1
|
Mezouar S, Mege J. Monitoring Macrophage Polarization in Infectious Disease, Lesson From SARS-CoV-2 Infection. Rev Med Virol 2025; 35:e70034. [PMID: 40148134 PMCID: PMC11976041 DOI: 10.1002/ird3.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The concept of macrophage polarization has been largely used in human diseases to define a typology of activation of myeloid cells reminiscent of lymphocyte functional subsets. In COVID-19, several studies have investigated myeloid compartment dysregulation and macrophage polarization as an indicator of disease prognosis and monitoring. SARS-CoV-2 induces an in vitro activation state in monocytes and macrophages that does not match the polarization categories in most studies. In COVID-19 patients, monocytes and macrophages are activated but they do not show a polarization profile. Therefore, the investigation of polarization under basic conditions was not relevant to assess monocyte and macrophage activation. The analysis of monocytes and macrophages with high-throughput methods has allowed the identification of new functional subsets in the context of COVID-19. This approach proposes an innovative stratification of myeloid cell activation. These new functional subsets of myeloid cells would be better biomarkers to assess the risk of complications in COVID-19, reserving the concept of polarization for pharmacological programme evaluation. This review reappraises the polarization of monocytes and macrophages in viral infections, particularly in COVID-19.
Collapse
Affiliation(s)
- Soraya Mezouar
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Faculty of Medical and Paramedical SciencesAix‐Marseille UniversityHIPE Human LabMarseilleFrance
| | - Jean‐Louis Mege
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Department of ImmunologyLa Timone HospitalMarseilleFrance
| |
Collapse
|
2
|
Tian J, Zhang L, Wang X, Huang G, Li Z, Gong L, Liu D. Clinical features and survival analysis of adult patients with severe lower respiratory tract infection positive for respiratory syncytial virus. BMC Pulm Med 2025; 25:184. [PMID: 40247280 PMCID: PMC12007204 DOI: 10.1186/s12890-025-03660-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND The awareness and attention toward lower respiratory tract infections caused by respiratory syncytial virus (RSV) are lower in adults than in children. Moreover, the clinical data on severe RSV infections in adults are lacking. This study aimed to augment the clinical understanding of RSV infections. METHOD This study was performed on 191 adult patients with severe lower respiratory tract infections admitted to the intensive care unit (ICU) of Zunyi Medical University Affiliated Hospital (Zhuhai). The sputum specimens were collected for RSV testing, and the clinical data were collected and analyzed statistically. RESULTS The findings revealed that patients in the RSV-positive group were older, had a higher prevalence of underlying diseases, were more likely to require noninvasive mechanical ventilation, and had prolonged hospital stays compared with those in the RSV-negative group. Furthermore, Kaplan-Meier survival analysis conducted within 90 days revealed comparable mortality rates between the RSV-positive and RSV-negative groups. In the RSV-positive group, elderly patients and patients with congestive heart failure or chronic obstructive pulmonary disease had lower survival rates. CONCLUSION Patients in the RSV-positive group, particularly elderly patients and patients with congestive heart failure or, experience reduced survival durations compared with those in the RSV-negative group.
Collapse
Affiliation(s)
- Jiahua Tian
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, China
| | - Xunling Wang
- Department of Clinical Medicine, Zunyi Medical University, Zunyi, 563000, China
| | - Guichuan Huang
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, China
| | - Zhu Li
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, China
| | - Ling Gong
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, China.
| | - Daishun Liu
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, 550000, China.
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China.
| |
Collapse
|
3
|
Sierra-López F, Iglesias-Vazquez V, Baylon-Pacheco L, Ríos-Castro E, Osorio-Trujillo JC, Lagunes-Guillén A, Chávez-Munguía B, Hernández SB, Acosta-Altamirano G, Talamás-Rohana P, Rosales-Encina JL, Sierra-Martínez M. A Fraction of Escherichia coli Bacteria Induces an Increase in the Secretion of Extracellular Vesicle Polydispersity in Macrophages: Possible Involvement of Secreted EVs in the Diagnosis of COVID-19 with Bacterial Coinfections. Int J Mol Sci 2025; 26:3741. [PMID: 40332365 PMCID: PMC12027499 DOI: 10.3390/ijms26083741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Extracellular vesicles (EVs) can transport molecules that combat viruses, such as RNA against SARS-CoV-2. Bacterial coinfections can help establish certain viruses and worsen diseases. Thus, we designed a model to induce the secretion of polydisperse EVs shown with SARS-CoV-2 and bacterial coinfection using macrophages and E. coli fractions as in vitro inducers. We obtained short and large macrophage EVs. The E. coli fraction was designated as SDS-soluble bacterial membrane fraction and its associated proteins (SDS-SBMF). The proteins were identified using a mass spectrometer. SDS-SBMF contained mainly OmpF, OmpA, OmpC, OmpX, and lpp. The SDS-SBMF macrophages induced the secretion of polydisperse EVs at 30 min, reaching optimal secretion at 120 min, as observed via scanning electron microscopy and confocal microscopy. Macrophage EVs contained mainly HSP7C, actin, apolipoprotein, GAPDH, annexin A5, PKM, moesin, and cofilin. We observed an increase in EVs in the bloodstream of patients with SARS-CoV-2 and bacterial coinfection, in addition to the presence of SARS-CoV-2 genes (E, ORF) in EVs. This in vitro method for inducing EVs has the potential to be used to obtain larger samples for study and for the detection of diagnostic and prognostic biomarkers of different diseases.
Collapse
Affiliation(s)
- Francisco Sierra-López
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco, Mexico City 07360, Mexico; (F.S.-L.); (L.B.-P.); (J.C.O.-T.); (A.L.-G.); (B.C.-M.); (P.T.-R.)
| | - Vanessa Iglesias-Vazquez
- Unidad de Investigación en Salud, Hospital Regional de Alta Especialidad de Ixtapaluca, Servicios de Salud del Instituto Mexicano del Seguro Social para el Bienestar (IMSS-BIENESTAR), Mexico, Carr Mex-Puebla Km 34.5 col., Zoquiapan, Mexico City 56530, Mexico; (V.I.-V.); (S.B.H.)
| | - Lidia Baylon-Pacheco
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco, Mexico City 07360, Mexico; (F.S.-L.); (L.B.-P.); (J.C.O.-T.); (A.L.-G.); (B.C.-M.); (P.T.-R.)
| | - Emmanuel Ríos-Castro
- Genomics, Proteomics and Metabolomics Core Facility (UGPM) LaNSE, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco, Mexico City 07360, Mexico
| | - Juan Carlos Osorio-Trujillo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco, Mexico City 07360, Mexico; (F.S.-L.); (L.B.-P.); (J.C.O.-T.); (A.L.-G.); (B.C.-M.); (P.T.-R.)
| | - Anel Lagunes-Guillén
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco, Mexico City 07360, Mexico; (F.S.-L.); (L.B.-P.); (J.C.O.-T.); (A.L.-G.); (B.C.-M.); (P.T.-R.)
| | - Bibiana Chávez-Munguía
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco, Mexico City 07360, Mexico; (F.S.-L.); (L.B.-P.); (J.C.O.-T.); (A.L.-G.); (B.C.-M.); (P.T.-R.)
| | - Susana Bernardo Hernández
- Unidad de Investigación en Salud, Hospital Regional de Alta Especialidad de Ixtapaluca, Servicios de Salud del Instituto Mexicano del Seguro Social para el Bienestar (IMSS-BIENESTAR), Mexico, Carr Mex-Puebla Km 34.5 col., Zoquiapan, Mexico City 56530, Mexico; (V.I.-V.); (S.B.H.)
| | - Gustavo Acosta-Altamirano
- Hospital General de México, Eje 2A Sur (Dr. Balmis) No. 148, Cuauhtémoc, Doctores, CDMX, Mexico City 06726, Mexico;
| | - Patricia Talamás-Rohana
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco, Mexico City 07360, Mexico; (F.S.-L.); (L.B.-P.); (J.C.O.-T.); (A.L.-G.); (B.C.-M.); (P.T.-R.)
| | - José Luis Rosales-Encina
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Av. IPN 2508, Zacatenco, Mexico City 07360, Mexico; (F.S.-L.); (L.B.-P.); (J.C.O.-T.); (A.L.-G.); (B.C.-M.); (P.T.-R.)
| | - Mónica Sierra-Martínez
- Unidad de Investigación en Salud, Hospital Regional de Alta Especialidad de Ixtapaluca, Servicios de Salud del Instituto Mexicano del Seguro Social para el Bienestar (IMSS-BIENESTAR), Mexico, Carr Mex-Puebla Km 34.5 col., Zoquiapan, Mexico City 56530, Mexico; (V.I.-V.); (S.B.H.)
| |
Collapse
|
4
|
Gallo A, Sammartino JC, Vazzana R, Giambruno R, Carcione C, Cuscino N, Castelbuono S, Miceli V, Bulati M, Lilleri D, Cassaniti I, Conaldi PG, Baldanti F. Transcriptomic profiles of monocyte-derived macrophages exposed to SARS-CoV-2 VOCs reveal immune-evasion escape driven by delta. J Transl Med 2025; 23:151. [PMID: 39905461 PMCID: PMC11796281 DOI: 10.1186/s12967-025-06158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Since the breakout of COVID-19, the mutated forms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have shown enhanced rates of transmission and adaptation to humans. The variants of concern (VOC), designated Alpha, Beta, Gamma, Delta, and Omicron emerged independent of one another, and in turn rapidly became dominant. The success of each VOC, as well as the virus fitness, were enabled by altered intrinsic functional properties and, reasonably, to virus antigenicity changes, conferring the ability to evade a primed immune response. METHODS We analysed the gene expression profiles of monocyte-derived macrophages (MDM) isolated from whole blood of healthy participants exposed to the 5 different SARS-CoV-2 VOC: D614G, Alpha (B.1.1.7), Gamma (P1), Delta (B.1.617.2), and Omicron BA.1 (B.1.1.529), and to the HCoV-OC43 strain, a coronavirus already present in the population before the SARS-CoV-2 pandemic. Whole transcriptome RNA-Seq, for both coding and non-coding RNAs, was then made. RESULTS After exposure to the 5 VOC of MDM, we initially assessed the presence of the viral SARS-CoV-2 transcripts to confirm viral entry. We then analysed the RNA-Seq data and observed a significant deregulation of both coding and non-coding RNAs. In particular, our RNA-Seq analysis showed a significant up-regulation of several genes involved in different immunological processes, such as PARP9/PARP14 axes, in macrophages exposed to D614G, Alpha, and Gamma variants. Surprisingly, our data showed that macrophages exposed to the Delta variant exhibited a transcriptional profile more similar to the naïve control group, while macrophages exposed to the Omicron variant showed intermediate differentially expressed genes (DEGs) between the two groups. By checking the canonical markers for M1/M2 differentiation states, we did not observe any expression in macrophages exposed to the Delta variant, suggesting an M0 status, comparable to the naïve control group. Finally, we observed a significant deregulation of 3 main types of non-coding RNAs (ncRNAs): long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and small nucleolar RNAs (snoRNAs), some of which are common to coronaviruses, and some specific to SARS-CoV-2. CONCLUSION The SARS-CoV-2-dependent alteration of the transcriptome of monocyte-derived macrophage (MDM)-infected cells can be linked to the chronological order of the variants' appearance in the human population. Our data suggest an evolution of VOC in modulating the host immune response, with a strong change in pace beginning with the advent of the Delta variant. MDMs exposed to Delta showed a failure in the activation of the adaptive immune response, and this correlates with the more severe symptoms developed by people affected with this SARS-CoV-2 variant.
Collapse
Affiliation(s)
- Alessia Gallo
- Department of Research, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy.
| | - Josè Camilla Sammartino
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Università degli Studi di Pavia, Pavia, Italy
| | - Roberta Vazzana
- Department of Research, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Roberto Giambruno
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | | | - Nicola Cuscino
- Department of Research, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Salvatore Castelbuono
- Department of Research, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Vitale Miceli
- Department of Research, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Matteo Bulati
- Department of Research, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Daniele Lilleri
- Microbiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Irene Cassaniti
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Università degli Studi di Pavia, Pavia, Italy
- Microbiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Pier Giulio Conaldi
- Department of Research, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Palermo, Italy
| | - Fausto Baldanti
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, Università degli Studi di Pavia, Pavia, Italy
- Microbiology and Virology Department, Fondazione Istituto di ricovero e cura a carattere scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
5
|
Amoroso D, Bongo S, Copponi A, Rossi V, Di Giorgio R, Bernardini S, Ippoliti L, Morello M. A Review of the Hematological Picture of Severe COVID-19 Infection. Cureus 2025; 17:e78797. [PMID: 39931501 PMCID: PMC11808344 DOI: 10.7759/cureus.78797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2025] [Indexed: 02/13/2025] Open
Abstract
Numerous hematological abnormalities have been documented in COVID-19 patients. We conducted an analysis of 82 articles from PubMed, focusing on the hematological characteristics observed in survivors (S) and non-survivors (NS) with moderate and severe COVID-19 symptoms, respectively. Our review underlines neutrophilia, lymphopenia, and thrombocytopenia as hallmark features of the disease. In severe cases, blood cell microscopy revealed the following abnormalities: i) an increased number of neutrophils, often displaying granularity, toxic granulation, and vacuolization; ii) lymphocytes with a notably blue cytoplasm; iii) several monocytes that contain vacuoles; iv) platelet aggregation; and v) basophilic stippling in red blood cells. Furthermore, scattergram analysis of COVID-19 patients revealed two common features: i) an increased neutrophil population and ii) the presence of a distinctive "sandglass pattern". This review underscores the critical role of hematochemical and cytomorphological blood cell analysis in COVID-19 patients, aiding clinicians in better recognizing and understanding the indicators of disease severity.
Collapse
Affiliation(s)
- Dominga Amoroso
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Stefania Bongo
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Anna Copponi
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Vanessa Rossi
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Roberta Di Giorgio
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Sergio Bernardini
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Lorenzo Ippoliti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, ITA
| | - Maria Morello
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| |
Collapse
|
6
|
Bae SM, Hyun CG. Insights on the Anti-Inflammatory and Anti-Melanogenic Effects of 2'-Hydroxy-2,6'-dimethoxychalcone in RAW 264.7 and B16F10 Cells. Curr Issues Mol Biol 2025; 47:85. [PMID: 39996806 PMCID: PMC11854153 DOI: 10.3390/cimb47020085] [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: 01/15/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Chalcones are recognized for their diverse pharmacological properties, including anti-inflammatory and anti-melanogenic effects. However, studies on 2'-hydroxy-2-methoxychalcone derivatives remain limited. This study investigated the anti-inflammatory and melanin synthesis-inhibitory effects of three derivatives: 2'-hydroxy-2,4-dimethoxychalcone (2,4-DMC), 2'-hydroxy-2,5'-dimethoxychalcone (2,5'-DMC), and 2'-hydroxy-2,6'-dimethoxychalcone (2,6'-DMC). In lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages, 2,6'-DMC demonstrated a superior inhibition of nitric oxide (NO) production, pro-inflammatory cytokines, and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) compared to the other derivatives. A mechanistic analysis revealed that 2,6'-DMC modulates the NF-κB and MAPK signaling pathways to attenuate inflammation. Additionally, 2,6'-DMC exhibited a significant inhibition of α-melanocyte-stimulating hormone (α-MSH)-induced melanin synthesis in B16F10 melanoma cells by downregulating tyrosinase, TRP-1, TRP-2, and MITF expression. This regulation was achieved through the suppression of the Wnt/β-catenin, PI3K/AKT, MAPK, and PKA/CREB pathways. Compared to 2,4-DMC and 2,5'-DMC, 2,6'-DMC's structural configuration, characterized by methoxy groups at the 2- and 6'-positions, contributed to its enhanced molecular stability and binding affinity, amplifying its inhibitory effects. A primary skin irritation test confirmed that 2,6'-DMC exhibited minimal irritation, demonstrating its safety for dermal applications. These findings suggest that 2,6'-DMC holds promise as a dual-function agent for managing inflammatory conditions and hyperpigmentation-related disorders.
