1
|
de Oliveira JN, Fernandes CYM, de Godoy SM, Frantine-Silva W, de Souza Cassela PLC, Trigo GL, Lozovoy MAB, Tano ZN, Simão ANC, de Oliveira KB. Association of IL10 gene SNVs rs1800896 (A>G), rs1800871 (C>T), rs1800872 (C>A) and haplotypes with COVID-19 severity and outcome in the Brazilian population. Hum Immunol 2025; 86:111261. [PMID: 39933261 DOI: 10.1016/j.humimm.2025.111261] [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: 08/13/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Elevated concentrations of IL-10 have been detected in coronavirus disease (COVID-19) patients and are a possible disease severity marker. Single nucleotide variants (SNVs) and their haplotypes can be associated with differences in IL-10 levels and with viral disease susceptibility. AIM Evaluate the associations of SNVs and their haplotypes in Brazilian patients with COVID-19 severity and outcome. METHODS In this cross-sectional and case-control study, the patients were selected from the University Hospital of State University of Londrina (HU-UEL) (n = 367) and were subdivided into mild (n = 165), moderate (n = 72) and severe (n = 130) groups. The DNA samples of the participants were subjected to real-time PCR for the detection of rs1800896 (A>G), rs1800871 (C>T) and rs1800872 (C>A) genotypes. The haplotypes were inferred with PHASE v2.1.1. RESULTS The severe cases of COVID-19 were independently associated with the GG genotype (rs1800896) (P = 0.038, OR 2.522, 95 % CI 1.053-6.038) as well as with the GCC haplotype in homozygosity (P = 0.037, OR 2.767, 95 % CI 1.065-7.191). CONCLUSION These results showed that the GG genotype of rs1800896 or the GCC haplotype are associated with COVID-19 severity in Brazilian patients.
Collapse
Affiliation(s)
- Janaina Nicolau de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, Pr 445 km 380 Celso Garcia Cid Highway 86.057-970 PR, Brazil
| | - Caroline Yukari Motoori Fernandes
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, Pr 445 km 380 Celso Garcia Cid Highway 86.057-970 PR, Brazil
| | - Sara Mataroli de Godoy
- Laboratory for Studies and Analysis of Polymorphisms, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina 86.057-970 PR, Brazil
| | - Wilson Frantine-Silva
- Laboratory for Studies and Analysis of Polymorphisms, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina 86.057-970 PR, Brazil
| | | | - Guilherme Lerner Trigo
- Department of Applied Pathology, Clinical and Toxicological Analysis, State University of Londrina 86.057-970 PR, Brazil
| | | | - Zuleica Naomi Tano
- Department of Clinical Medicine, University of Londrina, Londrina, PR, Brazil
| | - Andrea Name Colado Simão
- Department of Applied Pathology, Clinical and Toxicological Analysis, State University of Londrina 86.057-970 PR, Brazil
| | - Karen Brajão de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, Pr 445 km 380 Celso Garcia Cid Highway 86.057-970 PR, Brazil; Laboratory for Studies and Analysis of Polymorphisms, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina 86.057-970 PR, Brazil.
| |
Collapse
|
2
|
Nivean PD, Shetty R, Sethu S, Ghosh A, Kumaramanikavel G, Koka K, Webers CAB, Berendschot TT, Paridaens D. Role of biomarkers in South Indian Thyroid Eye Disease study (SITED). Orbit 2025:1-9. [PMID: 39937544 DOI: 10.1080/01676830.2025.2453536] [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: 10/26/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE Thyroid eye disease (TED) is a complex autoimmune disease. Early detection with routine disease monitoring using biomarker assessment would help in mitigating TED-associated vision loss. Hence, we performed a non-invasive tear fluid (TF) based screening in patients with TED as part of the South Indian Thyroid Eye Disease Study (SITED). MATERIALS AND METHODS We used TF from healthy controls (HC;13 eyes;13 subjects), patients with thyroid dysfunction but without TED (No TED;11 eyes;11 subjects) and patients with TED (18 eyes;18 subjects). TED subjects were further sub-divided into those with and without an active form of the disease. Patients with dysthyroid optic neuropathy (DON) were analyzed separately. The diagnosis of TED was based on Gorman and Bartley's criteria. Activity was defined as scoring more than 4 in the Vision, Inflammation, Strabismus and Appearance (VISA). Schirmer's strip was used to collect TF and the levels of IL-2, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-13, IL-17A, IL-17F, IL-22, IFNγ, TNFα, PDGF-AA and PDGF-BB were determined by multiplex ELISA using flow cytometry. RESULTS Significantly (p < 0.05) higher levels of IL-6 and IL-10 were observed in TED patients compared to HC and No TED subjects. TF levels of IL-6 and IL-10 were significantly higher in active TED patients compared to No TED subjects. Interestingly, TF levels of PDGF-AA were observed to be negatively associated with IL-4 and IL-13. CONCLUSION Elevated TF levels of IL-6 and IL-10 can be explored for their role as a non-invasive risk stratification biomarker or as targets to modulate management of TED.
