI, hypertension, and diabetes type 2.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
| | | | - Marcell Alysson Batisti Lozovoy
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
- Department of Pathology, Clinical Analysis and Toxicology, Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
| | | | - Tiago Danelli
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
| | - Elaine Regina Delicato de Almeida
- Department of Pathology, Clinical Analysis and Toxicology, Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
| | | | | | - Edna Maria Vissoci Reiche
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
- Department of Pathology, Clinical Analysis and Toxicology, Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
| | - Andréa Name Colado Simão
- Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
- Department of Pathology, Clinical Analysis and Toxicology, Laboratory of Research in Applied Immunology, University of Londrina, Londrina, PR, Brazil
| |
Collapse
|
34
|
Peluso MJ, Donatelli J, Henrich TJ. Long-term immunologic effects of SARS-CoV-2 infection: leveraging translational research methodology to address emerging questions. Transl Res 2022; 241:1-12. [PMID: 34780969 PMCID: PMC8588584 DOI: 10.1016/j.trsl.2021.11.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 11/01/2022]
Abstract
The current era of COVID-19 is characterized by emerging variants of concern, waning vaccine- and natural infection-induced immunity, debate over the timing and necessity of vaccine boosting, and the emergence of post-acute sequelae of SARS-CoV-2 infection. As a result, there is an ongoing need for research to promote understanding of the immunology of both natural infection and prevention, especially as SARS-CoV-2 immunology is a rapidly changing field, with new questions arising as the pandemic continues to grow in complexity. The next phase of COVID-19 immunology research will need focus on clearer characterization of the immune processes defining acute illness, development of a better understanding of the immunologic processes driving protracted symptoms and prolonged recovery (ie, post-acute sequelae of SARS-CoV-2 infection), and a growing focus on the impact of therapeutic and prophylactic interventions on the long-term consequences of SARS-CoV-2 infection. In this review, we address what is known about the long-term immune consequences of SARS-CoV-2 infection and propose how experience studying the translational immunology of other infections might inform the approach to some of the key questions that remain.
Collapse
Affiliation(s)
- Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, California
| | - Joanna Donatelli
- Division Experimental Medicine, University of California, San Francisco, California
| | - Timothy J Henrich
- Division Experimental Medicine, University of California, San Francisco, California.
| |
Collapse
|
35
|
Santinelli L, Laghi L, Innocenti GP, Pinacchio C, Vassalini P, Celani L, Lazzaro A, Borrazzo C, Marazzato M, Tarsitani L, Koukopoulos AE, Mastroianni CM, d'Ettorre G, Ceccarelli G. Oral Bacteriotherapy Reduces the Occurrence of Chronic Fatigue in COVID-19 Patients. Front Nutr 2022; 8:756177. [PMID: 35096923 PMCID: PMC8790565 DOI: 10.3389/fnut.2021.756177] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Long COVID refers to patients with symptoms as fatigue, “brain fog,” pain, suggesting the chronic involvement of the central nervous system (CNS) in COVID-19. The supplementation with probiotic (OB) would have a positive effect on metabolic homeostasis, negatively impacting the occurrence of symptoms related to the CNS after hospital discharge. On a total of 58 patients hospitalized for COVID-19, 24 (41.4%) received OB during hospitalization (OB+) while 34 (58.6%) taken only the standard treatment (OB–). Serum metabolomic profiling of patients has been performed at both hospital acceptance (T0) and discharge (T1). Six months after discharge, fatigue perceived by participants was assessed by administrating the Fatigue Assessment Scale. 70.7% of participants reported fatigue while 29.3% were negative for such condition. The OB+ group showed a significantly lower proportion of subjects reporting fatigue than the OB– one (p < 0.01). Furthermore, OB+ subjects were characterized by significantly increased concentrations of serum Arginine, Asparagine, Lactate opposite to lower levels of 3-Hydroxyisobutirate than those not treated with probiotics. Our results strongly suggest that in COVID-19, the administration of probiotics during hospitalization may prevent the development of chronic fatigue by impacting key metabolites involved in the utilization of glucose as well as in energy pathways.
Collapse
Affiliation(s)
- Letizia Santinelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Luca Laghi
- Department of Agricultural and Food Sciences, University of Bologna, Bologna, Italy.,Interdepartmental Centre for Agri-Food Industrial Research, University of Bologna, Bologna, Italy
| | | | - Claudia Pinacchio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Paolo Vassalini
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Luigi Celani
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Alessandro Lazzaro
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Cristian Borrazzo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Massimiliano Marazzato
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Lorenzo Tarsitani
- Department of Human Neurosciences, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Alexia E Koukopoulos
- Department of Human Neurosciences, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Claudio M Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
36
|
Abstract
High expression of the transmembrane protein angiotensin I converting enzyme 2 (ACE2), more than 100-times higher as in the lung, and transmembrane serine protease 2 (TMPRSS2) in the gastrointestinal tract leads to infection with SARS-CoV-2. According to meta-analysis data, 9.8–20% of COVID-19 patients experience gastrointestinal symptoms, where diarrhoea is the most frequent, and about 50% shed viruses with high titre through their faeces, where a first faecal transmission was reported. Furthermore, gut inflammation, intestinal damage, and weakening of the gut mucosal integrity that leads to increased permeability has been shown in different studies for COVID-19 patients. This can lead to increased inflammation and bacteraemia. Low mucosal integrity combined with low intestinal damage is a good predictor for disease progression and submission to the intensive care unit (ICU). Several pilot studies have shown that the gut microbiome of COVID-19 patients is changed, microbial richness and diversity were lower, and opportunistic pathogens that can cause bacteraemia were enriched compared to a healthy control group. In a large proportion of these patients, dysbiosis was not resolved at discharge from the hospital and one study showed dysbiosis is still present after 3 months post COVID-19. Consequently, there might be a link between dysbiosis of the gut microbiome in COVID-19 patients and chronic COVID-19 syndrome (CCS). Various clinical trials are investigating the benefit of probiotics for acute COVID-19 patients, the majority of which have not reported results yet. However, two clinical trials have shown that a certain combination of probiotics is beneficial and safe for acute COVID-19 patients. Mortality was 11% for the probiotic treatment group, and 22% for the control group. Furthermore, for the probiotic group, symptoms cleared faster, and an 8-fold decreased risk of developing a respiratory failure was calculated. In conclusion, evidence is arising that inflammation, increased permeability, and microbiome dysbiosis in the gut occur in COVID-19 patients and thus provide new targets for adjuvant treatments of acute and chronic COVID-19. More research in this area is needed.
Collapse
|