Collapse
Affiliation(s)
| | - Chang-Gu Hyun
- Department of Chemistry and Cosmetics, Jeju Inside Agency and Cosmetic Science Center, Jeju National University, Jeju 63243, Republic of Korea;
| |
Collapse
|
7
|
Yang Y, Yan L, Zhang H, Xiao C, Wang K. Atomized Neutrophil Membrane-coated MOF Nanoparticles for Direct Delivery of Dexamethasone for Severe Pneumonia. FRONT BIOSCI-LANDMRK 2025; 30:26721. [PMID: 39862093 DOI: 10.31083/fbl26721] [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: 09/24/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Dexamethasone has proven life-saving in severe acute respiratory syndrome (SARS) and COVID-19 cases. However, its systemic administration is accompanied by serious side effects. Inhalation delivery of dexamethasone (Dex) faces challenges such as low lung deposition, brief residence in the respiratory tract, and the pulmonary mucus barrier, limiting its clinical use. Neutrophil cell membrane-derived nanovesicles, with their ability to specifically target hyper-activated immune cells and excellent mucus permeability, emerge as a promising carrier for pulmonary inhalation therapy. METHODS We designed a novel UiO66 metal-organic framework nanoparticle loaded with Dex and coated with neutrophil cell membranes (UiO66-Dex@NMP) for targeted therapy of severe pneumonia. This was achieved by loading Dex into UiO66 pores and subsequently coating with neutrophil membranes for functionalization. RESULTS Drug release experiments revealed UiO66-Dex@NMP to exhibit favorable sustained-release properties. Additionally, UiO66-Dex@NMP demonstrated excellent targeting capabilities both in vitro and in vivo. In a mouse model of lipopolysaccharide (LPS)-induced pneumonia, UiO66-Dex@NMP significantly reduced lung inflammation compared to both the control model and Dex administered via inhalation. Histopathological analysis further confirmed UiO66-Dex@NMP's ability to alleviate lung tissue damage. CONCLUSIONS UiO66-Dex@NMP represents a novel and safe inhaled delivery carrier for Dex, offering valuable insights into the clinical management of respiratory diseases, including severe pneumonia.
Collapse
Affiliation(s)
- Yixiao Yang
- Institute of Translational Medicine, Shanghai University, 200444 Shanghai, China
- Institute of Burn Research, The First Affiliated Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Lizhen Yan
- Department of Respiratory and Critical Care Medicine, Zibo Municipal Hospital, 255400 Zibo, Shandong, China
| | - Han Zhang
- Institute of Translational Medicine, Shanghai University, 200444 Shanghai, China
- MPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical College, Nantong University, 226001 Nantong, Jiangsu, China
| | - Chuanguang Xiao
- Department of Breast Thyroid Surgery, Zibo Central Hospital, 255036 Zibo, Shandong, China
| | - Kai Wang
- Institute of Translational Medicine, Shanghai University, 200444 Shanghai, China
| |
Collapse
|
8
|
Li J, Ma W, Tang Z, Li Y, Zheng R, Xie Y, Li G. Macrophage‑driven pathogenesis in acute lung injury/acute respiratory disease syndrome: Harnessing natural products for therapeutic interventions (Review). Mol Med Rep 2025; 31:16. [PMID: 39513609 PMCID: PMC11551695 DOI: 10.3892/mmr.2024.13381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024] Open
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a common respiratory disease characterized by hypoxemia and respiratory distress. It is associated with high morbidity and mortality. Due to the complex pathogenesis of ALI, the clinical management of patients with ALI/ARDS is challenging, resulting in numerous post‑treatment sequelae and compromising the quality of life of patients. Macrophages, as a class of innate immune cells, play an important role in ALI/ARDS. In recent years, the functions and phenotypes of macrophages have been better understood due to the development of flow cytometry, immunofluorescence, single‑cell sequencing and spatial genomics. However, no macrophage‑targeted drugs for the treatment of ALI/ARDS currently exist in clinical practice. Natural products are important for drug development, and it has been shown that numerous natural compounds from herbal medicine can alleviate ALI/ARDS caused by various factors by modulating macrophage abnormalities. In the present review, the natural products from herbal medicine that can modulate macrophage abnormalities in ALI/ARDS to treat ALI/ARDS are introduced, and their mechanisms of action, discovered in the previous five years (2019‑2024), are presented. This will provide novel ideas and directions for further research, to develop new drugs for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Jincun Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Wenyu Ma
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Zilei Tang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yingming Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Ruiyu Zheng
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Yuhuan Xie
- Yunnan Innovation Team of Application Research on Traditional Chinese Medicine Theory of Disease Prevention, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Gang Li
- Yunnan Provincial University Key Laboratory of Aromatic Chinese Herb Research, Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
- Basic Medical School, Yunnan University of Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
9
|
Kim YJ, Lee SG, Park SY, Jeon SM, Kim SI, Kim KT, Roh T, Lee SH, Lee MJ, Lee J, Kim HJ, Lee SE, Kim JK, Heo JY, Kim IS, Park C, Paik S, Jo EK. Ubiquitin regulatory X (UBX) domain-containing protein 6 is essential for autophagy induction and inflammation control in macrophages. Cell Mol Immunol 2024; 21:1441-1458. [PMID: 39438692 PMCID: PMC11606977 DOI: 10.1038/s41423-024-01222-1] [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: 04/30/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Ubiquitin regulatory X (UBX) domain-containing protein 6 (UBXN6) is an essential cofactor for the activity of the valosin-containing protein p97, an adenosine triphosphatase associated with diverse cellular activities. Nonetheless, its role in cells of the innate immune system remains largely unexplored. In this study, we report that UBXN6 is upregulated in humans with sepsis and may serve as a pivotal regulator of inflammatory responses via the activation of autophagy. Notably, the upregulation of UBXN6 in sepsis patients was negatively correlated with inflammatory gene profiles but positively correlated with the expression of Forkhead box O3, an autophagy-driving transcription factor. Compared with those of control mice, the macrophages of mice subjected to myeloid cell-specific UBXN6 depletion exhibited exacerbated inflammation, increased mitochondrial oxidative stress, and greater impairment of autophagy and endoplasmic reticulum-associated degradation pathways. UBXN6-deficient macrophages also exhibited immunometabolic remodeling, characterized by a shift to aerobic glycolysis and elevated levels of branched-chain amino acids. These metabolic shifts amplify mammalian target of rapamycin pathway signaling, in turn reducing the nuclear translocation of the transcription factor EB and impairing lysosomal biogenesis. Together, these data reveal that UBXN6 serves as an activator of autophagy and regulates inflammation to maintain immune system suppression during human sepsis.
Collapse
Affiliation(s)
- Young Jae Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Sung-Gwon Lee
- Section of Genetics and Physiology, Laboratory of Molecular and Cellular Biology, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - So Young Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Kangdong Sacred Heart Hospital, Hallym Medical Center, Seoul, 05355, Republic of Korea
| | - Sang Min Jeon
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Soo In Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Kyung Tae Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Cheongju, Chungbuk, 28199, Republic of Korea
| | - Min Joung Lee
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Jinyoung Lee
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyeon Ji Kim
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - So Eui Lee
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Jun Young Heo
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Biochemistry, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - In Soo Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| | - Chungoo Park
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Seungwha Paik
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
10
|
Carolin A, Yan K, Bishop CR, Tang B, Nguyen W, Rawle DJ, Suhrbier A. Tracking inflammation resolution signatures in lungs after SARS-CoV-2 omicron BA.1 infection of K18-hACE2 mice. PLoS One 2024; 19:e0302344. [PMID: 39531435 PMCID: PMC11556745 DOI: 10.1371/journal.pone.0302344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes Coronavirus Disease 2019 (COVID-19), which can result in severe disease, often characterised by a 'cytokine storm' and the associated acute respiratory distress syndrome. However, many infections with SARS-CoV-2 are mild or asymptomatic throughout the course of infection. Although blood biomarkers of severe disease are well studied, less well understood are the inflammatory signatures in lung tissues associated with mild disease or silent infections, wherein infection and inflammation are rapidly resolved leading to sequelae-free recovery. Herein we described RNA-Seq and histological analyses of lungs over time in an omicron BA.1/K18-hACE2 mouse infection model, which displays these latter features. Although robust infection was evident at 2 days post infection (dpi), viral RNA was largely cleared by 10 dpi. Acute inflammatory signatures showed a slightly different pattern of cytokine signatures compared with severe infection models, and where much diminished 30 dpi and absent by 66 dpi. Cellular deconvolution identified significantly increased abundance scores for a number of anti-inflammatory pro-resolution cell types at 5/10 dpi. These included type II innate lymphoid cells, T regulatory cells, and interstitial macrophages. Genes whose expression trended downwards over 2-66 dpi included biomarkers of severe disease and were associated with 'cytokine storm' pathways. Genes whose expression trended upward during this period were associated with recovery of ciliated cells, AT2 to AT1 transition, reticular fibroblasts and innate lymphoid cells, indicating a return to homeostasis. Very few differentially expressed host genes were identified at 66 dpi, suggesting near complete recovery. The parallels between mild or subclinical infections in humans and those observed in this BA.1/K18-hACE2 mouse model are discussed with reference to the concept of "protective inflammation".
Collapse
Affiliation(s)
- Agnes Carolin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kexin Yan
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Cameron R. Bishop
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bing Tang
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Wilson Nguyen
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Daniel J. Rawle
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- GVN Centre of Excellence, Australian Infectious Disease Research Centre, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
Liu Z, Petinrin OO, Chen N, Toseef M, Liu F, Zhu Z, Qi F, Wong KC. Identification and evaluation of candidate COVID-19 critical genes and medicinal drugs related to plasma cells. BMC Infect Dis 2024; 24:1099. [PMID: 39363208 PMCID: PMC11451256 DOI: 10.1186/s12879-024-10000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 09/25/2024] [Indexed: 10/05/2024] Open
Abstract
The ongoing COVID-19 pandemic, caused by the SARS-CoV-2 virus, represents one of the most significant global health crises in recent history. Despite extensive research into the immune mechanisms and therapeutic options for COVID-19, there remains a paucity of studies focusing on plasma cells. In this study, we utilized the DESeq2 package to identify differentially expressed genes (DEGs) between COVID-19 patients and controls using datasets GSE157103 and GSE152641. We employed the xCell algorithm to perform immune infiltration analyses, revealing notably elevated levels of plasma cells in COVID-19 patients compared to healthy individuals. Subsequently, we applied the Weighted Gene Co-expression Network Analysis (WGCNA) algorithm to identify COVID-19 related plasma cell module genes. Further, positive cluster biomarker genes for plasma cells were extracted from single-cell RNA sequencing data (GSE171524), leading to the identification of 122 shared genes implicated in critical biological processes such as cell cycle regulation and viral infection pathways. We constructed a robust protein-protein interaction (PPI) network comprising 89 genes using Cytoscape, and identified 20 hub genes through cytoHubba. These genes were validated in external datasets (GSE152418 and GSE179627). Additionally, we identified three potential small molecules (GSK-1070916, BRD-K89997465, and idarubicin) that target key hub genes in the network, suggesting a novel therapeutic approach. These compounds were characterized by their ability to down-regulate AURKB, KIF11, and TOP2A effectively, as evidenced by their low free binding energies determined through computational analyses using cMAP and AutoDock. This study marks the first comprehensive exploration of plasma cells' role in COVID-19, offering new insights and potential therapeutic targets. It underscores the importance of a systematic approach to understanding and treating COVID-19, expanding the current body of knowledge and providing a foundation for future research.
Collapse
Affiliation(s)
- Zhe Liu
- Institute for Hepatology, The Second Affiliated Hospital, School of Medicine, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518112, China
- Department of Computer Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | | | - Nanjun Chen
- Department of Computer Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Muhammad Toseef
- Department of Computer Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fang Liu
- Rocgene (Beijing) Technology Co., Ltd, Beijing, Beijing, 102200, China
| | - Zhongxu Zhu
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
| | - Furong Qi
- Institute for Hepatology, The Second Affiliated Hospital, School of Medicine, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518112, China.
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China.
| |
Collapse
|
12
|
Mitrofanova L, Korneva L, Makarov I, Bortsova M, Sitnikova M, Ryzhkova D, Kudlay D, Starshinova A. CD68-Negative Histiocytoses with Cardiac Involvement, Associated with COVID-19. Int J Mol Sci 2024; 25:10086. [PMID: 39337571 PMCID: PMC11432257 DOI: 10.3390/ijms251810086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Histiocytoses are rare diseases characterised by infiltration of affected organs by myeloid cells with a monocyte or dendritic cell phenotype. Symptoms can range from self-resolving localised forms to multisystemic lesions requiring specific treatment. To demonstrate extremely rare cases of CD68-negative cardiac histiocytosis with expression of SARS-CoV-2 antigen in infiltrate cells. We demonstrated a case of Erdheim-Chester disease in a 67-year-old man with pericardial involvement and positive dynamics with vemurafenib treatment, an autopsy case of xanthogranulomatous myopericarditis in a 63-year-old man, surgical material of xanthogranulomatous constrictive pericarditis in a 57-year-old man, and an autopsy case of xanthogranulomatosis in a 1-month-old girl. In all cases, xanthogranuloma cells expressed CD163, many of them spike protein SARS-CoV-2, while CD68 expression was detected only in single cells. In this article, we demonstrated four cases of extremely rare CD68-negative cardiac xanthogranulomatosis in three adults and one child with expression of the spike protein SARS-CoV-2 in M2 macrophages. This potential indirect association between COVID-19 and the development of histiocytosis in these patients warrants further investigation. To substantiate this hypothesis, more extensive research is needed.
Collapse
Affiliation(s)
- Lubov Mitrofanova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (L.K.); (M.B.); (M.S.); (D.R.)
| | - Lubov Korneva
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (L.K.); (M.B.); (M.S.); (D.R.)
| | - Igor Makarov
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (L.K.); (M.B.); (M.S.); (D.R.)
| | - Maria Bortsova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (L.K.); (M.B.); (M.S.); (D.R.)
| | - Maria Sitnikova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (L.K.); (M.B.); (M.S.); (D.R.)
| | - Daria Ryzhkova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (L.K.); (M.B.); (M.S.); (D.R.)
| | - Dmitry Kudlay
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia;
- Institute of Immunology, 115478 Moscow, Russia
| | - Anna Starshinova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (L.M.); (L.K.); (M.B.); (M.S.); (D.R.)