Collapse
Affiliation(s)
| | - Rohit Shetty
- Department of Ophthalmology, Narayana Nethralaya, Bangalore, India
| | - Swaminathan Sethu
- Department of Ophthalmology, GROW Research Lab, Narayana Netralaya Foundation, Bangalore, India
| | - Arkasubhra Ghosh
- Department of Ophthalmology, GROW Research Lab, Narayana Netralaya Foundation, Bangalore, India
| | | | - Keerti Koka
- Department of Ophthalmology, Medical Research Foundation, Sankara Nethralaya, Chennai, India
| | - Carroll A B Webers
- Department of Ophthalmology, University Eye Clinic Maastricht, Maastricht, The Netherlands
| | - Tos Tjm Berendschot
- Department of Ophthalmology, University Eye Clinic Maastricht, Maastricht, The Netherlands
| | - Dion Paridaens
- Department of Ophthalmology, Erasmus Medical Centre, Rotterdam, The Netherlands
- Department of Ophthalmology, Rotterdam Eye Hospital, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Majidpour M, Azizi SG, Davodabadi F, Sabeti Akbar-Abad M, Abdollahi Z, Sargazi S, Shahriari H. Recent advances in TGF-β signaling pathway in COVID-19 pathogenesis: A review. Microb Pathog 2025; 199:107236. [PMID: 39701478 DOI: 10.1016/j.micpath.2024.107236] [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: 09/02/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
The coronavirus disease 2019 (COVID-19) has resulted in approximately 7.0 million fatalities between 2019 and 2022, underscoring a pressing need for comprehensive research into its underlying mechanisms and therapeutic avenues. A distinctive feature of severe COVID-19 is the dysregulated immune response characterized by excessive activation of immune cells and the consequent cytokine storms. Recent advancements in our understanding of cellular signaling pathways have illuminated the role of Transforming Growth Factor Beta (TGF-β) as a pivotal signaling molecule with significant implications for the pathogenesis of infectious diseases, including COVID-19. Emerging evidence reveals that TGF-β signaling, when activated by viral components or secondary pathways, adversely affects diverse cell types, particularly immune cells, and lung tissue, leading to complications such as pulmonary fibrosis. In our review article, we critically evaluate recent literature on the involvement of TGF-β signaling in the progression of COVID-19. We discuss a range of pharmacological interventions, including nintedanib, pirfenidone, corticosteroids, proton pump inhibitors, and histone deacetylase inhibitors, and their potential to modulate the TGF-β pathway in the context of COVID-19 treatment. Additionally, we explore ongoing clinical trials involving mesenchymal stem cells, low-dose radiation therapy, and artemisinin derivatives to assess their impact on TGF-β levels and subsequent clinical outcomes in COVID-19 patients. This review is particularly relevant at this juncture as the global health community continues to grapple with the ramifications of the COVID-19 pandemic, highlighting the urgent need for targeted therapeutic strategies aimed at TGF-β modulation to mitigate disease severity and improve patient outcomes.
Collapse
Affiliation(s)
- Mahdi Majidpour
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Seyed Ghader Azizi
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Fatemeh Davodabadi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahboobeh Sabeti Akbar-Abad
- Department of Clinical Biochemistry, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Zahra Abdollahi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran.
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Hossein Shahriari
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
4
|
Hirt N, Manchon E, Chen Q, Delaroque C, Corneau A, Hemon P, Saker-Delye S, Bataille P, Bouaziz JD, Bourrat E, Hovnanian A, Le Buanec H, Aoudjit F, El Costa H, Jabrane-Ferrat N, Al-Daccak R. Systems immunology integrates the complex endotypes of recessive dystrophic epidermolysis bullosa. Nat Commun 2025; 16:664. [PMID: 39809737 PMCID: PMC11733305 DOI: 10.1038/s41467-025-55934-7] [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: 04/02/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Endotypes are characterized by the immunological, inflammatory, metabolic, and remodelling pathways that explain the mechanisms underlying the clinical presentation (phenotype) of a disease. Recessive dystrophic epidermolysis bullosa (RDEB) is a severe blistering disease caused by COL7A1 pathogenic variants. Although underscored by animal studies, the endotypes of human RDEB are poorly understood. To fill this gap, we apply systems immunology approaches using single-cell high-dimensional techniques to capture the signature of peripheral immune cells and the diversity of metabolic profiles in RDEB adults, sampled outside of any opportunistic infection and active cancer. Our study, demonstrates the particular inflammation and immunity characteristics of RDEB adults, with activated / effector T and dysfunctional natural killer cell signatures, concomitant with an overall pro-inflammatory lipid signature. Artificial intelligence prediction models and principal component analysis stress that RDEB is not solely confined to cutaneous issues but has complex systemic endotypes marked by immune dysregulation and hyperinflammation. By characterising the phenotype-endotype association in RDEB adults, our study lays the groundwork for translational interventions that could by lessening inflammation, alleviate the everlasting suffering of RDEB patients, while awaiting curative genetic therapies.
Collapse
Affiliation(s)
- Nell Hirt
- National Institute of Health and Medical Research (INSERM) UMRS-976 HIPI, Paris Cité University, Saint-Louis Hospital, 75010, Paris, France
| | - Enzo Manchon
- National Institute of Health and Medical Research (INSERM) UMRS-976 HIPI, Paris Cité University, Saint-Louis Hospital, 75010, Paris, France
| | - Qian Chen
- Boston Childrens Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Clara Delaroque
- INSERM U1016, The National Centre for Scientific Research (CNRS) UMR 8104, Paris Cité University, 75014, Paris, France
| | - Aurelien Corneau
- Pitié-Salpêtrière Cytometry, UMS037, Sorbonne University, 75013, Paris, France
| | - Patrice Hemon
- LBAI, INSERM UMR1227, Brest University, 29200, Brest, France
| | | | - Pauline Bataille
- Dermatology Department, AP-HP, Saint-Louis Hospital, 75010, Paris, France
| | - Jean-David Bouaziz
- National Institute of Health and Medical Research (INSERM) UMRS-976 HIPI, Paris Cité University, Saint-Louis Hospital, 75010, Paris, France
- Dermatology Department, AP-HP, Saint-Louis Hospital, 75010, Paris, France
| | - Emmanuelle Bourrat
- Dermatology Department, AP-HP, Saint-Louis Hospital, 75010, Paris, France
| | - Alain Hovnanian
- Laboratory of Genetic Skin Diseases, Imagine Institute, Paris Cité University, INSERM UMR 1163, 75015, Paris, France
| | - Helene Le Buanec
- National Institute of Health and Medical Research (INSERM) UMRS-976 HIPI, Paris Cité University, Saint-Louis Hospital, 75010, Paris, France
| | - Fawzi Aoudjit
- Division of Immune and Infectious Diseases, CHU de Quebec Research Centre, Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Hicham El Costa
- Institute for Infectious and Inflammatory Diseases, CNRS UMR5051, INSERM UMR1291, Toulouse III University, 31059, Toulouse, France
| | - Nabila Jabrane-Ferrat
- Institute for Infectious and Inflammatory Diseases, CNRS UMR5051, INSERM UMR1291, Toulouse III University, 31059, Toulouse, France
| | - Reem Al-Daccak
- National Institute of Health and Medical Research (INSERM) UMRS-976 HIPI, Paris Cité University, Saint-Louis Hospital, 75010, Paris, France.