- Department of Mathematics and Computer Science, St-Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
13
|
Tolomeo M, Cascio A. STAT4 and STAT6, their role in cellular and humoral immunity and in diverse human diseases. Int Rev Immunol 2024; 43:394-418. [PMID: 39188021 DOI: 10.1080/08830185.2024.2395274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/23/2023] [Accepted: 08/17/2024] [Indexed: 08/28/2024]
Abstract
Signal transducer and activator of transcription (STAT) 4 and STAT6 play a crucial role in immune cells by transducing signals from specific cytokine receptors, and inducing transcription of genes involved in cell-mediated and humoral immunity. These two different defense mechanisms against pathogens are regulated by two specific CD4+ T helper (Th) cells known as Th1 and Th2 cells. Many studies have shown that several diseases including cancer, inflammatory, autoimmune and allergic diseases are associated with a Th1/Th2 imbalance caused by increased or decreased expression/activity of STAT4 or STAT6 often due to genetic and epigenetic aberrances. An altered expression of STAT4 has been observed in different tumors and autoimmune diseases, while a dysregulation of STAT6 signaling pathway is frequently observed in allergic conditions, such as atopic dermatitis, allergic asthma, food allergy, and tumors such as Hodgkin and non-Hodgkin lymphomas. Recently, dysregulations of STAT4 and STAT6 expression have been observed in SARS-CoV2 and monkeypox infections, which are still public health emergencies in many countries. SARS-CoV-2 can induce an imbalance in Th1 and Th2 responses with a predominant activation of STAT6 in the cytosol and nuclei of pneumocytes that drives Th2 polarization and cytokine storm. In monkeypox infection the virus can promote an immune evasion by inducing a Th2 response that in turn inhibits the Th1 response essential for virus elimination. Furthermore, genetic variations of STAT4 that are associated with an increased risk of developing systemic lupus erythematosus seem to play a role in defense against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Manlio Tolomeo
- Department of Infectious Diseases, A.O.U.P. Palermo, Palermo, Italy
| | - Antonio Cascio
- Department of Infectious Diseases, A.O.U.P. Palermo, Palermo, Italy
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, Palermo, Italy
| |
Collapse
|
14
|
Chen J, Zhu X, Wang Z, Rützler M, Lu Q, Xu H, Andersson R, Dai Y, Shen Z, Calamita G, Xie S, Bai Y, Chen B. Inhibition of aquaporin-9 ameliorates severe acute pancreatitis and associated lung injury by NLRP3 and Nrf2/HO-1 pathways. Int Immunopharmacol 2024; 137:112450. [PMID: 38906007 DOI: 10.1016/j.intimp.2024.112450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024]
Abstract
Inflammation, apoptosis and oxidative stress play crucial roles in the deterioration of severe acute pancreatitis-associated acute respiratory distress syndrome (SAP-ARDS). Unfortunately, despite a high mortality rate of 45 %[1], there are limited treatment options available for ARDS outside of last resort options such as mechanical ventilation and extracorporeal support strategies[2]. This study investigated the potential therapeutic role and mechanisms of AQP9 inhibitor RG100204 in two animal models of severe acute pancreatitis, inducing acute respiratory distress syndrome: 1) a sodium-taurocholate induced rat model, and 2) and Cerulein and lipopolysaccharide induced mouse model. RG100204 treatment led to a profound reduction in inflammatory cytokine expression in pancreatic, and lung tissue, in both models. In addition, infiltration of CD68 + and CD11b + cells into these tissues were reduced in RG100204 treated SAP animals, and edema and SAP associated tissue damage were improved. Moreover, we demonstrate that RG100204 reduced apoptosis in the lungs of rat SAP animals, and reduces NF-κB signaling, NLRP3, expression, while profoundly increasing the Nrf2-dependent anti oxidative stress response. We conclude that AQP9 inhibition is a promising strategy for the treatment of pancreatitis and its systemic complications, such as ARDS.
Collapse
Affiliation(s)
- Jiawei Chen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiandong Zhu
- Department of Thyroid Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Ziqiong Wang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Michael Rützler
- ApoGlyx AB, Lund, Sweden, & Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Qiaohong Lu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Hongjie Xu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Roland Andersson
- Department of Surgery, Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Yinwei Dai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Zouwen Shen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Giuseppe Calamita
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Shangjing Xie
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yongheng Bai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| | - Bicheng Chen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
15
|
Chen T, Tan W, Zhan X, Zhou C, Zhu J, Wu S, Qin B, He R, Qin X, Wei W, Huang C, Zhang B, Feng S, Liu C. The shared role of neutrophils in ankylosing spondylitis and ulcerative colitis. Genes Immun 2024; 25:324-335. [PMID: 39060428 DOI: 10.1038/s41435-024-00286-3] [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: 04/27/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
This study aimed to analyze single-cell sequencing data to investigate immune cell interactions in ankylosing spondylitis (AS) and ulcerative colitis (UC). Vertebral bone marrow blood was collected from three AS patients for 10X single-cell sequencing. Analysis of single-cell data revealed distinct cell types in AS and UC patients. Cells significantly co-expressing immune cells (P < 0.05) were subjected to communication analysis. Overlapping genes of these co-expressing immune cells were subjected to GO and KEGG analyses. Key genes were identified using STRING and Cytoscape to assess their correlation with immune cell expression. The results showed the significance of neutrophils in both diseases (P < 0.01), with notable interactions identified through communication analysis. XBP1 emerged as a Hub gene for both diseases, with AUC values of 0.760 for AS and 0.933 for UC. Immunohistochemistry verified that the expression of XBP1 was significantly lower in the AS group and significantly greater in the UC group than in the control group (P < 0.01). This finding highlights the critical role of neutrophils in both AS and UC, suggesting the presence of shared immune response elements. The identification of XBP1 as a potential therapeutic target offers promising intervention avenues for both diseases.
Collapse
Affiliation(s)
- Tianyou Chen
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Weiming Tan
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Xinli Zhan
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Chenxing Zhou
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jichong Zhu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Shaofeng Wu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Boli Qin
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Rongqing He
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Xiaopeng Qin
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Wendi Wei
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Chengqian Huang
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Bin Zhang
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Sitan Feng
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China
| | - Chong Liu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China.
| |
Collapse
|
16
|
Liu Z, Xue X, Geng S, Jiang Z, Ge Z, Zhao C, Xu Y, Wang X, Zhang W, Lin L, Chen Z. The differences in cytokine signatures between severe fever with thrombocytopenia syndrome (SFTS) and hemorrhagic fever with renal syndrome (HFRS). J Virol 2024; 98:e0078624. [PMID: 38916398 PMCID: PMC11265425 DOI: 10.1128/jvi.00786-24] [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: 05/02/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) virus and hantavirus are categorized under the Bunyavirales order. The severe disease progression in both SFTS and hemorrhagic fever with renal syndrome (HFRS) is associated with cytokine storms. This study aimed to explore the differences in cytokine profiles and immune responses between the two diseases. A cross-sectional, single-center study involved 100 participants, comprising 46 SFTS patients, 48 HFRS patients, and 6 healthy controls. The study employed the Luminex cytokine detection platform to measure 48 cytokines. The differences in cytokine profiles and immune characteristics between the two diseases were further analyzed using multiple linear regression, principal component analysis, and random forest method. Among the 48 cytokines tested, 30 showed elevated levels in SFTS and/or HFRS compared to the healthy control group. Furthermore, there were 19 cytokines that exhibited significant differences between SFTS and HFRS. Random forest analysis suggested that TRAIL and CTACK were predictive of SFTS, while IL2Ralpha, MIG, IL-8, IFNalpha2, HGF, SCF, MCP-3, and PDGFBB were more common with HFRS. It was further verified by the receiver operating characteristic with area under the curve >0.8 and P-values <0.05, except for TRAIL. Significant differences were observed in the cytokine profiles of SFTS and HFRS, with TRAIL, IL2Ralpha, MIG, and IL-8 being the top 4 cytokines that most clearly distinguished the two diseases. IMPORTANCE SFTS and HFRS differ in terms of cytokine immune characteristics. TRAIL, IL-2Ralpha, MIG, and IL-8 were the top 4 that differed markedly between SFTS and HFRS.
Collapse
Affiliation(s)
- Zishuai Liu
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyu Xue
- Department of Infectious Disease, Beijing Ditan Hospital, Peking University, Beijing, China
| | - Shuying Geng
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, China
| | - Zhouling Jiang
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ziruo Ge
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Chenxi Zhao
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yanli Xu
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, China
| | - Xiaolei Wang
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ling Lin
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, China
| | - Zhihai Chen
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Infectious Disease, Beijing Ditan Hospital, Peking University, Beijing, China
| |
Collapse
|
17
|
Krishna VD, Chang A, Korthas H, Var SR, Seelig DM, Low WC, Li L, Cheeran MCJ. Impact of age and sex on neuroinflammation following SARS-CoV-2 infection in a murine model. Front Microbiol 2024; 15:1404312. [PMID: 39077737 PMCID: PMC11284165 DOI: 10.3389/fmicb.2024.1404312] [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: 03/20/2024] [Accepted: 06/24/2024] [Indexed: 07/31/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the etiological agent of COVID-19, is known to infect people of all ages and both sexes. Senior populations have the greatest risk of severe COVID-19, and sexual dimorphism in clinical outcomes has been reported. Neurological symptoms are widely observed in COVID-19 patients, with many survivors exhibiting persistent neurological and cognitive impairment. The present study aims to investigate the impact of age and sex on the neuroinflammatory response to SARS-CoV-2 infection using a mouse model. Wild-type C57BL/6J mice were intranasally inoculated with SARS-CoV-2 lineage B.1.351, a variant known to infect mice. Older male mice exhibited a significantly greater weight loss and higher viral loads in the lung at 3 days post infection. Notably, no viral RNA was detected in the brains of infected mice. Nevertheless, expression of IL-6, TNF-α, and CCL-2 in the lung and brain increased with viral infection. RNA-seq transcriptomic analysis of brains showed that SARS-CoV-2 infection caused significant changes in gene expression profiles, implicating innate immunity, defense response to virus, and cerebrovascular and neuronal functions. These findings demonstrate that SARS-CoV-2 infection triggers a neuroinflammatory response, despite the lack of detectable virus in the brain. Aberrant activation of innate immune response, disruption of blood-brain barrier and endothelial cell integrity, and suppression of neuronal activity and axonogenesis underlie the impact of SARS-CoV-2 infection on the brain. Understanding the role of these affected pathways in SARS-CoV-2 pathogenesis helps identify appropriate points of therapeutic interventions to alleviate neurological dysfunction observed during COVID-19.
Collapse
Affiliation(s)
- Venkatramana D. Krishna
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States
| | - Allison Chang
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Holly Korthas
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Susanna R. Var
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Davis M. Seelig
- Comparative Pathology Shared Resource, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Walter C. Low
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Ling Li
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Maxim C. -J. Cheeran
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States
| |
Collapse
|
18
|
Bhuvaneshwar K, Madhavan S, Gusev Y. Integrative genomic analysis of the lung tissue microenvironment in SARS-CoV-2 and NL63 patients. Heliyon 2024; 10:e32772. [PMID: 39183848 PMCID: PMC11341340 DOI: 10.1016/j.heliyon.2024.e32772] [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: 11/17/2023] [Revised: 05/28/2024] [Accepted: 06/09/2024] [Indexed: 08/27/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-CoV-2 virus has affected over 700 million people, and caused over 7 million deaths throughout the world as of April 2024, and continues to affect people through seasonal waves. While over 675 million people have recovered from this disease globally, the lingering effects of the disease are still under study. Long term effects of SARS-CoV-2 infection, known as 'long COVID,' include a wide range of symptoms including fatigue, chest pain, cellular damage, along with a strong innate immune response characterized by inflammatory cytokine production. Three years after the pandemic, data about long covid studies are finally emerging. More clinical studies and clinical trials are needed to understand and determine the factors that predispose individuals to these long-term side effects. In this methodology paper, our goal was to apply data driven approaches in order to explore the multidimensional landscape of infected lung tissue microenvironment to better understand complex interactions between viral infection, immune response and the lung microbiome of patients with (a) SARS-CoV-2 virus and (b) NL63 coronavirus. The samples were analyzed with several machine learning tools allowing simultaneous detection and quantification of viral RNA amount at genome and gene level; human gene expression and fractions of major types of immune cells, as well as metagenomic analysis of bacterial and viral abundance. To contrast and compare specific viral response to SARS-COV-2, we analyzed deep sequencing data from additional cohort of patients infected with NL63 strain of corona virus. Our correlation analysis of three types of RNA-seq based measurements in patients i.e. fraction of viral RNA (at genome and gene level), Human RNA (transcripts and gene level) and bacterial RNA (metagenomic analysis), showed significant correlation between viral load as well as level of specific viral gene expression with the fractions of immune cells present in lung lavage as well as with abundance of major fractions of lung microbiome in COVID-19 patients. Our methodology-based proof-of-concept study has provided novel insights into complex regulatory signaling interactions and correlative patterns between the viral infection, inhibition of innate and adaptive immune response as well as microbiome landscape of the lung tissue. These initial findings could provide better understanding of the diverse dynamics of immune response and the side effects of the SARS-CoV-2 infection and demonstrates the possibilities of the various types of analyses that could be performed from this type of data.
Collapse
Affiliation(s)
- Krithika Bhuvaneshwar
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| | - Subha Madhavan
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| | - Yuriy Gusev
- Georgetown-Innovation Center for Biomedical Informatics (Georgetown-ICBI), Georgetown University Medical Center, Washington DC, 20007, USA
| |
Collapse
|
19
|
Mangiaterra S, Gavazza A, Biagini L, Rossi G. Study of Macrophage Activity in Cats with FIP and Naturally FCoV-Shedding Healthy Cats. Pathogens 2024; 13:437. [PMID: 38921735 PMCID: PMC11206276 DOI: 10.3390/pathogens13060437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 06/27/2024] Open
Abstract
Coronavirus frequently infects humans and animals, showing the ability to recombine and cross over to different species. Cats can be considered a model for studying coronavirus infection, in which feline coronavirus (FCoV) represents a major enteric pathogen related to gastroenteric disease. In this animal, the virus can acquire tropism for macrophage cells, leading to a deadly disease called feline infectious peritonitis (FIP). In this study, monocyte-derived macrophages were isolated by CD14-positive selection in venous whole blood from 26 cats with FIP and 32 FCoV-positive healthy cats. Phagocytosis and respiratory burst activities were investigated and compared between the groups. This is the first study comparing macrophage activity in cats affected by FIP and healthy cats positive for FCoV infection. Our results showed that in cats with FIP, the phagocytic and respiratory burst activities were significantly lower. Our results support the possible role of host immunity in Coronaviridae pathogenesis in cats, supporting future research on the immune defense against this systemic disease.