| |
Collapse
|
5
|
Gultom M, Lin L, Brandt CB, Milusev A, Despont A, Shaw J, Döring Y, Luo Y, Rieben R. Sustained Vascular Inflammatory Effects of SARS-CoV-2 Spike Protein on Human Endothelial Cells. Inflammation 2024:10.1007/s10753-024-02208-x. [PMID: 39739157 DOI: 10.1007/s10753-024-02208-x] [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: 08/30/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has been associated with systemic inflammation and vascular injury, which contribute to the development of acute respiratory syndrome (ARDS) and the mortality of COVID-19 infection. Moreover, multiorgan complications due to persistent endothelial dysfunction have been suspected as the cause of post-acute sequelae of SARS-CoV-2 infection. Therefore, elucidation of the vascular inflammatory effect of SARS-CoV-2 will increase our understanding of how endothelial cells (ECs) contribute to the short- and long-term consequences of SARS-CoV-2 infection. Here, we investigated the interaction of SARS-CoV-2 spike protein with human ECs from aortic (HAoEC) and pulmonary microvascular (HPMC) origins, cultured under physiological flow conditions. We showed that the SARS-CoV-2 spike protein triggers prolonged expression of cell adhesion markers in both ECs, similar to the effect of TNF-α. SARS-CoV-2 spike treatment also led to the release of various cytokines and chemokines observed in severe COVID-19 patients. Moreover, increased binding of leucocytes to the endothelial surface and a procoagulant state of the endothelium were observed. Transcriptomic profiles of SARS-CoV-2 spike-activated HPMC and HAoEC showed prolonged upregulation of genes and pathways associated with responses to virus, cytokine-mediated signaling, pattern recognition, as well as complement and coagulation pathways. Our findings support experimental and clinical observations of the vascular consequences of SARS-CoV-2 infection and highlight the importance of EC protection as one of the strategies to mitigate the severe effects as well as the possible post-acute complications of COVID-19 disease.
Collapse
Affiliation(s)
- Mitra Gultom
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Camilla Blunk Brandt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Anastasia Milusev
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Alain Despont
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Jane Shaw
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Yvonne Döring
- Department for Biomedical Research, University of Bern, Bern, Switzerland
- Department of Angiology, Inselspital, Bern University Hospital, Bern, Switzerland
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian University, Munich, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum Für Herz-Kreislauf-Forschung, DZHK), Munich Heart Alliance Partner Site, Munich, Germany
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Robert Rieben
- Department for Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
6
|
Asgari A, Franczak A, Herchen A, Jickling GC, Jurasz P. Elevated levels of pro-thrombotic eNOS-negative platelets in COVID-19 patients. Thromb Res 2024; 244:109178. [PMID: 39369655 DOI: 10.1016/j.thromres.2024.109178] [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: 05/09/2024] [Revised: 09/13/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Platelet-rich microvascular thrombi are common in severe COVID-19. Endogenous nitric oxide (NO)-signaling limits thrombus formation and previously we identified platelet subpopulations with a differential ability to produce NO based on the presence or absence of endothelial nitric oxide synthase (eNOS). eNOS expression is counter-regulated by cytokines, and COVID-19-associated immune/inflammatory responses may affect the transcriptome profile of megakaryocytes and their platelet progeny. OBJECTIVES We investigated whether the percentage of eNOS-negative to eNOS-positive platelets increases in COVID-19 patients and whether this change may be due to the actions of pro-inflammatory cytokines on megakaryocytes. METHODS Platelets were isolated from hospitalized COVID-19 patients and COVID-19-negative controls. Platelet eNOS was measured by flow cytometry and plasma inflammatory cytokines by ELISA. Megakaryocytes from eNOS-GFP transgenic mice and the Meg-01 cell line were characterized to identify an appropriate model to study eNOS-based platelet subpopulation formation in response to inflammatory cytokines. RESULTS COVID-19 patients demonstrated a significant increase in eNOS-negative and a concomitant decrease in eNOS-positive platelets compared to controls, and this change was associated with disease severity as assessed by ICU admission. A higher eNOS-negative to -positive platelet percentage was associated with enhanced platelet activation as measured by surface CD62P. Accordingly, COVID-19 patients demonstrated higher TNF-α, IL-6, and IL-1β plasma concentrations than controls. Inflammatory cytokines associated with COVID-19 promoted eNOS-negative Meg-01 formation and enhanced subsequent eNOS-negative platelet-like particle formation. CONCLUSIONS COVID-19 patients have a higher percentage of eNOS-negative to -positive platelets, likely as a result of inflammatory response reducing megakaryocyte eNOS expression, which predisposes to thrombosis.
Collapse
Affiliation(s)
- Amir Asgari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Aleksandra Franczak
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Alex Herchen
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada; School of Medicine and Dentistry, Griffith University, Queensland, Australia
| | - Glen C Jickling
- Department of Medicine, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
7
|
Zhang W, Wang X, Zhang H, Pan Y, Ma W, Xu Y, Tian Z, Xia C, Fu L, Wang Y. Comparison of pathogenicity and host responses of emerging porcine reproductive and respiratory syndrome virus variants in piglets. J Virol 2024; 98:e0154223. [PMID: 39445829 PMCID: PMC11575335 DOI: 10.1128/jvi.01542-23] [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: 08/16/2024] [Accepted: 09/15/2024] [Indexed: 10/25/2024] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a highly variable virus with genetic diversity. This study comparatively examines the pathogenicity and immunological impact of two emergent PRRSV strains, SD53 and HuN4, in piglets. Our results indicate that SD53 strain induces milder clinical syndromes and less severe tissue damage than HuN4, despite similar replication rates. Hematological tests showed less perturbations in peripheral blood cell profiles after SD53 infection, suggesting a less systemic impact. The neutrophil-to-lymphocyte ratio was notably lower in SD53-infected piglets, suggesting a less intense inflammatory reaction. Moreover, SD53 infection led to lower levels of pro-inflammatory cytokines, further supporting a less pronounced inflammatory profile. Both strains induced the production of PRRSV-specific antibodies. However, transcriptomic analysis of lung and lymph node tissues from infected piglets disclosed a more moderate up-regulation of core genes, including ISGs, in the SD53 group. Further analysis indicated that SD53 primarily enhanced immune-related signaling, particularly in T cell response modules, while HuN4 caused a more robust pro-inflammatory reaction and a dampening of T cell functionality. Flow cytometry analyses confirmed these findings, showing higher CD4/CD8 ratios and increased CD4+ T cell percentages in SD53-infected piglets, implying a more robust T cell response. Collectively, these findings broaden our comprehension of PRRSV pathogenesis and may inform the development of future therapeutic or prophylactic strategies for controlling PRRSV infections more effectively. IMPORTANCE The high mutation rate of porcine reproductive and respiratory syndrome virus (PRRSV) poses significant challenges to its accurate diagnosis and the implementation of effective control measures. This research explores the pathogenic profiles of two emerging PRRSV stains: the NADC30-like strain SD53 and the highly pathogenic strain HuN4. Our investigation reveals that SD53 initiates distinct immunopathological responses in vivo compared with those provoked by HuN4. By conducting a transcriptome analysis of differential gene expression in the lungs and lymph nodes of infected piglets, we unveil the intricate molecular mechanisms underlying the contrasting pathogenicity of these two strains. The comprehensive insights yielded by this study are instrumental in advancing our understanding of the dominant NADC30-like PRRSV strain, which has become increasingly prevalent in China's swine industry.
Collapse
Affiliation(s)
- Wenli Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xinrong Wang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - He Zhang
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yu Pan
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Wenjie Ma
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- Chongqing Academy of Animal Science, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Yunfei Xu
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhijun Tian
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Changyou Xia
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Lizhi Fu
- Chongqing Academy of Animal Science, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Yue Wang
- College of Veterinary Medicine, Southwest University, Chongqing, China
- Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| |
Collapse
|
8
|
Amoddeo A. In silico assessment of CAR macrophages activity against SARS-CoV-2 infection. Heliyon 2024; 10:e39689. [PMID: 39524874 PMCID: PMC11550025 DOI: 10.1016/j.heliyon.2024.e39689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Macrophage engineering with chimeric antigen receptor is a promising technique first applied to the treatment of tumours and recently suggested as a possible immunotherapeutic route against the COVID-19 disease. Four immunotherapies based on engineered macrophages have been tested in vitro revealing promising, with one of them acting without increasing the cytokines level. We present a mathematical model aimed at the evaluation of both the SARS-CoV-2 virions dynamics and the cytokines production induced, while such newly developed constructs interact with the immune system once administered. The importance of the study lies both in monitoring the dynamics of the infection and in evaluating the cytokine production, since clinical studies show that in critical COVID-19 patients an abnormal cytokines production occurs, a concern to be accounted for in designing appropriate therapeutic strategies. The mathematical model was built in the context of the continuum approach of the mass conservation, while the numerical simulations have been performed introducing parameters deduced from the experiments, using the finite element method. The model simulations allow to analyse and to compare the immune mechanisms underlying the virus dynamics, deepening the investigation for two selected immunotherapies, suggesting that a synergistic work of involved cytokines with phagocytic activity of macrophages occurs. The best SARS-CoV-2 clearance relies not only on the phagocytic capacity of the engineered macrophages, but also on the production of T-lymphocytes, pro- and anti-inflammatory cytokines which in the two cases examined in depth can decrease by 99.7 %, 99.6 % and 69 % respectively, passing from the most effective immunotherapy to the least effective one. This study is the first mathematical model that analyses the dynamics of macrophages engineered to fight the COVID-19, and paves the way for their possible exploitation against such a challenging disease, going beyond existing models involving other immune cells.
Collapse
Affiliation(s)
- Antonino Amoddeo
- Department of Civil, Energy, Environment and Materials Engineering, Università’Mediterranea’ di Reggio Calabria, Via R. Zehender 1, Feo di Vito, I-89122, Reggio Calabria, Italy
| |
Collapse
|
9
|
Basting CM, Langat R, Broedlow CA, Guerrero CR, Bold TD, Bailey M, Velez A, Schroeder T, Short-Miller J, Cromarty R, Mayer ZJ, Southern PJ, Schacker TW, Safo SE, Bramante CT, Tignanelli CJ, Schifanella L, Klatt NR. SARS-CoV-2 infection is associated with intestinal permeability, systemic inflammation, and microbial dysbiosis in hospitalized patients. Microbiol Spectr 2024; 12:e0068024. [PMID: 39345212 PMCID: PMC11537016 DOI: 10.1128/spectrum.00680-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) and its associated severity have been linked to uncontrolled inflammation and may be associated with changes in the microbiome of mucosal sites including the gastrointestinal tract and oral cavity. These sites play an important role in host-microbe homeostasis, and disruption of epithelial barrier integrity during COVID-19 may potentially lead to exacerbated inflammation and immune dysfunction. Outcomes in COVID-19 are highly disparate, ranging from asymptomatic to fatal, and the impact of microbial dysbiosis on disease severity is unclear. Here, we obtained plasma, rectal swabs, oropharyngeal swabs, and nasal swabs from 86 patients hospitalized with COVID-19 and 12 healthy volunteers. We performed 16S rRNA sequencing to characterize the microbial communities in the mucosal swabs and measured concentrations of circulating cytokines, markers of gut barrier integrity, and fatty acids in the plasma samples. We compared these plasma concentrations and microbiomes between healthy volunteers and COVID-19 patients, some of whom had unfortunately died by the end of the study enrollment, and performed a correlation analysis between plasma variables and bacterial abundances. Rectal swabs of COVID-19 patients had reduced abundances of several commensal bacteria including Faecalibacterium prausnitzii and an increased abundance of the opportunistic pathogens Eggerthella lenta and Hungatella hathewayi. Furthermore, the oral pathogen Scardovia wiggsiae was more abundant in the oropharyngeal swabs of COVID-19 patients who died. The abundance of both H. hathewayi and S. wiggsiae correlated with circulating inflammatory markers including IL-6, highlighting the possible role of the microbiome in COVID-19 severity and providing potential therapeutic targets for managing COVID-19.IMPORTANCEOutcomes in coronavirus disease 2019 (COVID-19) are highly disparate and are associated with uncontrolled inflammation; however, the individual factors that lead to this uncontrolled inflammation are not fully understood. Here, we report that severe COVID-19 is associated with systemic inflammation, microbial translocation, and microbial dysbiosis. The rectal and oropharyngeal microbiomes of COVID-19 patients were characterized by a decreased abundance of commensal bacteria and an increased abundance of opportunistic pathogens, which positively correlated with markers of inflammation and microbial translocation. These microbial perturbations may, therefore, contribute to disease severity in COVID-19 and highlight the potential for microbiome-based interventions in improving COVID-19 outcomes.