Collapse
Affiliation(s)
- Sara Mangiaterra
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (A.G.); (L.B.); (G.R.)
| | | | | | | |
Collapse
|
20
|
Feng S, Wang S, Liu C, Wu S, Zhang B, Lu C, Huang C, Chen T, Zhou C, Zhu J, Chen J, Xue J, Wei W, Zhan X. Prediction model for spinal cord injury in spinal tuberculosis patients using multiple machine learning algorithms: a multicentric study. Sci Rep 2024; 14:7691. [PMID: 38565845 PMCID: PMC10987632 DOI: 10.1038/s41598-024-56711-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/09/2024] [Indexed: 04/04/2024] Open
Abstract
Spinal cord injury (SCI) is a prevalent and serious complication among patients with spinal tuberculosis (STB) that can lead to motor and sensory impairment and potentially paraplegia. This research aims to identify factors associated with SCI in STB patients and to develop a clinically significant predictive model. Clinical data from STB patients at a single hospital were collected and divided into training and validation sets. Univariate analysis was employed to screen clinical indicators in the training set. Multiple machine learning (ML) algorithms were utilized to establish predictive models. Model performance was evaluated and compared using receiver operating characteristic (ROC) curves, area under the curve (AUC), calibration curve analysis, decision curve analysis (DCA), and precision-recall (PR) curves. The optimal model was determined, and a prospective cohort from two other hospitals served as a testing set to assess its accuracy. Model interpretation and variable importance ranking were conducted using the DALEX R package. The model was deployed on the web by using the Shiny app. Ten clinical characteristics were utilized for the model. The random forest (RF) model emerged as the optimal choice based on the AUC, PRs, calibration curve analysis, and DCA, achieving a test set AUC of 0.816. Additionally, MONO was identified as the primary predictor of SCI in STB patients through variable importance ranking. The RF predictive model provides an efficient and swift approach for predicting SCI in STB patients.
Collapse
Affiliation(s)
- Sitan Feng
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Shujiang Wang
- Department of Outpatient, General Hospital of Eastern Theater Command, Nanjing, Jiangsu, People's Republic of China
| | - Chong Liu
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Shaofeng Wu
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Bin Zhang
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
- Department of Spine Ward, Bei Jing Ji Shui Tan Hospital Gui Zhou Hospital, Guiyang, Guizhou, People's Republic of China
| | - Chunxian Lu
- Department of Spine and Osteopathy Ward, Bai Se People's Hospital, Baise, Guangxi, People's Republic of China
| | - Chengqian Huang
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Tianyou Chen
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Chenxing Zhou
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jichong Zhu
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jiarui Chen
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jiang Xue
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Wendi Wei
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Xinli Zhan
- Department of Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China.
| |
Collapse
|
21
|
Vajdi M, Karimi A, Hassanizadeh S, Farhangi MA, Bagherniya M, Askari G, Roufogalis BD, Davies NM, Sahebkar A. Effect of polyphenols against complications of COVID-19: current evidence and potential efficacy. Pharmacol Rep 2024; 76:307-327. [PMID: 38498260 DOI: 10.1007/s43440-024-00585-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024]
Abstract
The COVID-19 pandemic that started in 2019 and resulted in significant morbidity and mortality continues to be a significant global health challenge, characterized by inflammation, oxidative stress, and immune system dysfunction.. Developing therapies for preventing or treating COVID-19 remains an important goal for pharmacology and drug development research. Polyphenols are effective against various viral infections and can be extracted and isolated from plants without losing their therapeutic potential. Researchers have developed methods for separating and isolating polyphenols from complex matrices. Polyphenols are effective in treating common viral infections, including COVID-19, and can also boost immunity. Polyphenolic-based antiviral medications can mitigate SARS-CoV-2 enzymes vital to virus replication and infection. Individual polyphenolic triterpenoids, flavonoids, anthraquinonoids, and tannins may also inhibit the SARS-CoV-2 protease. Polyphenol pharmacophore structures identified to date can explain their action and lead to the design of novel anti-COVID-19 compounds. Polyphenol-containing mixtures offer the advantages of a well-recognized safety profile with few known severe side effects. However, studies to date are limited, and further animal studies and randomized controlled trials are needed in future studies. The purpose of this study was to review and present the latest findings on the therapeutic impact of plant-derived polyphenols on COVID-19 infection and its complications. Exploring alternative approaches to traditional therapies could aid in developing novel drugs and remedies against coronavirus infection.
Collapse
Affiliation(s)
- Mahdi Vajdi
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arash Karimi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shirin Hassanizadeh
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Abbasalizad Farhangi
- Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Bagherniya
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Department of Community Nutrition, Food Security Research Center, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Basil D Roufogalis
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2P5, Canada
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
22
|
Yang Q, Barbachano-Guerrero A, Fairchild LM, Rowland TJ, Dowell RD, Allen MA, Warren CJ, Sawyer SL. Macrophages derived from human induced pluripotent stem cells (iPSCs) serve as a high-fidelity cellular model for investigating HIV-1, dengue, and influenza viruses. J Virol 2024; 98:e0156323. [PMID: 38323811 PMCID: PMC10949493 DOI: 10.1128/jvi.01563-23] [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: 10/05/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Macrophages are important target cells for diverse viruses and thus represent a valuable system for studying virus biology. Isolation of primary human macrophages is done by culture of dissociated tissues or from differentiated blood monocytes, but these methods are both time consuming and result in low numbers of recovered macrophages. Here, we explore whether macrophages derived from human induced pluripotent stem cells (iPSCs)-which proliferate indefinitely and potentially provide unlimited starting material-could serve as a faithful model system for studying virus biology. Human iPSC-derived monocytes were differentiated into macrophages and then infected with HIV-1, dengue virus, or influenza virus as model human viruses. We show that iPSC-derived macrophages support the replication of these viruses with kinetics and phenotypes similar to human blood monocyte-derived macrophages. These iPSC-derived macrophages were virtually indistinguishable from human blood monocyte-derived macrophages based on surface marker expression (flow cytometry), transcriptomics (RNA sequencing), and chromatin accessibility profiling. iPSC lines were additionally generated from non-human primate (chimpanzee) fibroblasts. When challenged with dengue virus, human and chimpanzee iPSC-derived macrophages show differential susceptibility to infection, thus providing a valuable resource for studying the species-tropism of viruses. We also show that blood- and iPSC-derived macrophages both restrict influenza virus at a late stage of the virus lifecycle. Collectively, our results substantiate iPSC-derived macrophages as an alternative to blood monocyte-derived macrophages for the study of virus biology. IMPORTANCE Macrophages have complex relationships with viruses: while macrophages aid in the removal of pathogenic viruses from the body, macrophages are also manipulated by some viruses to serve as vessels for viral replication, dissemination, and long-term persistence. Here, we show that iPSC-derived macrophages are an excellent model that can be exploited in virology.
Collapse
Affiliation(s)
- Qing Yang
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | | | - Laurence M. Fairchild
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Teisha J. Rowland
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Robin D. Dowell
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome Boulder Branch, BioFrontiers Institute, Boulder, Colorado, USA
- Department of Computer Science, University of Colorado Boulder, Boulder, Colorado, USA
| | - Mary A. Allen
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Linda Crnic Institute for Down Syndrome Boulder Branch, BioFrontiers Institute, Boulder, Colorado, USA
| | - Cody J. Warren
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, Ohio, USA
- Center for RNA Biology, The Ohio State University, Columbus, Ohio, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, USA
| | - Sara L. Sawyer
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
23
|
Stakišaitis D, Kapočius L, Tatarūnas V, Gečys D, Mickienė A, Tamošuitis T, Ugenskienė R, Vaitkevičius A, Balnytė I, Lesauskaitė V. Effects of Combined Treatment with Sodium Dichloroacetate and Sodium Valproate on the Genes in Inflammation- and Immune-Related Pathways in T Lymphocytes from Patients with SARS-CoV-2 Infection with Pneumonia: Sex-Related Differences. Pharmaceutics 2024; 16:409. [PMID: 38543303 PMCID: PMC10974540 DOI: 10.3390/pharmaceutics16030409] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 12/10/2024] Open
Abstract
The study presents data on the anti-inflammatory effects of a combination of sodium dichloroacetate and sodium valproate (DCA-VPA) on the expression of inflammation- and immune response-related genes in T lymphocytes of SARS-CoV-2 patients. The study aimed to assess the effects of DCA-VPA on the genes of cytokine activity, chemokine-mediated signaling, neutrophil chemotaxis, lymphocyte chemotaxis, T-cell chemotaxis, and regulation of T-cell proliferation pathways. The study included 21 patients with SARS-CoV-2 infection and pneumonia: 9 male patients with a mean age of 68.44 ± 15.32 years and 12 female patients with a mean age of 65.42 ± 15.74 years. They were hospitalized between December 2022 and March 2023. At the time of testing, over 90% of sequences analyzed in Lithuania were found to be of the omicron variant of SARS-CoV-2. The T lymphocytes from patients were treated with 5 mmol DCA and 2 mmol VPA for 24 h in vitro. The effect of the DCA-VPA treatment on gene expression in T lymphocytes was analyzed via gene sequencing. The study shows that DCA-VPA has significant anti-inflammatory effects and apparent sex-related differences. The effect is more potent in T cells from male patients with SARS-CoV-2 infection and pneumonia than in females.
Collapse
Affiliation(s)
- Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (I.B.)
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania
| | - Linas Kapočius
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (I.B.)
| | - Vacis Tatarūnas
- Institute of Cardiology, Laboratory of Molecular Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.T.); (D.G.); (V.L.)
| | - Dovydas Gečys
- Institute of Cardiology, Laboratory of Molecular Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.T.); (D.G.); (V.L.)
| | - Auksė Mickienė
- Department of Infectious Diseases, Lithuanian University of Health Sciences, 47116 Kaunas, Lithuania;
| | - Tomas Tamošuitis
- Department of Intensive Care Medicine, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania;
| | - Rasa Ugenskienė
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania;
| | - Arūnas Vaitkevičius
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University Hospital Santaros Klinikos, Vilnius University, 08661 Vilnius, Lithuania;
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (L.K.); (I.B.)
| | - Vaiva Lesauskaitė
- Institute of Cardiology, Laboratory of Molecular Cardiology, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania; (V.T.); (D.G.); (V.L.)
| |
Collapse
|
24
|
Mahmoodi M, Mohammadi Henjeroei F, Hassanshahi G, Nosratabadi R. Do chemokine/chemokine receptor axes play paramount parts in trafficking and oriented locomotion of monocytes/macrophages toward the lungs of COVID-19 infected patients? A systematic review. Cytokine 2024; 175:156497. [PMID: 38190792 DOI: 10.1016/j.cyto.2023.156497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/19/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024]
Abstract
The COVID-19 (coronavirus disease 2019) is a well-defined viral infection, resulting from SARS-CoV-2 (severe acute respiratory syndrome- coronavirus-2). The innate immune system serves as the first line of defense to limit viral spreading and subsequently stimulate adaptive immune responses by the prominent aids of its cellular and molecular arms. Monocytes are defined as the most prominent innate immune cells (IICs) that are reactive against invading pathogens. These cells support host protection against the virus that is mediated by several non-specific mechanisms such as phagocytosis, producing antiviral enzymes, and recruitment of immune cells toward and into the infected tissues. They have the ability to egress from blood and migrate to the SARS-CoV-2 infected regions by the aid of some defense-related functions like chemotaxis, which is mediated by chemical compounds, e.g., chemokines. Chemokines, in addition to their related ligands are categorized within the most important and deserved agents involved in oriented trafficking of monocytes/macrophages towards and within the lung parenchyma in both steady state and pathological circumstances, including COVID-19-raised infection. However, the overexpression of chemokines could have deleterious effects on various organs through the induction of cytokine storm and may be the most important leading mechanisms in the pathogenesis of COVID-19. Authors have aimed the current review article to describe present knowledge about the interplay between monocytes/macrophages and SARS-CoV-2 with a focus on the ability of IICs to migrate and home into the lung of COVID-19 patients through various chemokine-chemokine receptor axes to promote our understanding regarding this disease.
Collapse
Affiliation(s)
- Merat Mahmoodi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Mohammadi Henjeroei
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, RafsanjanUniversity of Medical Sciences, Rafsanjan, Iran
| | - Reza Nosratabadi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Gastroenterology and Hepatology Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
25
|
Healey AM, Fenner KN, O'Dell CT, Lawrence BP. Aryl hydrocarbon receptor activation alters immune cell populations in the lung and bone marrow during coronavirus infection. Am J Physiol Lung Cell Mol Physiol 2024; 326:L313-L329. [PMID: 38290163 PMCID: PMC11281796 DOI: 10.1152/ajplung.00236.2023] [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: 07/26/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/01/2024] Open
Abstract
Respiratory viral infections are one of the major causes of illness and death worldwide. Symptoms associated with respiratory infections can range from mild to severe, and there is limited understanding of why there is large variation in severity. Environmental exposures are a potential causative factor. The aryl hydrocarbon receptor (AHR) is an environment-sensing molecule expressed in all immune cells. Although there is considerable evidence that AHR signaling influences immune responses to other immune challenges, including respiratory pathogens, less is known about the impact of AHR signaling on immune responses during coronavirus (CoV) infection. In this study, we report that AHR activation significantly altered immune cells in the lungs and bone marrow of mice infected with a mouse CoV. AHR activation transiently reduced the frequency of multiple cells in the mononuclear phagocyte system, including monocytes, interstitial macrophages, and dendritic cells in the lung. In the bone marrow, AHR activation altered myelopoiesis, as evidenced by a reduction in granulocyte-monocyte progenitor cells and an increased frequency of myeloid-biased progenitor cells. Moreover, AHR activation significantly affected multiple stages of the megakaryocyte lineage. Overall, these findings indicate that AHR activation modulates multiple aspects of the immune response to a CoV infection. Given the significant burden of respiratory viruses on human health, understanding how environmental exposures shape immune responses to infection advances our knowledge of factors that contribute to variability in disease severity and provides insight into novel approaches to prevent or treat disease.NEW & NOTEWORTHY Our study reveals a multifaceted role for aryl hydrocarbon receptor (AHR) signaling in the immune response to coronavirus (CoV) infection. Sustained AHR activation during in vivo mouse CoV infection altered the frequency of mature immune cells in the lung and modulated emergency hematopoiesis, specifically myelopoiesis and megakaryopoiesis, in bone marrow. This provides new insight into immunoregulation by the AHR and extends our understanding of how environmental exposures can impact host responses to respiratory viral infections.
Collapse
Affiliation(s)
- Alicia M Healey
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - Kristina N Fenner
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - Colleen T O'Dell
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States
| |
Collapse
|
26
|
Tuo T, Chen D, Wang L, Zhang Y, Zhou L, Ge X, Han J, Guo X, Yang H. Infection of PRRSV inhibits CSFV C-strain replication by inducing macrophages polarization to M1. Vet Microbiol 2024; 289:109957. [PMID: 38160508 DOI: 10.1016/j.vetmic.2023.109957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
It is a common sense that porcine reproductive and respiratory syndrome virus (PRRSV) infection could cause immune failure of classical swine fever (CSF) vaccine, and porcine alveolar macrophages (PAMs) are the target cells of both. To elucidate the role of macrophage polarization in PRRSV infection induced CSF vaccine failure, an immortal porcine alveolar macrophage line PAM39 cell line was used to investigate the effect of PRRSV or/and CSFV C-strain (CSFV-C) infection on macrophage polarization in vitro. Interestingly, PRRSV single infection or PRRSV co-infection with CSFV-C promoted PAM39 cells to M1, while CSFV-C single infection induced PAM39 cells to M2. After the construction of M1 and M2 PAM39 cells polarization models, M1 polarized PAM39 cells were found to inhibit the replication of CSFV-C, and Chinese medicine such as matrine, ginsenosides and astragalus polysaccharides could alleviate the polarization of PAM39 cells and the replication of CSFV-C. Furthermore, interferon (IFN)-γ and lipopolysaccharide (LPS) co-stimulation induced NF-κB activation while matrine treatment blocked M1 polarization-induced NF-κB pathway activation. These findings provided a theoretical basis for designing a new strategy to improve the immune effect of CSFV-C based on porcine alveolar macrophage polarization subtypes.