Collapse
Affiliation(s)
| | - Robert Langat
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Candace R. Guerrero
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minnesota, Minneapolis, USA
- College of Biological Sciences, University of Minnesota, Minnesota, Minneapolis, USA
| | - Tyler D. Bold
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | - Melisa Bailey
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Adrian Velez
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ty Schroeder
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jonah Short-Miller
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ross Cromarty
- Masonic Cancer Center, University of Minnesota, Minnesota, Minneapolis, USA
| | - Zachary J. Mayer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minnesota, Minneapolis, USA
- College of Biological Sciences, University of Minnesota, Minnesota, Minneapolis, USA
| | - Peter J. Southern
- Department of Microbiology and Immunology, University of Minnesota, Minnesota, Minneapolis, USA
| | - Timothy W. Schacker
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | - Sandra E. Safo
- Department of Biostatistics and Health Data Science, University of Minnesota, Minnesota, Minneapolis, USA
| | - Carolyn T. Bramante
- Department of Medicine, University of Minnesota, Minnesota, Minneapolis, USA
| | | | - Luca Schifanella
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
- National Cancer Institute, Center for Cancer Research, Vaccine Branch, Animal Models and Retroviral Vaccines Section, National Institutes of Health, Bethesda, Maryland, USA
| | - Nichole R. Klatt
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
10
|
Ferreira RR, Martins RB, Pires I, Marques BL, Costa KC, Lirio PH, Scomparin DS, Scarante FF, Batah SS, Hallak JE, Crippa JA, Rodrigues LC, Tostes RC, Fabro AT, Arruda E, Campos AC. Cardiovascular and kidney diseases are positively associated with neuroinflammation and reduced brain-derived neurotrophic factor in patients with severe COVID-19. Brain Behav Immun Health 2024; 41:100855. [PMID: 39391797 PMCID: PMC11466569 DOI: 10.1016/j.bbih.2024.100855] [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: 01/23/2024] [Revised: 05/25/2024] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Even though respiratory dysfunctions are the primary symptom associated with SARS-CoV-2 infection, cerebrovascular events, and neurological symptoms are described in many patients. However, the connection between the neuroimmune profile and the lung's inflammatory condition during COVID-19 and its association with the neurological symptoms reported by COVID-19 patients still needs further exploration. The present study characterizes the SARS-CoV-2 infectivity profile in postmortem nervous and lung tissue samples of patients who died due to severe COVID-19, and the pro-inflammatory factors present in both nervous and lung tissue samples, via a proteomic profiling array. Additionally, Brain-Derived Neurotrophic Factor (BDNF) levels and intracellular pathways related to neuroplasticity/neuroprotection were assessed in the samples. Out of the 16 samples analyzed, all samples but 1 were positive for the viral genome (genes E or N2, but only 3.9% presented E and N2) in the olfactory brain pathway. The E or N2 gene were also detected in all lung samples, with 43.7% of the samples being positive for the E and N2 genes. In the E/N2 positive brain samples, the Spike protein of SARS-CoV-2 co-localized with TUJ-1+ (neuron-specific class III beta-tubulin) and GFAP+ (glial fibrillary acidic protein) astrocytes. IL-6, but not IL-10, expression was markedly higher in most nervous tissue samples compared to the lung specimens. While intracellular adhesion molecule-1 (ICAM-1), interleukin-8 (IL-8), macrophage migration inhibitory factor (MIF), and plasminogen activator inhibitor 1 (PAI-1) were increased in lung samples from SARS-Cov-2 patients, only MIF and IL-18 were detected in nervous tissue samples. Correlation analysis suggested that high levels of IL-6 are followed by increased levels of IL-10 in the brain, but not in lung samples. Our analysis also demonstrated that the presence of comorbidities, such as cardiovascular disease, hypertension, and hypothyroidism, is associated with neuroinflammation, while chronic kidney conditions predict the presence of neurological symptoms, which correlate with lower levels of BDNF in the brain samples. Our results corroborate the hypothesis that a pro-inflammatory state might further impair neural homeostasis and induce brain abnormalities found in COVID-19 patients.