Collapse
Affiliation(s)
- Tianbei Tuo
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Dengjin Chen
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Lihong Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Yongning Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Lei Zhou
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Xinna Ge
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Jun Han
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| | - Xin Guo
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China.
| | - Hanchun Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, People's Republic of China
| |
Collapse
|
27
|
Henao-Agudelo JS, Ayala S, Badiel M, Zea-Vera AF, Matta Cortes L. Classical monocytes-low expressing HLA-DR is associated with higher mortality rate in SARS-CoV-2+ young patients with severe pneumonia. Heliyon 2024; 10:e24099. [PMID: 38268832 PMCID: PMC10803910 DOI: 10.1016/j.heliyon.2024.e24099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
Aims This study aimed to investigate whether monocyte dysregulation and hyperinflammation serve as predictive markers for mortality in young patients with SARS-CoV-2 severe pneumonia. Methods A prospective cohort study was conducted in a tertiary-level public University Hospital in Colombia. Forty young adults (18-50 years of age) with severe pneumonia and SARS-CoV-2 infection confirmed by qPCR, were enrroled. Serum cytokines and the monocyte phenotype profile, including PDL1/HLA-DR expression, were determined during the first 24 h of hospitalization. Routine laboratory parameters were measured throughout patient follow-up until either death or hospital discharge. We also included a cohort of twenty-five healthy control subjects. Key findings Elevated levels of IL-10, IL-8, and IL-6 cytokines emerged as robust predictors of mortality in young adults with severe pneumonia due to SARS-CoV-2 infected. A descriptive analysis revealed a cumulative mortality rate of 30 % in unvaccinated and ICU-admitted patients. Patients who died had significantly lower expression of HLA-DR on their classical monocytes subsets (CD14+CD16-) than survivors and healthy controls. Lower expression of HLA-DR was associated with greater clinical severity (APACHE≥12) and bacterial coinfection (relative risk 2.5 95%CI [1.18-5.74]). Notably, the expression of HLA-DR in 27.5 % of CD14+/CD16- monocytes was associated with a significantly lower probability of survival. Significance The early reduction in HLA-DR expression within classical monocytes emerged as an independent predictor of mortality, irrespective of comorbidities. Together with PD-L1 expression and cytokine alterations, these findings support the notion that monocyte immunosuppression plays a fundamental role in the pathogenesis and mortality of young patients infected with SARS-CoV-2. These findings hold significant implications for risk assessment and therapeutic strategies in managing critically ill young adults with SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | - Sebastian Ayala
- Department of Internal Medicine, Universidad del Valle, Cali, Colombia
- Hospital Universitario del Valle Evaristo García, Cali, Colombia
| | - Marisol Badiel
- Department of Internal Medicine, Universidad del Valle, Cali, Colombia
- Hospital Universitario del Valle Evaristo García, Cali, Colombia
| | - Andrés F. Zea-Vera
- School of Basic Sciences, Universidad del Valle, Cali, Colombia
- LCIM//Division of Intramural Research, NIAID, National Institutes of Health, Bethesda, MD, USA
| | - Lorena Matta Cortes
- Department of Internal Medicine, Universidad del Valle, Cali, Colombia
- Hospital Universitario del Valle Evaristo García, Cali, Colombia
| |
Collapse
|
28
|
Degenfeld-Schonburg L, Sadovnik I, Smiljkovic D, Peter B, Stefanzl G, Gstoettner C, Jaksch P, Hoetzenecker K, Aigner C, Radtke C, Arock M, Sperr WR, Valent P. Coronavirus Receptor Expression Profiles in Human Mast Cells, Basophils, and Eosinophils. Cells 2024; 13:173. [PMID: 38247864 PMCID: PMC10814915 DOI: 10.3390/cells13020173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
A major problem in SARS-CoV-2-infected patients is the massive tissue inflammation in certain target organs, including the lungs. Mast cells (MC), basophils (BA), and eosinophils (EO) are key effector cells in inflammatory processes. These cells have recently been implicated in the pathogenesis of SARS-CoV-2 infections. We explored coronavirus receptor (CoV-R) expression profiles in primary human MC, BA, and EO, and in related cell lines (HMC-1, ROSA, MCPV-1, KU812, and EOL-1). As determined using flow cytometry, primary MC, BA, and EO, and their corresponding cell lines, displayed the CoV-R CD13 and CD147. Primary skin MC and BA, as well as EOL-1 cells, also displayed CD26, whereas primary EO and the MC and BA cell lines failed to express CD26. As assessed using qPCR, most cell lines expressed transcripts for CD13, CD147, and ABL2, whereas ACE2 mRNA was not detectable, and CD26 mRNA was only identified in EOL-1 cells. We also screened for drug effects on CoV-R expression. However, dexamethasone, vitamin D, and hydroxychloroquine did not exert substantial effects on the expression of CD13, CD26, or CD147 in the cells. Together, MC, BA, and EO express distinct CoV-R profiles. Whether these receptors mediate virus-cell interactions and thereby virus-induced inflammation remains unknown at present.
Collapse
Affiliation(s)
- Lina Degenfeld-Schonburg
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dubravka Smiljkovic
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Barbara Peter
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gabriele Stefanzl
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Clemens Gstoettner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria (C.A.)
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria (C.A.)
| | - Clemens Aigner
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria (C.A.)
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Michel Arock
- Laboratory of Hematology, Pitié-Salpêtrière Hospital, 75651 Paris, France;
| | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
29
|
Zaidi AK, Singh RB, A A Rizvi S, Dehgani-Mobaraki P, Palladino N. COVID-19 pathogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 202:67-112. [PMID: 38237991 DOI: 10.1016/bs.pmbts.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
The pathogenesis of COVID-19 involves a complex interplay between host factors and the SARS-CoV-2 virus, leading to a multitude of clinical manifestations beyond the respiratory system. This chapter provides an overview of the risk factors, genetic predisposition, and multisystem manifestations of COVID-19, shedding light on the underlying mechanisms that contribute to extrapulmonary manifestations. The chapter discusses the direct invasion of SARS-CoV-2 into various organs as well as the indirect mechanisms such as dysregulation of the renin-angiotensin-aldosterone system (RAAS), immune response dysfunctions within the innate and adaptive immune systems, endothelial damage, and immunothrombosis. Furthermore, the multisystem manifestations of COVID-19 across different organ systems, including the cardiovascular, renal, gastrointestinal, hepatobiliary, nervous, endocrine and metabolic, ophthalmic, ear-nose-throat, reproductive, hematopoietic, and immune systems are discussed in detail. Each system exhibits unique manifestations that contribute to the complexity of the disease.
Collapse
Affiliation(s)
| | - Rohan Bir Singh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States; Department of Population, Policy and Practice, Greater Ormond Street Institute of Child Health, University College London, United Kingdom; Discipline of Ophthalmology and Visual Sciences, Adelaide Medical School, University of Adelaide, Australia
| | - Syed A A Rizvi
- College of Biomedical Sciences, Larkin University, Miami, Florida, United States.
| | - Puya Dehgani-Mobaraki
- Founder and President, Associazione Naso Sano, Ringgold Institution ID 567754, San Mariano, Italy.
| | | |
Collapse
|
30
|
Cianciosi D, Diaz YA, Gaddi AV, Capello F, Savo MT, Palí Casanova RDJ, Martínez Espinosa JC, Pascual Barrera AE, Navarro‐Hortal M, Tian L, Bai W, Giampieri F, Battino M. Can alpha‐linolenic acid be a modulator of “cytokine storm,” oxidative stress and immune response in SARS‐CoV‐2 infection? FOOD FRONTIERS 2024; 5:73-93. [DOI: 10.1002/fft2.319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
AbstractAlpha‐linolenic acid (ALA) is a long‐chain polyunsaturated essential fatty acid of the Ω3 series found mainly in vegetables, especially in the fatty part of oilseeds, dried fruit, berries, and legumes. It is very popular for its preventive use in several diseases: It seems to reduce the risk of the onset or decrease some phenomena related to inflammation, oxidative stress, and conditions of dysregulation of the immune response. Recent studies have confirmed these unhealthy situations also in patients with severe coronavirus disease 2019 (COVID‐19). Different findings (in vitro, in vivo, and clinical ones), summarized and analyzed in this review, have showed an important role of ALA in other various non‐COVID physiological and pathological situations against “cytokines storm,” chemokines secretion, oxidative stress, and dysregulation of immune cells that are also involved in the infection of the 2019 novel coronavirus. According to the effects of ALA against all the aforementioned situations (also present in patients with a severe clinical picture of severe acute respiratory syndrome‐(CoV‐2) infection), there may be the biologic plausibility of a prophylactic effect of this compound against COVID‐19 symptoms and fatality.
Collapse
Affiliation(s)
- Danila Cianciosi
- Dipartimento di Scienze Cliniche Specialistiche, Facoltà di Medicina Università Politecnica delle Marche Ancona Italy
| | - Yasmany Armas Diaz
- Dipartimento di Scienze Cliniche Specialistiche, Facoltà di Medicina Università Politecnica delle Marche Ancona Italy
| | | | - Fabio Capello
- International Study Center of Society of Telemedicine and Digital Health Bologna Italy
| | | | - Ramón del Jesús Palí Casanova
- Universidad Europea del Atlántico Santander Spain
- Universidad Internacional Iberoamericana Campeche México México
- Universidad Internacional Iberoamericana Arecibo Puerto Rico USA
| | - Julio César Martínez Espinosa
- Universidad Europea del Atlántico Santander Spain
- Universidad Internacional Iberoamericana Campeche México México
- Fundación Universitaria Internacional de Colombia Bogotá Colombia
| | - Alina Eugenia Pascual Barrera
- Universidad Europea del Atlántico Santander Spain
- Universidad Internacional Iberoamericana Campeche México México
- Fundación Universitaria Internacional de Colombia Bogotá Colombia
| | - Maria‐Dolores Navarro‐Hortal
- Department of Physiology, Institute of Nutrition and Food Technology “José Mataix Verdú”, Biomedical Research Centre University of Granada Armilla Spain
| | - Lingmin Tian
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection Jinan University Guangzhou China
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection Jinan University Guangzhou China
| | - Francesca Giampieri
- Dipartimento di Scienze Cliniche Specialistiche, Facoltà di Medicina Università Politecnica delle Marche Ancona Italy
- Research Group on Food, Nutritional Biochemistry and Health Universidad Europea del Atlántico Santander Spain
| | - Maurizio Battino
- Dipartimento di Scienze Cliniche Specialistiche, Facoltà di Medicina Università Politecnica delle Marche Ancona Italy
- Research Group on Food, Nutritional Biochemistry and Health Universidad Europea del Atlántico Santander Spain
- International Joint Research Laboratory of Intelligent Agriculture and Agri‐Products Processing Jiangsu University Zhenjiang China
| |
Collapse
|
31
|
Spadera L, Lugarà M, Spadera M, Conticelli M, Oliva G, Bassi V, Apuzzi V, Calderaro F, Fattoruso O, Guzzi P, D'Amora M, Catapano O, Marra R, Galdo M, Zappalà M, Inui T, Mette M, Vitiello G, Corvino M, Tortoriello G. Adjunctive use of oral MAF is associated with no disease progression or mortality in hospitalized patients with COVID-19 pneumonia: The single-arm COral-MAF1 prospective trial. Biomed Pharmacother 2023; 169:115894. [PMID: 37988850 DOI: 10.1016/j.biopha.2023.115894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023] Open
Abstract
Based on a growing body of evidence that a dysregulated innate immune response mediated by monocytes/macrophages plays a key role in the pathogenesis of COVID-19, a clinical trial was conducted to investigate the therapeutic potential and safety of oral macrophage activating factor (MAF) plus standard of care (SoC) in the treatment of hospitalized patients with COVID-19 pneumonia. Ninety-seven hospitalized patients with confirmed COVID-19 pneumonia were treated with oral MAF and a vitamin D3 supplement, in combination with SoC, in a single-arm, open label, multicentre, phase II clinical trial. The primary outcome measure was a reduction in an intensive care unit transfer rate below 13% after MAF administration. At the end of the study, an additional propensity score matching (PSM) analysis was performed to compare the MAF group with a control group treated with SoC alone. Out of 97 patients treated with MAF, none needed care in the ICU and/or intubation with mechanical ventilation or died during hospitalization. Oxygen therapy was discontinued after a median of nine days of MAF treatment. The median length of viral shedding and hospital stay was 14 days and 18 days, respectively. After PSM, statistically significant differences were found in all of the in-hospital outcomes between the two groups. No mild to serious adverse events were recorded during the study. Notwithstanding the limitations of a single-arm study, which prevented definitive conclusions, a 21-day course of MAF treatment plus SoC was found to be safe and promising in the treatment of hospitalized adult patients with COVID-19 pneumonia. Further research will be needed to confirm these preliminary findings.
Collapse
Affiliation(s)
- Lucrezia Spadera
- Department of Otolaryngology-Head and Neck Surgery, Ospedale del Mare hospital, ASL Napoli 1 Centro, Naples, Italy.
| | - Marina Lugarà
- Department of General Medicine, Ospedale del Mare hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Maria Spadera
- Department of Anesthesiology and Intensive Care, San Giovanni Bosco hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Mariano Conticelli
- Department of Clinical Pathology, Ospedale del Mare Hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Gabriella Oliva
- Department of General Medicine, Ospedale del Mare hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Vincenzo Bassi
- Department of General Medicine, San Giovanni Bosco Hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Valentina Apuzzi
- Department of General Medicine, San Giovanni Bosco Hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Francesco Calderaro
- Department of General Medicine, San Giovanni Bosco Hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Olimpia Fattoruso
- Department of Clinical Pathology, San Giovanni Bosco Hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Pietro Guzzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Campus Universitario, Germaneto, 88100 Catanzaro, Italy
| | - Maurizio D'Amora
- Department of Laboratory Medicine and Clinical Pathology, San Paolo Hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Oriana Catapano
- Department of Laboratory Medicine and Clinical Pathology, San Paolo Hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Roberta Marra
- Department of Pharmacy, Ospedale del Mare Hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Maria Galdo
- Department of Pharmacy, AORN Ospedali dei Colli Monaldi - Cotugno - C.T.O. Hospital, Naples, Italy
| | - Michele Zappalà
- Department of Medicine, Vesuvio Clinic, ASL Napoli 1 Centro, Naples, Italy
| | - Toshio Inui
- Department of Life System, Institute of Technology and Science, Graduate School, Tokushima University, Tokushima, Japan; Saisei Mirai Cell Processing Center, Osaka, Japan; Saisei Mirai Keihan Clinic, Osaka, Japan; Saisei Mirai Kobe Clinic, Kobe, Japan
| | - Martin Mette
- Saisei Mirai Keihan Clinic, Osaka, Japan; Saisei Mirai Kobe Clinic, Kobe, Japan
| | - Giuseppe Vitiello
- Department of Health Management, Ospedale del Mare hospital, ASL Napoli 1 Centro, Naples, Italy
| | - Maria Corvino
- Department of Health Management, ASL Napoli 1 Centro, Naples, Italy
| | - Giuseppe Tortoriello
- Department of Otolaryngology-Head and Neck Surgery, AORN Ospedali dei Colli Monaldi - Cotugno - C.T.O. Hospital, Naples, Italy
| |
Collapse
|
32
|
Zhang J, Askenase P, Jaenisch R, Crumpacker CS. Approaches to pandemic prevention - the chromatin vaccine. Front Immunol 2023; 14:1324084. [PMID: 38143744 PMCID: PMC10739501 DOI: 10.3389/fimmu.2023.1324084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/13/2023] [Indexed: 12/26/2023] Open
Abstract
Developing effective vaccines against viral infections have significant impacts on development, prosperity and well-being of human populations. Thus, successful vaccines such as smallpox and polio vaccines, have promoted global societal well-being. In contrast, ineffective vaccines may fuel arguments that retard scientific progress. We aim to stimulate a multilevel discussion on how to develop effective vaccines against recent and future pandemics by focusing on acquired immunodeficiency syndrome (AIDS), coronavirus disease (COVID) and other viral infections. We appeal to harnessing recent achievements in this field specifically towards a cure for current pandemics and prevention of the next pandemics. Among these, we propose to apply the HIV DNA in chromatin format - an end product of aborted HIV integration in episomal forms, i.e., the chromatin vaccines (cVacc), to elicit the epigenetic silencing and memory that prevent viral replication and infection.