Collapse
Affiliation(s)
- Rafael R. Ferreira
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Ronaldo B. Martins
- Department of Cell and Molecular Biology, Ribeirão Preto School of Medicine, University of Sao Paulo, Ribeirão Preto, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil, 14049-900, São Paulo, Brazil
| | - Isabela Pires
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Bruno L. Marques
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Karla C.M. Costa
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Pedro H.C. Lirio
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Davi S. Scomparin
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Franciele F. Scarante
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Sabrina S. Batah
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, 3900, Bandeirantes Avenue - Monte Alegre- Ribeirão Preto-SP-Brazil, 14049-900, Brazil
| | - Jaime E.C. Hallak
- Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirão Preto, University of São Paulo, 2650, Tenente Catão Roxo Street - Monte Alegre, Ribeirão Preto – SP- Brazil, 14051-140, São Paulo, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT TM) - CNPQ/FAPESP/CAPES - Brazil
| | - Jose A. Crippa
- Department of Neuroscience and Behavior, Faculty of Medicine of Ribeirão Preto, University of São Paulo, 2650, Tenente Catão Roxo Street - Monte Alegre, Ribeirão Preto – SP- Brazil, 14051-140, São Paulo, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT TM) - CNPQ/FAPESP/CAPES - Brazil
| | - Livia C.M. Rodrigues
- Department of Physiological Sciences, Health Sciences Center, Universidade Federal do Espírito Santo, Vitória 1468, Marechal Campos Avenue - Maruípe, Vitória, ES, 29047-105, Brazil
| | - Rita C. Tostes
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
| | - Alexandre T. Fabro
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, 3900, Bandeirantes Avenue - Monte Alegre- Ribeirão Preto-SP-Brazil, 14049-900, Brazil
| | - Eurico Arruda
- Department of Cell and Molecular Biology, Ribeirão Preto School of Medicine, University of Sao Paulo, Ribeirão Preto, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil, 14049-900, São Paulo, Brazil
| | - Alline C. Campos
- Department of Pharmacology- Ribeirão Preto Medical School- University of São Paulo, Ribeirão Preto, São Paulo, Brazil, 3900, Bandeirantes Avenue- Monte Alegre- Ribeirão Preto-SP-Brazil - 14049-900
- National Institute of Science and Technology for Translational Medicine (INCT TM) - CNPQ/FAPESP/CAPES - Brazil
| |
Collapse
|
11
|
Nizam R, Malik MZ, Jacob S, Alsmadi O, Koistinen HA, Tuomilehto J, Alkandari H, Al-Mulla F, Thanaraj TA. Circulating hsa-miR-320a and its regulatory network in type 1 diabetes mellitus. Front Immunol 2024; 15:1376416. [PMID: 39464889 PMCID: PMC11502356 DOI: 10.3389/fimmu.2024.1376416] [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: 01/25/2024] [Accepted: 09/26/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction Increasing evidence from human and animal model studies indicates the significant role of microRNAs (miRNAs) in pancreatic beta cell function, insulin signaling, immune responses, and pathogenesis of type 1 diabetes (T1D). Methods We aimed, using next-generation sequencing, to screen miRNAs from peripheral blood mononuclear cells of eight independent Kuwaiti-Arab families with T1D affected siblings, consisting of 18 T1D patients and 18 unaffected members, characterized by no parent-to-child inheritance pattern. Results Our analysis revealed 20 miRNAs that are differentially expressed in T1D patients compared with healthy controls. Module-based weighted gene co-expression network analysis prioritized key consensus miRNAs in T1D pathogenesis. These included hsa-miR-320a-3p, hsa-miR-139-3p, hsa-miR-200-3p, hsa-miR-99b-5p and hsa-miR-6808-3p. Functional enrichment analysis of differentially expressed miRNAs indicated that PI3K-AKT is one of the key pathways perturbed in T1D. Gene ontology analysis of hub miRNAs also implicated PI3K-AKT, along with mTOR, MAPK, and interleukin signaling pathways, in T1D. Using quantitative RT-PCR, we validated one of the key predicted miRNA-target gene-transcription factor networks in an extended cohort of children with new-onset T1D positive for islet autoantibodies. Our analysis revealed that hsa-miR-320a-3p and its key targets, including PTEN, AKT1, BCL2, FOXO1 and MYC, are dysregulated in T1D, along with their interacting partners namely BLIMP3, GSK3B, CAV1, CXCL3, TGFB, and IL10. Receiver Operating Characteristic analysis highlighted the diagnostic potential of hsa-miR-320a-3p, CAV1, GSK3B and MYC for T1D. Discussion Our study presents a novel link between hsa-miR-320a-3p and T1D, and highlights its key regulatory role in the network of mRNA markers and transcription factors involved in T1D pathogenesis.
Collapse
Affiliation(s)
- Rasheeba Nizam
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Md Zubbair Malik
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Sindhu Jacob
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Osama Alsmadi
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Heikki A. Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Metabolism Group, Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hessa Alkandari
- Department of Population Health, Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Pediatrics, Farwaniya Hospital, Ministry of Health, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | | |
Collapse
|
12
|
Cetinkaya PG, Abras IF, Evcili I, Yildirim T, Ceylan Y, Kara Eroglu F, Kayaoglu B, İpekoglu EM, Akarsu A, Yıldırım M, Kahraman T, Cengiz AB, Sahiner UM, Sekerel BE, Ozsurekci Y, Soyer O, Gursel I. Plasma Extracellular Vesicles Derived from Pediatric COVID-19 Patients Modulate Monocyte and T Cell Immune Responses Based on Disease Severity. Immunol Invest 2024; 53:1141-1175. [PMID: 39115924 DOI: 10.1080/08820139.2024.2385992] [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] [Indexed: 10/02/2024]
Abstract
BACKGROUND The COVID-19 pandemic has caused significant morbidity and mortality globally. The role of plasma-derived extracellular vesicles (EVs) in pediatric COVID-19 patients remains unclear. METHODS We isolated EVs from healthy controls (n = 13) and pediatric COVID-19 patients (n = 104) with varying severity during acute and convalescent phases using serial ultracentrifugation. EV effects on healthy PBMCs, naïve CD4+ T cells, and monocytes were assessed through in vitro assays, flow cytometry, and ELISA. RESULTS Our findings indicate that COVID-19 severity correlates with diverse immune responses. Severe acute cases exhibited increased cytokine levels, decreased IFNγ levels, and lower CD4+ T cell and monocyte counts, suggesting immunosuppression. EVs from severe acute patients stimulated healthy cells to express higher PDL1, increased Th2 and Treg cells, reduced IFNγ secretion, and altered Th1/Th17 ratios. Patient-derived EVs significantly reduced proinflammatory cytokine production by monocytes (p < .001 for mild, p = .0025 for severe cases) and decreased CD4+ T cell (p = .043) and monocyte (p = .033) populations in stimulated healthy PBMCs. CONCLUSION This study reveals the complex relationship between immunological responses and EV-mediated effects, emphasizing the impact of COVID-19 severity. We highlight the potential role of plasma-derived EVs in early-stage immunosuppression in severe COVID-19 patients.