Collapse
Affiliation(s)
- Jielin Zhang
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Philip Askenase
- Allergy & Clinical Immunology, Yale School of Medicine, New Haven, CT, United States
| | - Rudolf Jaenisch
- Department of Biology, Whitehead Institute, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Clyde S. Crumpacker
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
Zelaya H, Arellano-Arriagada L, Fukuyama K, Matsumoto K, Marranzino G, Namai F, Salva S, Alvarez S, Agüero G, Kitazawa H, Villena J. Lacticaseibacillus rhamnosus CRL1505 Peptidoglycan Modulates the Inflammation-Coagulation Response Triggered by Poly(I:C) in the Respiratory Tract. Int J Mol Sci 2023; 24:16907. [PMID: 38069229 PMCID: PMC10707514 DOI: 10.3390/ijms242316907] [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: 10/27/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Lacticaseibacillus rhamnosus CRL1505 beneficially modulates the inflammation-coagulation response during respiratory viral infections. This study evaluated the capacity of the peptidoglycan obtained from the CRL1505 strain (PG-Lr1505) to modulate the immuno-coagulative response triggered by the viral pathogen-associated molecular pattern poly(I:C) in the respiratory tract. Adult BALB/c mice were nasally treated with PG-Lr1505 for two days. Treated and untreated control mice were then nasally challenged with poly(I:C). Mice received three doses of poly(I:C) with a 24 h rest period between each administration. The immuno-coagulative response was studied after the last administration of poly(I:C). The challenge with poly(I:C) significantly increased blood and respiratory pro-inflammatory mediators, decreased prothrombin activity (PT), and increased von Willebrand factor (vWF) levels in plasma. Furthermore, tissue factor (TF), tissue factor pathway inhibitor (TFPI), and thrombomodulin (TM) expressions were increased in the lungs. PG-Lr1505-treated mice showed significant modulation of hemostatic parameters in plasma (PT in %, Control = 71.3 ± 3.8, PG-Lr1505 = 94.0 ± 4.0, p < 0.01) and lungs. Moreover, PG-Lr1505-treated mice demonstrated reduced TF in F4/80 cells from lungs, higher pro-inflammatory mediators, and increased IL-10 compared to poly(I:C) control mice (IL-10 in pg/mL, Control = 379.1 ± 12.1, PG-Lr1505 = 483.9 ± 11.3, p < 0.0001). These changes induced by PG-Lr1505 correlated with a significant reduction in lung tissue damage. Complementary in vitro studies using Raw 264.7 cells confirmed the beneficial effect of PG-Lr1505 on poly(I:C)-induced inflammation, since increased IL-10 expression, as well as reduced damage, production of inflammatory mediators, and hemostatic parameter expressions were observed. In addition, protease-activated receptor-1 (PAR1) activation in lungs and Raw 264.7 cells was observed after TLR3 stimulation, which was differentially modulated by PG-Lr1505. The peptidoglycan from L. rhamnosus CRL1505 is able to regulate inflammation, the procoagulant state, and PAR1 activation in mice and macrophages in the context of the activation of TLR3 signaling pathways, contributing to a beneficial modulation of inflammation-hemostasis crosstalk.
Collapse
Affiliation(s)
- Hortensia Zelaya
- Institute of Applied Biochemistry, Tucuman University, Tucuman 4000, Argentina; (H.Z.); (S.A.); (G.A.)
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
| | - Luciano Arellano-Arriagada
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
| | - Kohtaro Fukuyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
| | - Kaho Matsumoto
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
| | - Gabriela Marranzino
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
- Facultad de Ciencias de la Salud, Universidad del Norte Santo Tomás de Aquino (UNSTA), Tucuman 4000, Argentina
| | - Fu Namai
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan
| | - Susana Salva
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
| | - Susana Alvarez
- Institute of Applied Biochemistry, Tucuman University, Tucuman 4000, Argentina; (H.Z.); (S.A.); (G.A.)
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
| | - Graciela Agüero
- Institute of Applied Biochemistry, Tucuman University, Tucuman 4000, Argentina; (H.Z.); (S.A.); (G.A.)
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina; (L.A.-A.); (G.M.); (S.S.)
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8576, Japan; (K.F.); (K.M.); (F.N.)
| |
Collapse
|
34
|
Felkle D, Zięba K, Kaleta K, Czaja J, Zyzdorf A, Sobocińska W, Jarczyński M, Bryniarski K, Nazimek K. Overreactive macrophages in SARS-CoV-2 infection: The effects of ACEI. Int Immunopharmacol 2023; 124:110858. [PMID: 37708705 DOI: 10.1016/j.intimp.2023.110858] [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: 06/02/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
Among various factors influencing the course of SARS-CoV-2 infection in humans, macrophage overactivation is considered the main cause of the cytokine storm that leads to severe complications of COVID-19. Moreover, the increased expression of angiotensin converting enzyme 2 (ACE2), an obligatory entry receptor of the coronavirus, caused by treatment with ACE inhibitors (ACEI) lowered overall confidence in the safety of these drugs. However, analysis of the course of coronavirus infection in patients treated with ACEI does not support these concerns. Instead, the beneficial effect of ACEI on macrophages has increasingly been emphasized. This includes their anti-inflammatory activation and the consequent reduction in the risk of severe disease and life-threatening complications. Herein, we summarize the current knowledge and understanding of the dual role of macrophages in SARS-CoV-2 infection, with a special focus on the postulated mechanisms underlying the beneficial effects of macrophage targeting by ACEI. These seem to involve the stimulation of macrophage angiotensin II type 2 and Mas receptors by angiotensin 1-7, intensively produced due to the up-regulation of ACE2 expression on macrophages, as well as the direct inhibition of macrophage hyper-responsiveness by ACEI. The impact of ACEI on macrophages may also lead to the activation of an effective antiviral response due to the increased expression of ACE2.
Collapse
Affiliation(s)
- Dominik Felkle
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Katarzyna Zięba
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Konrad Kaleta
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Julia Czaja
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Amanda Zyzdorf
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Wiktoria Sobocińska
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Mateusz Jarczyński
- Students' Scientific Group at the Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland
| | - Katarzyna Nazimek
- Department of Immunology, Jagiellonian University Medical College, Czysta 18, 31-121 Kraków, Poland.
| |
Collapse
|
35
|
Asghari F, Asghary A, Majidi Zolbanin N, Faraji F, Jafari R. Immunosenescence and Inflammaging in COVID-19. Viral Immunol 2023; 36:579-592. [PMID: 37797216 DOI: 10.1089/vim.2023.0045] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023] Open
Abstract
Despite knowledge gaps in understanding the full spectrum of the hyperinflammatory phase caused by SARS-CoV-2, according to the World Health Organization (WHO), COVID-19 is still the leading cause of death worldwide. Susceptible people to severe COVID-19 are those with underlying medical conditions or those with dysregulated and senescence-associated immune responses. As the immune system undergoes aging in the elderly, such drastic changes predispose them to various diseases and affect their responsiveness to infections, as seen in COVID-19. At-risk groups experience poor prognosis in terms of disease recovery. Changes in the quantity and quality of immune cell function have been described in numerous literature sites. Impaired immune cell function along with age-related metabolic changes can lead to features such as hyperinflammatory response, immunosenescence, and inflammaging in COVID-19. Inflammaging is related to the increased activity of the most inflammatory factors and is the main cause of age-related diseases and tissue failure in the elderly. Since hyperinflammation is a common feature of most severe cases of COVID-19, this pathway, which is not fully understood, leads to immunosenescence and inflammaging in some individuals, especially in the elderly and those with comorbidities. In this review, we shed some light on the age-related abnormalities of innate and adaptive immune cells and how hyperinflammatory immune responses contribute to the inflammaging process, leading to clinical deterioration. Further, we provide insights into immunomodulation-based therapeutic approaches, which are potentially important considerations in vaccine design for elderly populations.
Collapse
Affiliation(s)
- Faezeh Asghari
- Department of Immunology, School of Medicine, Tarbiat Modares University, Tehran, Iran
| | - Amir Asghary
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Naime Majidi Zolbanin
- Experimental and Applied Pharmaceutical Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
36
|
Liu Z, Zhang J, Li S, Wang H, Ren B, Li J, Bao Z, Liu J, Guo M, Yang G, Chen L. Circadian control of ConA-induced acute liver injury and inflammatory response via Bmal1 regulation of Junb. JHEP Rep 2023; 5:100856. [PMID: 37791375 PMCID: PMC10542646 DOI: 10.1016/j.jhepr.2023.100856] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 10/05/2023] Open
Abstract
Background & Aims Circadian rhythms play significant roles in immune responses, and many inflammatory processes in liver diseases are associated with malfunctioning molecular clocks. However, the significance of the circadian clock in autoimmune hepatitis (AIH), which is characterised by immune-mediated hepatocyte destruction and extensive inflammatory cytokine production, remains unclear. Methods We tested the difference in susceptibility to the immune-mediated liver injury induced by concanavalin A (ConA) at various time points throughout a day in mice and analysed the effects of global, hepatocyte, or myeloid cell deletion of the core clock gene, Bmal1 (basic helix-loop-helix ARNT-like 1), on liver injury and inflammatory responses. Multiple molecular biology techniques and mice with macrophage-specific knockdown of Junb, a Bmal1 target gene, were used to investigate the involvement of Junb in the circadian control of ConA-induced hepatitis. Results The susceptibility to ConA-induced liver injury is highly dependent on the timing of ConA injection. The treatment at Zeitgeber time 0 (lights on) triggers the highest mortality as well as the severest liver injury and inflammatory responses. Further study revealed that this timing effect was driven by macrophage, but not hepatocyte, Bmal1. Mechanistically, Bmal1 controls the diurnal variation of ConA-induced hepatitis by directly regulating the circadian transcription of Junb and promoting M1 macrophage activation. Inhibition of Junb in macrophages blunts the administration time-dependent effect of ConA and attenuates liver injury. Moreover, we demonstrated that Junb promotes macrophage inflammation by regulating AKT and extracellular signal-regulated kinase (ERK) signalling pathways. Conclusions Our findings uncover a critical role of the Bmal1-Junb-AKT/ERK axis in the circadian control of ConA-induced hepatitis and provide new insights into the prevention and treatment of AIH. Impact and Implications This study unveils a critical role of the Bmal1-Junb-AKT/ERK axis in the circadian control of ConA-induced liver injury, providing new insights into the prevention and treatment of immune-mediated hepatitis, including autoimmune hepatitis (AIH). The findings have scientific implications as they enhance our understanding of the circadian regulation of immune responses in liver diseases. Furthermore, clinically, this research offers opportunities for optimising treatment strategies in immune-mediated hepatitis by considering the timing of therapeutic interventions.
Collapse
Affiliation(s)
- Zhaiyi Liu
- School of Bioengineering, Dalian University of Technology, Dalian, China
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Jiayang Zhang
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, China
| | - Shuyao Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hui Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Baoyin Ren
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Jiazhi Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhiyue Bao
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Jiaxin Liu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Meina Guo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Guangrui Yang
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lihong Chen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
37
|
Pereira VIC, de Brito Junior LC, Falcão LFM, da Costa Vasconcelos PF, Quaresma JAS, Berg AVVD, Paixão APS, Ferreira RIS, Diks IBC. Monocytes subpopulations pattern in the acute respiratory syndrome coronavirus 2 virus infection and after long COVID-19. Int Immunopharmacol 2023; 124:110994. [PMID: 37804653 DOI: 10.1016/j.intimp.2023.110994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/09/2023]
Abstract
INTRODUTION AND OBJECTIVE The present study sought to characterize the pattern of monocyte subpopulations in patients during the course of the infections caused by SARS-CoV-2 virus or who presented long COVID-19 syndrome compared to monocytes from patients with zika virus (Zika) or chikungunya virus (CHIKV). CASUISTRY Study with 89 peripheral blood samples from patients, who underwent hemogram and serology (IgG and IgM) for detection of Zika (Control Group 1, n = 18) or CHIKV (Control Group 2, n = 9), and from patients who underwent hemogram and reverse transcription polymerase chain reaction for detection of SARS-CoV-2 at the acute phase of the disease (Group 3, n = 19); and of patients who presented long COVID-19 syndrome (Group 4, n = 43). The monocyte and subpopulations counts were performed by flow cytometry. RESULTS No significant difference was observed in the total number of monocytes between the groups. The classical (CD14++CD16-) and intermediate (CD14+CD16+) monocytes counts were increased in patients with acute infection or with long COVID-19 syndrome. The monocytes subpopulations counts were lower in patients with infection Zika or CHIKV. CONCLUSION Increase in the monocyte subpopulations in patients with acute infection or with long COVID-19 syndrome may be an important finding of differentiated from the infection Zika or CHIKV.
Collapse
Affiliation(s)
| | - Lacy Cardoso de Brito Junior
- Institute of Biological Sciences at UFPA. Laboratory of General Pathology - Immunopathology and Cytology at FederalUniversity of Pará. Belém, Pará, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Negi V, Gavlock D, Miedel MT, Lee JK, Shun T, Gough A, Vernetti L, Stern AM, Taylor DL, Yechoor VK. Modeling mechanisms underlying differential inflammatory responses to COVID-19 in type 2 diabetes using a patient-derived microphysiological organ-on-a-chip system. LAB ON A CHIP 2023; 23:4514-4527. [PMID: 37766577 DOI: 10.1039/d3lc00285c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Background: COVID-19 pandemic has caused more than 6 million deaths worldwide. Co-morbid conditions such as Type 2 Diabetes (T2D) have increased mortality in COVID-19. With limited translatability of in vitro and small animal models to human disease, human organ-on-a-chip models are an attractive platform to model in vivo disease conditions and test potential therapeutics. Methods: T2D or non-diabetic patient-derived macrophages and human liver sinusoidal endothelial cells were seeded, along with normal hepatocytes and stellate cells in the liver-on-a-chip (LAMPS - liver acinus micro physiological system), perfused with media mimicking non-diabetic fasting or T2D (high levels of glucose, fatty acids, insulin, glucagon) states. The macrophages and endothelial cells were transduced to overexpress the SARS-CoV2-S (spike) protein with appropriate controls before their incorporation into LAMPS. Cytokine concentrations in the efflux served as a read-out of the effects of S-protein expression in the different experimental conditions (non-diabetic-LAMPS, T2D-LAMPS), including incubation with tocilizumab, an FDA-approved drug for severe COVID-19. Findings: S-protein expression in the non-diabetic LAMPS led to increased cytokines, but in the T2D-LAMPS, this was significantly amplified both in the number and magnitude of key pro-inflammatory cytokines (IL6, CCL3, IL1β, IL2, TNFα, etc.) involved in cytokine storm syndrome (CSS), mimicking severe COVID-19 infection in T2D patients. Compared to vehicle control, tocilizumab (IL6-receptor antagonist) decreased the pro-inflammatory cytokine secretion in T2D-COVID-19-LAMPS but not in non-diabetic-COVID-19-LAMPS. Interpretation: macrophages and endothelial cells play a synergistic role in the pathophysiology of the hyper-inflammatory response seen with COVID-19 and T2D. The effect of Tocilizumab was consistent with large clinical trials that demonstrated Tocilizumab's efficacy only in critically ill patients with severe disease, providing confirmatory evidence that the T2D-COVID-19-LAMPS is a robust platform to model human in vivo pathophysiology of COVID-19 in T2D and for screening potential therapeutics.
Collapse
Affiliation(s)
- Vinny Negi
- Diabetes and Beta Cell Biology Center, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Dillon Gavlock
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark T Miedel
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeong Kyung Lee
- Diabetes and Beta Cell Biology Center, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Tongying Shun
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Albert Gough
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence Vernetti
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew M Stern
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - D Lansing Taylor
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vijay K Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
39
|
Tascini C, Cotrufo M, Sozio E, Fanin M, Dellai F, Zanus Forte A, Cesselli D, DE Stefanis P, Ripoli A, Sbrana F, Giuliano S, Fabris M, Girardis M, Curcio F, Bassi F. Potential role of IgM-enriched immunoglobulin as adjuvant treatment in severe SARS-CoV-2 infection. Minerva Anestesiol 2023; 89:884-894. [PMID: 37822148 DOI: 10.23736/s0375-9393.23.17244-0] [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: 10/13/2023]
Abstract
BACKGROUND Severe COVID-19 patients are characterized by a dysregulated host response to an infection, with uncontrolled pro- and anti- inflammatory pathway activation. Consistent proportion of patients require admission in intensive care units and are at risk of progression to severe forms of disease. These patients are generally admitted during later stages of the disease, when effective antiviral and monoclonal antibody are not indicated. We aimed to assess the potential role of IgM-enriched intra venous immunoglobulins (IGAM) preparations in this setting. METHODS This retrospective, observational case-controlled study was conducted at a single-center University Hospital of Udine in the Friuli Venezia Giulia Region of Italy. Patients referring to the center between March 2020 and April 2021 was included. During the study period, patient who received Pentaglobin® IGAM treatment (N.=56), administered as compassionate use, was compared with a control group (N.=169) to assess, by propensity score analysis, clinical outcome. RESULTS Untreated controls required, respect to patient treated with IGAM therapy, longer time to hospitalization with no significant differences in death and orotracheal intubation requirement. Significant differences in the two cohort were in: SOFA was higher in treated, while D-dimer and P/F ratio was better in the treatment cohort. Multivariate logistic regression analysis performed on the "matched sample," obtained by a weighting propensity score approach, identify, as significant protective factor for death outcome, the Pentaglobin® treatment (0.820 [0.698-0.963], P=0.016) and low C-reactive protein (1.001 [1.000-1.002], P=0.031) value while the delay of onset hospitalization is associate with a worst outcome (0.983 [0.967-0.999], P=0.041). CONCLUSIONS The present study offers a significant insight concerning the use of IgM-enriched immunoglobulin preparations in patients with SARS-CoV-2 severe infection and also could identifying the specific immunological and biochemical profile of the patient who can more benefit from this therapeutic option.
Collapse
Affiliation(s)
- Carlo Tascini
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy -
- Department of Medical Area (DAME), University of Udine, Udine, Italy -
| | - Marco Cotrufo
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Emanuela Sozio
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Matteo Fanin
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Fabiana Dellai
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Agnese Zanus Forte
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Daniela Cesselli
- Institute of Clinical Pathology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Paola DE Stefanis
- Section of Anesthesia and Resuscitation2, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Andrea Ripoli
- Department of Bioengineering, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Francesco Sbrana
- Lipoapheresis Unit, Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Simone Giuliano
- Infectious Diseases Clinic, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| | - Martina Fabris
- Institute of Clinical Pathology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Massimo Girardis
- Department of Anesthesia and Intensive Care, University Hospital of Modena, Modena, Italy
| | - Francesco Curcio
- Institute of Clinical Pathology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Flavio Bassi
- Section of Anesthesia and Resuscitation2, Azienda Sanitaria Universitaria del Friuli Centrale (ASUFC), Udine, Italy
| |
Collapse
|
40
|
Luan Y, Luan Y, He H, Jue B, Yang Y, Qin B, Ren K. Glucose metabolism disorder: a potential accomplice of SARS-CoV-2. Int J Obes (Lond) 2023; 47:893-902. [PMID: 37542197 DOI: 10.1038/s41366-023-01352-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/06/2023]
Abstract
Globally, 265,713,467 confirmed cases of SARS-CoV-2 (CoV-2), including 5,260,888 deaths, have been reported by the WHO. It is important to study the mechanism of this infectious disease. A variety of evidences show the potential association between CoV-2 and glucose metabolism. Notably, people with type 2 diabetes mellitus (T2DM) and other metabolic complications were prone to have a higher risk of developing a more severe infection course than people who were metabolically normal. The correlations between glucose metabolism and CoV-2 progression have been widely revealed. This review will discuss the association between glucose metabolism disorders and CoV-2 progression, showing the promoting effect of diabetes and other diseases related to glucose metabolism disorders on the progression of CoV-2. We will further conclude the effects of key proteins and pathways in glucose metabolism regulation on CoV-2 progression and potential interventions by targeting glucose metabolism disorders for CoV-2 treatment. Therefore, this review will provide systematic insight into the treatment of CoV-2 from the perspective of glucose metabolism.
Collapse
Affiliation(s)
- Yi Luan
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100000, China
| | - Hongbo He
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Bolin Jue
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453000, China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Bo Qin
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
41
|
Mazzocco YL, Bergero G, Del Rosso S, Eberhardt N, Sola C, Saka HA, Villada SM, Bocco JL, Aoki MP. Differential expression patterns of purinergic ectoenzymes and the antioxidative role of IL-6 in hospitalized COVID-19 patient recovery. Front Immunol 2023; 14:1227873. [PMID: 37818368 PMCID: PMC10560791 DOI: 10.3389/fimmu.2023.1227873] [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: 05/23/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023] Open
Abstract
Introduction We have acquired significant knowledge regarding the pathogenesis of severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2). However, the underlying mechanisms responsible for disease recovery still need to be fully understood. Methods To gain insights into critical immune markers involved in COVID-19 etiopathogenesis, we studied the evolution of the immune profile of peripheral blood samples from patients who had recovered from COVID-19 and compared them to subjects with severe acute respiratory illness but negative for SARS-CoV-2 detection (controls). In addition, linear and clustered correlations between different parameters were determined. Results The data obtained revealed a significant reduction in the frequency of inflammatory monocytes (CD14+CD16+) at hospital discharge vs. admission. Remarkably, nitric oxide (NO) production by the monocyte compartment was significantly reduced at discharge. Furthermore, interleukin (IL)-6 plasma levels were negatively correlated with the frequency of NO+CD14+CD16+ monocytes at hospital admission. However, at the time of hospital release, circulating IL-6 directly correlated with the NO production rate by monocytes. In line with these observations, we found that concomitant with NO diminution, the level of nitrotyrosine (NT) on CD8 T-cells significantly diminished at the time of hospital release. Considering that purinergic signaling constitutes another regulatory system, we analyzed the kinetics of CD39 and CD73 ectoenzyme expression in CD8 T-cells. We found that the frequency of CD39+CD8+ T-cells significantly diminished while the percentage of CD73+ cells increased at hospital discharge. In vitro, IL-6 stimulation of PBMCs from COVID-19 patients diminished the NT levels on CD8 T-cells. A clear differential expression pattern of CD39 and CD73 was observed in the NT+ vs. NT-CD8+ T-cell populations. Discussion The results suggest that early after infection, IL-6 controls the production of NO, which regulates the levels of NT on CD8 T-cells modifying their effector functions. Intriguingly, in this cytotoxic cell population, the expression of purinergic ectoenzymes is tightly associated with the presence of nitrated surface molecules. Overall, the data obtained contribute to a better understanding of pathogenic mechanisms associated with COVID-19 outcomes.
Collapse
Affiliation(s)
- Yanina Luciana Mazzocco
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Córdoba, Argentina
| | - Gastón Bergero
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Córdoba, Argentina
| | - Sebastian Del Rosso
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Córdoba, Argentina
| | - Natalia Eberhardt
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Córdoba, Argentina
| | - Claudia Sola
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Córdoba, Argentina
| | - Héctor Alex Saka
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Córdoba, Argentina
| | - Sofía María Villada
- Servicio de Enfermedades Infecciosas, Hospital Privado Universitario de Córdoba, Córdoba, Argentina
| | - José Luis Bocco
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Córdoba, Argentina
| | - Maria Pilar Aoki
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Córdoba, Argentina
| |
Collapse
|
42
|
Bergersen KV, Pham K, Li J, Ulrich MT, Merrill P, He Y, Alaama S, Qiu X, Harahap-Carrillo IS, Ichii K, Frost S, Kaul M, Godzik A, Heinrich EC, Nair MG. Health disparities in COVID-19: immune and vascular changes are linked to disease severity and persist in a high-risk population in Riverside County, California. BMC Public Health 2023; 23:1584. [PMID: 37598150 PMCID: PMC10439554 DOI: 10.1186/s12889-023-16462-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/05/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Health disparities in underserved communities, such as inadequate healthcare access, impact COVID-19 disease outcomes. These disparities are evident in Hispanic populations nationwide, with disproportionately high infection and mortality rates. Furthermore, infected individuals can develop long COVID with sustained impacts on quality of life. The goal of this study was to identify immune and endothelial factors that are associated with COVID-19 outcomes in Riverside County, a high-risk and predominantly Hispanic community, and investigate the long-term impacts of COVID-19 infection. METHODS 112 participants in Riverside County, California, were recruited according to the following criteria: healthy control (n = 23), outpatients with moderate infection (outpatient, n = 33), ICU patients with severe infection (hospitalized, n = 33), and individuals recovered from moderate infection (n = 23). Differences in outcomes between Hispanic and non-Hispanic individuals and presence/absence of co-morbidities were evaluated. Circulating immune and vascular biomarkers were measured by ELISA, multiplex analyte assays, and flow cytometry. Follow-up assessments for long COVID, lung health, and immune and vascular changes were conducted after recovery (n = 23) including paired analyses of the same participants. RESULTS Compared to uninfected controls, the severe infection group had a higher proportion of Hispanic individuals (n = 23, p = 0.012) than moderate infection (n = 8, p = 0.550). Disease severity was associated with changes in innate monocytes and neutrophils, lymphopenia, disrupted cytokine production (increased IL-8 and IP-10/CXCL10 but reduced IFNλ2/3 and IFNγ), and increased endothelial injury (myoglobin, VCAM-1). In the severe infection group, a machine learning model identified LCN2/NGAL, IL-6, and monocyte activation as parameters associated with fatality while anti-coagulant therapy was associated with survival. Recovery from moderate COVID infection resulted in long-term immune changes including increased monocytes/lymphocytes and decreased neutrophils and endothelial markers. This group had a lower proportion of co-morbidities (n = 8, p = 1.0) but still reported symptoms associated with long COVID despite recovered pulmonary function. CONCLUSION This study indicates increased severity of COVID-19 infection in Hispanic individuals of Riverside County, California. Infection resulted in immunological and vascular changes and long COVID symptoms that were sustained for up to 11 months, however, lung volume and airflow resistance was recovered. Given the immune and behavioral impacts of long COVID, the potential for increased susceptibility to infections and decreased quality of life in high-risk populations warrants further investigation.
Collapse
Affiliation(s)
- Kristina V Bergersen
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Kathy Pham
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Jiang Li
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Michael T Ulrich
- Riverside University Health System Medical Center, Riverside, CA, U.S
| | - Patrick Merrill
- Kaiser Permanente Riverside Medical Center, Riverside, CA, U.S
| | - Yuxin He
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Sumaya Alaama
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Xinru Qiu
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Indira S Harahap-Carrillo
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Keita Ichii
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Shyleen Frost
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Marcus Kaul
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Adam Godzik
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S
| | - Erica C Heinrich
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S..
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, U.S..
| |
Collapse
|
43
|
Aghamohamadi N, Shahba F, Zarezadeh Mehrabadi A, Khorramdelazad H, Karimi M, Falak R, Emameh RZ. Age-dependent immune responses in COVID-19-mediated liver injury: focus on cytokines. Front Endocrinol (Lausanne) 2023; 14:1139692. [PMID: 37654571 PMCID: PMC10465349 DOI: 10.3389/fendo.2023.1139692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/21/2023] [Indexed: 09/02/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is potentially pathogenic and causes severe symptoms; in addition to respiratory syndromes, patients might experience other severe conditions such as digestive complications and liver complications injury. The abnormality in the liver is manifested by hepatobiliary dysfunction and enzymatic elevation, which is associated with morbidity and mortality. The direct cytopathic effect, immune dysfunction, cytokine storm, and adverse effects of therapeutic regimens have a crucial role in the severity of liver injury. According to aging and immune system alterations, cytokine patterns may also change in the elderly. Moreover, hyperproduction of cytokines in the inflammatory response to SARS-CoV-2 can lead to multi-organ dysfunction. The mortality rate in elderly patients, particularly those with other comorbidities, is also higher than in adults. Although the pathogenic effect of SARS-CoV-2 on the liver has been widely studied, the impact of age and immune-mediated responses at different ages remain unclear. This review discusses the association between immune system responses in coronavirus disease 2019 (COVID-19) patients of different ages and liver injury, focusing on cytokine alterations.
Collapse
Affiliation(s)
- Nazanin Aghamohamadi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Shahba
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zarezadeh Mehrabadi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Milad Karimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Zolfaghari Emameh
- Department of Energy and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
44
|
Krishna VD, Chang A, Korthas H, Var SR, Low WC, Li L, Cheeran MCJ. Impact of age and sex on neuroinflammation following SARS-CoV-2 infection in a murine model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.11.552998. [PMID: 37645925 PMCID: PMC10462071 DOI: 10.1101/2023.08.11.552998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the etiological agent for the worldwide COVID-19 pandemic, is known to infect people of all ages and both sexes. Senior populations have the greatest risk of severe disease, and sexual dimorphism in clinical outcomes has been reported in COVID-19. SARS-CoV-2 infection in humans can cause damage to multiple organ systems, including the brain. Neurological symptoms are widely observed in patients with COVID-19, with many survivors suffering from persistent neurological and cognitive impairment, potentially accelerating Alzheimer's disease. The present study aims to investigate the impact of age and sex on the neuroinflammatory response to SARS-CoV-2 infection using a mouse model. Wild-type C57BL/6 mice were inoculated, by intranasal route, with SARS-CoV-2 lineage B.1.351 variant known to infect mice. Older animals and in particular males exhibited a significantly greater weight loss starting at 4 dpi. In addition, male animals exhibited higher viral RNA loads and higher titers of infectious virus in the lung, which was particularly evident in males at 16 months of age. Notably, no viral RNA was detected in the brains of infected mice, regardless of age or sex. Nevertheless, expression of IL-6, TNF-α, and CCL-2 in the lung and brain was increased with viral infection. An unbiased brain RNA-seq/transcriptomic analysis showed that SARS-CoV-2 infection caused significant changes in gene expression profiles in the brain, with innate immunity, defense response to virus, cerebravascular and neuronal functions, as the major molecular networks affected. The data presented in this study show that SARS-CoV-2 infection triggers a neuroinflammatory response despite the lack of detectable virus in the brain. Age and sex have a modifying effect on this pathogenic process. Aberrant activation of innate immune response, disruption of blood-brain barrier and endothelial cell integrity, and supression of neuronal activity and axonogenesis underlie the impact of SARS-CoV-2 infection on the brain. Understanding the role of these affected pathways in SARS-CoV-2 pathogenesis helps identify appropriate points of therapeutic interventions to alleviate neurological dysfunction observed during COVID-19.