Collapse
Affiliation(s)
- Pınar Gur Cetinkaya
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Irem Fatma Abras
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Irem Evcili
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Tugçe Yildirim
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Yasemin Ceylan
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Fehime Kara Eroglu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Başak Kayaoglu
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Emre Mert İpekoglu
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Aysegul Akarsu
- Division of Pediatric Allergy and Asthma Unit, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Muzaffer Yıldırım
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Tamer Kahraman
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Ali Bülent Cengiz
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Umit Murat Sahiner
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Bulent Enis Sekerel
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Yasemin Ozsurekci
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ozge Soyer
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ihsan Gursel
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| |
Collapse
|
13
|
Thom RE, D’Elia RV. Future applications of host direct therapies for infectious disease treatment. Front Immunol 2024; 15:1436557. [PMID: 39411713 PMCID: PMC11473292 DOI: 10.3389/fimmu.2024.1436557] [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: 05/22/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
New and emerging pathogens, such as SARS-CoV2 have highlighted the requirement for threat agnostic therapies. Some antibiotics or antivirals can demonstrate broad-spectrum activity against pathogens in the same family or genus but efficacy can quickly reduce due to their specific mechanism of action and for the ability of the disease causing agent to evolve. This has led to the generation of antimicrobial resistant strains, making infectious diseases more difficult to treat. Alternative approaches therefore need to be considered, which include exploring the utility of Host-Directed Therapies (HDTs). This is a growing area with huge potential but difficulties arise due to the complexity of disease profiles. For example, a HDT given early during infection may not be appropriate or as effective when the disease has become chronic or when a patient is in intensive care. With the growing understanding of immune function, a new generation of HDT for the treatment of disease could allow targeting specific pathways to augment or diminish the host response, dependent upon disease profile, and allow for bespoke therapeutic management plans. This review highlights promising and approved HDTs that can manipulate the immune system throughout the spectrum of disease, in particular to viral and bacterial pathogens, and demonstrates how the advantages of HDT will soon outweigh the potential side effects.
Collapse
Affiliation(s)
- Ruth E. Thom
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Porton Down, Salisbury, United Kingdom
| | - R V. D’Elia
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
14
|
Oğuzoğlu TÇ, Hanifehnezhad A, Khabbazi SD, Karayel-Hacıoğlu İ, Kaynarcalıdan O, Fırat Z, Filazi N, Erdem-Şahinkesen E, Gül B, Karabulut MC, Koba E, Adıgüzel E, Şenlik Eİ, Korkulu E, Demirden C, Şahinkesen İ, Ceylan A, Muratoğlu H, Vural S, Demirbağ Z, Özkul A. Immunogenicity and Protective Efficacy of Baculovirus-Expressed SARS-CoV-2 Envelope Protein in Mice as a Universal Vaccine Candidate. Vaccines (Basel) 2024; 12:977. [PMID: 39340009 PMCID: PMC11435448 DOI: 10.3390/vaccines12090977] [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: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/30/2024] Open
Abstract
The envelope (env) protein of SARS-CoV-2, a pivotal component of the viral architecture, plays a multifaceted role in viral assembly, replication, pathogenesis, and ion channel activity. These features make it a significant target for understanding virus-host interactions and developing vaccines to combat COVID-19. Recent structural studies provide valuable insights into the conformational dynamics and membrane topology of the SARS-CoV-2 env protein, shedding light on its functional mechanisms. The strong homology and highly conserved structure of the SARS-CoV-2 env protein shape its immunogenicity and functional characteristics. This study examines the ability of the recombinant SARS-CoV-2 env protein to stimulate an immune response. In this study, recombinant envelope proteins were produced using the baculovirus expression system, and their potential efficacy was evaluated in both in vivo and in vitro models. Our results reveal that the env protein of SARS-CoV-2 stimulates humoral and cellular responses and highlight its potential as a promising vaccine candidate for combating the ongoing pandemic.
Collapse
Affiliation(s)
- Tuba Çiğdem Oğuzoğlu
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Alireza Hanifehnezhad
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Saber Delpasand Khabbazi
- Department of Agriculture and Food, Institute of Hemp Research, Yozgat Bozok University, Yozgat 66900, Türkiye
| | - İlke Karayel-Hacıoğlu
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Onur Kaynarcalıdan
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Zehra Fırat
- Biotechnology Institute, Ankara University, Ankara 06560, Türkiye
| | - Nazlıcan Filazi
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
- Department of Virology, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Hatay 31040, Türkiye
| | - Eda Erdem-Şahinkesen
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
- Department of Vaccine Technology, Vaccine Institute, Hacettepe University, Ankara 06100, Türkiye
| | - Buket Gül
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
| | - Muhammed Cesim Karabulut
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
| | - Enes Koba
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
| | - Ece Adıgüzel
- Republic of Türkiye Ministry of Agriculture and Forestry, Atkaracalar District Directorate, Çankırı 18310, Türkiye
| | - Elif İrem Şenlik
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
| | - Emrah Korkulu
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
- Department of Virology, Faculty of Veterinary Medicine, Kafkas University, Kars 36000, Türkiye
| | - Cansu Demirden
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
- Republic of Türkiye Ministry of Agriculture and Forestry, East Anatolian Agricultural Research Institute, Erzurum 25090, Türkiye
| | - İlker Şahinkesen
- Diagen Biotechnological Systems Health Services and Automation Industry, Ankara 06070, Türkiye
| | - Ahmet Ceylan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Hacer Muratoğlu
- Department of Molecular Biology and Genetics, Faculty of Sciences, Karadeniz Technical University, Trabzon 61080, Türkiye
| | - Sevil Vural
- Department of Pathology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Zihni Demirbağ
- Department of Biology, Faculty of Sciences, Karadeniz Technical University, Trabzon 61080, Türkiye
| | - Aykut Özkul
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| |
Collapse
|
15
|
Rajaiah R, Pandey K, Acharya A, Ambikan A, Kumar N, Guda R, Avedissian SN, Montaner LJ, Cohen SM, Neogi U, Byrareddy SN. Differential immunometabolic responses to Delta and Omicron SARS-CoV-2 variants in golden syrian hamsters. iScience 2024; 27:110501. [PMID: 39171289 PMCID: PMC11338146 DOI: 10.1016/j.isci.2024.110501] [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: 08/24/2023] [Revised: 02/07/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
Delta (B.1.617.2) and Omicron (B.1.1.529) variants of SARS-CoV-2 represents unique clinical characteristics. However, their role in altering immunometabolic regulations during acute infection remains convoluted. Here, we evaluated the differential immunopathogenesis of Delta vs. Omicron variants in Golden Syrian hamsters (GSH). The Delta variant resulted in higher virus titers in throat swabs and the lungs and exhibited higher lung damage with immune cell infiltration than the Omicron variant. The gene expression levels of immune mediators and metabolic enzymes, Arg-1 and IDO1 in the Delta-infected lungs were significantly higher compared to Omicron. Further, Delta/Omicron infection perturbed carbohydrates, amino acids, nucleotides, and TCA cycle metabolites and was differentially regulated compared to uninfected lungs. Collectively, our data provide a novel insight into immunometabolic/pathogenic outcomes for Delta vs. Omicron infection in the GSH displaying concordance with COVID-19 patients associated with inflammation and tissue injury during acute infection that offered possible new targets to develop potential therapeutics.