Collapse
Affiliation(s)
- Venkatramana D. Krishna
- Department of Veterinary Population Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | | | - Holly Korthas
- Department of Experimental and Clinical Pharmacology
| | - Susanna R. Var
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Walter C. Low
- Graduate Program in Neuroscience
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ling Li
- Graduate Program in Neuroscience
- Department of Experimental and Clinical Pharmacology
| | - Maxim C-J. Cheeran
- Department of Veterinary Population Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
45
|
Chen S, Wang S. The immune mechanism of the nasal epithelium in COVID-19-related olfactory dysfunction. Front Immunol 2023; 14:1045009. [PMID: 37529051 PMCID: PMC10387544 DOI: 10.3389/fimmu.2023.1045009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 06/29/2023] [Indexed: 08/03/2023] Open
Abstract
During the first waves of the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, olfactory dysfunction (OD) was reported as a frequent clinical sign. The nasal epithelium is one of the front-line protections against viral infections, and the immune responses of the nasal mucosa may be associated with OD. Two mechanisms underlying OD occurrence in COVID-19 have been proposed: the infection of sustentacular cells and the inflammatory reaction of the nasal epithelium. The former triggers OD and the latter likely prolongs OD. These two alternative mechanisms may act in parallel; the infection of sustentacular cells is more important for OD occurrence because sustentacular cells are more likely to be the entry point of SARS-CoV-2 than olfactory neurons and more susceptible to early injury. Furthermore, sustentacular cells abundantly express transmembrane protease, serine 2 (TMPRSS2) and play a major role in the olfactory epithelium. OD occurrence in COVID-19 has revealed crucial roles of sustentacular cells. This review aims to elucidate how immune responses of the nasal epithelium contribute to COVID-19-related OD. Understanding the underlying immune mechanisms of the nasal epithelium in OD may aid in the development of improved medical treatments for COVID-19-related OD.
Collapse
Affiliation(s)
| | - Shufen Wang
- Biomedical Engineering Research Institute, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
46
|
Ligi D, Lo Sasso B, Henry BM, Ciaccio M, Lippi G, Plebani M, Mannello F. Deciphering the role of monocyte and monocyte distribution width (MDW) in COVID-19: an updated systematic review and meta-analysis. Clin Chem Lab Med 2023; 61:960-973. [PMID: 36626568 DOI: 10.1515/cclm-2022-0936] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023]
Abstract
The SARS-CoV-2 infection is characterized by both systemic and organ hyper-thromboinflammation, with a clinical course ranging from mild up-to critical systemic dysfunction and death. In patients with coronavirus disease 2019 (COVID-19) the monocyte/macrophage population is deeply involved as both trigger and target, assuming the value of useful diagnostic/prognostic marker of innate cellular immunity. Several studies correlated morphological and immunophenotypic alterations of circulating monocytes with clinical outcomes in COVID-19 patients, concluding that monocyte distribution width (MDW) may retain clinical value in stratifying the risk of disease worsening. Through an electronic search in Medline and Scopus we performed an updated literature review and meta-analysis aimed to explore the association between increased MDW levels and illness severity in COVID-19 patients, deciphering role(s) and function(s) of monocytes in the harmful network underlining SARS-CoV-2 infection. We found that significantly elevated MDW values were frequently present in COVID-19 patients who developed unfavorable clinical outcomes, compounded by a significant association between monocyte anisocytosis and SARS-CoV-2 outcomes. These findings suggest that blood MDW index and its scatter plot could represent useful routine laboratory tools for early identification of patients at higher risk of unfavorable COVID-19 and for monitoring the progression of viral infection, clinical outcomes, and therapeutic efficacy throughout hospitalization. According to this evidence, therapeutic decisions in patients with SARS-CoV-2 infection could benefit from monitoring MDW value, with administration of drugs limiting thrombo-inflammation due to monocyte hyper-activation in patients with severe/critical COVID-19 disease.
Collapse
Affiliation(s)
- Daniela Ligi
- Unit of Clinical Biochemistry, Section of Biochemistry and Biotechnology, Department of Biomolecular Sciences-DISB, University of Urbino Carlo Bo, Urbino, Italy
| | - Bruna Lo Sasso
- Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, BiND, University of Palermo, Palermo, Italy
| | - Brandon M Henry
- Clinical Laboratory, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Marcello Ciaccio
- Institute of Clinical Biochemistry, Clinical Molecular Medicine and Clinical Laboratory Medicine, Department of Biomedicine, Neurosciences and Advanced Diagnostics, BiND, University of Palermo, Palermo, Italy
| | - Giuseppe Lippi
- Section of Clinical Biochemistry and School of Medicine, University Hospital of Verona, Verona, Italy
| | - Mario Plebani
- Department of Laboratory Medicine, University-Hospital of Padova, Padova, Italy
| | - Ferdinando Mannello
- Unit of Clinical Biochemistry, Section of Biochemistry and Biotechnology, Department of Biomolecular Sciences-DISB, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
47
|
Abbasi Habashi S, Koyuncu M, Alizadehsani R. A Survey of COVID-19 Diagnosis Using Routine Blood Tests with the Aid of Artificial Intelligence Techniques. Diagnostics (Basel) 2023; 13:1749. [PMID: 37238232 PMCID: PMC10217633 DOI: 10.3390/diagnostics13101749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/19/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), causing a disease called COVID-19, is a class of acute respiratory syndrome that has considerably affected the global economy and healthcare system. This virus is diagnosed using a traditional technique known as the Reverse Transcription Polymerase Chain Reaction (RT-PCR) test. However, RT-PCR customarily outputs a lot of false-negative and incorrect results. Current works indicate that COVID-19 can also be diagnosed using imaging resolutions, including CT scans, X-rays, and blood tests. Nevertheless, X-rays and CT scans cannot always be used for patient screening because of high costs, radiation doses, and an insufficient number of devices. Therefore, there is a requirement for a less expensive and faster diagnostic model to recognize the positive and negative cases of COVID-19. Blood tests are easily performed and cost less than RT-PCR and imaging tests. Since biochemical parameters in routine blood tests vary during the COVID-19 infection, they may supply physicians with exact information about the diagnosis of COVID-19. This study reviewed some newly emerging artificial intelligence (AI)-based methods to diagnose COVID-19 using routine blood tests. We gathered information about research resources and inspected 92 articles that were carefully chosen from a variety of publishers, such as IEEE, Springer, Elsevier, and MDPI. Then, these 92 studies are classified into two tables which contain articles that use machine Learning and deep Learning models to diagnose COVID-19 while using routine blood test datasets. In these studies, for diagnosing COVID-19, Random Forest and logistic regression are the most widely used machine learning methods and the most widely used performance metrics are accuracy, sensitivity, specificity, and AUC. Finally, we conclude by discussing and analyzing these studies which use machine learning and deep learning models and routine blood test datasets for COVID-19 detection. This survey can be the starting point for a novice-/beginner-level researcher to perform on COVID-19 classification.
Collapse
Affiliation(s)
| | - Murat Koyuncu
- Department of Information Systems Engineering, Atilim University, 06830 Ankara, Turkey;
| | - Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, Waurn Ponds, Geelong, VIC 3216, Australia
| |
Collapse
|
48
|
Zafar H, Saier MH. Understanding the Relationship of the Human Bacteriome with COVID-19 Severity and Recovery. Cells 2023; 12:cells12091213. [PMID: 37174613 PMCID: PMC10177376 DOI: 10.3390/cells12091213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) first emerged in 2019 in China and has resulted in millions of human morbidities and mortalities across the globe. Evidence has been provided that this novel virus originated in animals, mutated, and made the cross-species jump to humans. At the time of this communication, the Coronavirus disease (COVID-19) may be on its way to an endemic form; however, the threat of the virus is more for susceptible (older and immunocompromised) people. The human body has millions of bacterial cells that influence health and disease. As a consequence, the bacteriomes in the human body substantially influence human health and disease. The bacteriomes in the body and the immune system seem to be in constant association during bacterial and viral infections. In this review, we identify various bacterial spp. In major bacteriomes (oral, nasal, lung, and gut) of the body in healthy humans and compare them with dysbiotic bacteriomes of COVID-19 patients. We try to identify key bacterial spp. That have a positive effect on the functionality of the immune system and human health. These select bacterial spp. Could be used as potential probiotics to counter or prevent COVID-19 infections. In addition, we try to identify key metabolites produced by probiotic bacterial spp. That could have potential anti-viral effects against SARS-CoV-2. These metabolites could be subject to future therapeutic trials to determine their anti-viral efficacies.
Collapse
Affiliation(s)
- Hassan Zafar
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, CA 92093-0116, USA
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic
| | - Milton H Saier
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, CA 92093-0116, USA
| |
Collapse
|
49
|
Santinelli L, Lazzaro A, Sciarra F, Maddaloni L, Frasca F, Fracella M, Moretti S, Borsetti A, Bugani G, Alessandri F, Zullino V, Ruberto F, Pugliese F, Sorrentino L, Gianfrilli D, Isidori A, Venneri MA, Mastroianni CM, Ceccarelli G, d’Ettorre G. Cellular Immune Profiling of Lung and Blood Compartments in Patients with SARS-CoV-2 Infection. Pathogens 2023; 12:pathogens12030442. [PMID: 36986364 PMCID: PMC10057444 DOI: 10.3390/pathogens12030442] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Background: SARS-CoV-2 related immunopathology may be the driving cause underlying severe COVID-19. Through an immunophenotyping analysis on paired bronchoalveolar lavage fluid (BALF) and blood samples collected from mechanically ventilated patients with COVID-19-associated Acute Respiratory Distress Syndrome (CARDS), this study aimed to evaluate the cellular immune responses in survivors and non-survivors of COVID-19. Methods: A total of 36 paired clinical samples of bronchoalveolar lavage fluid (BALF) mononuclear cells (BALF-MC) and peripheral blood mononuclear cells (PBMC) were collected from 18 SARS-CoV-2-infected subjects admitted to the intensive care unit (ICU) of the Policlinico Umberto I, Sapienza University Hospital in Rome (Italy) for severe interstitial pneumonia. The frequencies of monocytes (total, classical, intermediate and non-classical) and Natural Killer (NK) cell subsets (total, CD56bright and CD56dim), as well as CD4+ and CD8+ T cell subsets [naïve, central memory (TCM) and effector memory (TEM)], and those expressing CD38 and/or HLADR were evaluated by multiparametric flow cytometry. Results: Survivors with CARDS exhibited higher frequencies of classical monocytes in blood compared to non-survivors (p < 0.05), while no differences in the frequencies of the other monocytes, NK cell and T cell subsets were recorded between these two groups of patients (p > 0.05). The only exception was for peripheral naïve CD4+ T cells levels that were reduced in non-survivors (p = 0.04). An increase in the levels of CD56bright (p = 0.012) and a decrease in CD56dim (p = 0.002) NK cell frequencies was also observed in BALF-MC samples compared to PBMC in deceased COVID-19 patients. Total CD4+ and CD8+ T cell levels in the lung compartment were lower compared to blood (p = 0.002 and p < 0.01, respectively) among non-survivors. Moreover, CD38 and HLA-DR were differentially expressed by CD4+ and CD8+ T cell subsets in BALF-MC and in PBMC among SARS-CoV-2-infected patients who died from COVID-19 (p < 0.05). Conclusions: These results show that the immune cellular profile in blood and pulmonary compartments was similar in survivors and non-survivors of COVID-19. T lymphocyte levels were reduced, but resulted highly immune-activated in the lung compartment of patients who faced a fatal outcome.
Collapse
Affiliation(s)
- Letizia Santinelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandro Lazzaro
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesca Sciarra
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Luca Maddaloni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Frasca
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy
| | - Matteo Fracella
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Italian Institute of Health, 00161 Rome, Italy
- Correspondence: (S.M.); (G.C.); Tel.: +39-0649903591 (S.M.); +39-0649970311 (G.C.)
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Italian Institute of Health, 00161 Rome, Italy
| | - Ginevra Bugani
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Alessandri
- Department of General and Specialistic Surgery, Sapienza University of Rome, 00185 Rome, Italy
| | - Veronica Zullino
- Department of General and Specialistic Surgery, Sapienza University of Rome, 00185 Rome, Italy
| | - Franco Ruberto
- Department of General and Specialistic Surgery, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Pugliese
- Department of General and Specialistic Surgery, Sapienza University of Rome, 00185 Rome, Italy
| | - Leonardo Sorrentino
- Laboratory of Virology, Department of Molecular Medicine, Affiliated to Istituto Pasteur Italia, Sapienza University, 00185 Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Andrea Isidori
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Claudio M. Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
- Azienda Ospedaliero Universitaria Policlinico Umberto I, 00185 Rome, Italy
- Correspondence: (S.M.); (G.C.); Tel.: +39-0649903591 (S.M.); +39-0649970311 (G.C.)
| | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
50
|
Santos LD, Antunes KH, Cassão G, Gonçalves JI, Abbadi BL, Bizarro CV, Basso LA, Machado P, de Souza APD, Porto BN. SARS-CoV-2 immune complex triggers human monocyte necroptosis. Int Immunopharmacol 2023; 117:109954. [PMID: 36870284 PMCID: PMC9968621 DOI: 10.1016/j.intimp.2023.109954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/01/2023]
Abstract
We analyzed the ability of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) itself and SARS-CoV-2-IgG immune complexes to trigger human monocyte necroptosis. SARS-CoV-2 was able to induce monocyte necroptosis dependently of MLKL activation. Necroptosis-associated proteins (RIPK1, RIPK3 and MLKL) were involved in SARS-CoV-2N1 gene expression in monocytes. SARS-CoV-2 immune complexes promoted monocyte necroptosis in a RIPK3- and MLKL-dependent manner, and Syk tyrosine kinase was necessary for SARS-CoV-2 immune complex-induced monocyte necroptosis, indicating the involvement of Fcγ receptors on necroptosis. Finally, we provide evidence that elevated LDH levels as a marker of lytic cell death are associated with COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Leonardo Duarte Santos
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Life and Health Science, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Krist Helen Antunes
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Life and Health Science, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Gisele Cassão
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Life and Health Science, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - João Ismael Gonçalves
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Life and Health Science, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Bruno Lopes Abbadi
- Research Center of Functional and Molecular Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristiano Valim Bizarro
- Research Center of Functional and Molecular Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiz Augusto Basso
- Research Center of Functional and Molecular Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Pablo Machado
- Research Center of Functional and Molecular Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana Paula D de Souza
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Life and Health Science, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil.
| | - Bárbara Nery Porto
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|