Collapse
Affiliation(s)
- Rajesh Rajaiah
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kabita Pandey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anoop Ambikan
- The Systems Virology Lab, Department of Laboratory Medicine, Division of Clinical Microbiology, ANA Futura, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Narendra Kumar
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Reema Guda
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sean N. Avedissian
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Luis J. Montaner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Samuel M. Cohen
- Havlik Wall Professor of Oncology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ujjwal Neogi
- The Systems Virology Lab, Department of Laboratory Medicine, Division of Clinical Microbiology, ANA Futura, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
- Havlik Wall Professor of Oncology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
16
|
Liu Y, Guo Y, Zhan H, Liu X, Li X, Cui J, Li H, Feng S, Cheng L, Li X, Guo S, Li Y. Immune and inflammation features of severe and critical Omicron infected patients during Omicron wave in China. BMC Infect Dis 2024; 24:809. [PMID: 39123106 PMCID: PMC11316362 DOI: 10.1186/s12879-024-09652-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
OBJECTIVE The current study aimed to investigate the baseline immune and inflammatory features and in-hospital outcomes of patients infected with the Omicron variant (PIWO) who presented with different disease severities during the first wave of mass Omicron infections in the Chinese population has occurred. METHOD A cross-sectional study was conducted on 140 hospitalized PIWO between December 11, 2022, and February 16, 2023. The clinical features, antibodies against SARS-CoV-2, immune cells, and inflammatory cytokines among mildly, severely, and critically ill PIWO at baseline and during follow-up period were compared. RESULT Patients with severe (n = 49) and critical (n = 35) disease were primarily male, needed invasive mechanical ventilation treatment, and exhibited higher mortality than those with mild disease (n = 56). During acute infection, SARS-CoV-2-specific antibody levels fluctuated with disease severity, serum antibodies increased and the incidence of severe cases decreased in critically ill PIWO over time. Antibody titers in severe or critical PIWO with no antibody responses at baseline did not increase significantly over time. Meanwhile, CD4+T cell, CD8+T cell, and natural killer cell counts were negatively correlated with disease severity, whereas interleukin (IL)-6 and IL-10 levels were positively correlated. In addition, combined diabetes, immunosuppressive therapy before infection, serum amyloid A, IL-10 and neutrophil counts were independently associated with severe and critical illness in PIWO. Among the 11 nonsurvivors, 8, 2, 1 died of respiratory failure, sudden cardiac death, and renal failure, respectively. Compared with survivors, nonsurvivors exhibited lower seropositivity of SARS-CoV-2-specific antibody, reduced CD3+T and CD4+T cell counts, and higher IL-2R, IL-6, IL-8, and IL-10 levels. Of note, lactate dehydrogenase was a significant risk factor of death in severe or critically ill PIWO. CONCLUSION This present study assessed the dynamic changes of SARS-CoV-2-specific antibodies, immune cells and inflammatory indexes between severely and critically ill PIWO. Critical and dead PIWO featured compromised humoral immune response and excessive inflammation, which broadened the understanding of the pathophysiology of Omicron infection and provides warning markers for severe disease and poor prognosis.
Collapse
Affiliation(s)
- Yongmei Liu
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100010, China
| | - Yaping Guo
- Department of Clinical Laboratory, Baoding First Central HospitalBaoding NO.1 Central Hospital, No. 320, Great Wall North Street, Baoding, Hebei, 071000, China
| | - Haoting Zhan
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100010, China
| | - Xin Liu
- Department of Clinical Laboratory, Baoding First Central HospitalBaoding NO.1 Central Hospital, No. 320, Great Wall North Street, Baoding, Hebei, 071000, China
| | - Xiaomeng Li
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100010, China
- Department of Clinical Laboratory, Peking University People's Hospital, NO.11, Xizhimen South Street, Beijing, 100035, China
| | - Jingjing Cui
- Department of Clinical Laboratory, Baoding First Central HospitalBaoding NO.1 Central Hospital, No. 320, Great Wall North Street, Baoding, Hebei, 071000, China
| | - Haolong Li
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100010, China
| | - Sha Feng
- Department of Clinical Laboratory, Baoding First Central HospitalBaoding NO.1 Central Hospital, No. 320, Great Wall North Street, Baoding, Hebei, 071000, China
| | - Linlin Cheng
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100010, China
| | - Xiaoyan Li
- Department of Clinical Laboratory, Baoding First Central HospitalBaoding NO.1 Central Hospital, No. 320, Great Wall North Street, Baoding, Hebei, 071000, China
| | - Shuqin Guo
- Department of Endocrinology, Baoding NO.1 Central Hospital, No. 320, Great Wall North Street, Baoding, Hebei, 071000, China.
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100010, China.
| |
Collapse
|
17
|
Aphale P, Sanap A, Sharma D, Kharat A, Kheur S, Gawade C, Somasundaram I, Bhonde R. Stem Cell Secretome Modulated by Arsenicum album 30C Ameliorates Lipopolysaccharide-induced Cytokine Storm in Blood Mononuclear Cells in vitro. HOMEOPATHY 2024; 113:132-141. [PMID: 38061387 DOI: 10.1055/s-0043-1776039] [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] [Indexed: 07/28/2024]
Abstract
BACKGROUND The therapeutic effectiveness of mesenchymal stem cells (MSCs) and their secretome can be enhanced by means of physical, chemical and biological preconditioning. Arsenicum album 30C (AA30) has been one of the leading homeopathic medicines used in prophylaxis against SARS-CoV-2 infection. AIMS This study aimed to investigate whether AA30 preconditioning could influence the growth factors and cytokine profile of the human dental pulp-derived MSC (DPD-MSC) secretome. Also, to test the efficacy of the AA30-preconditioned DPD-MSC secretome in ameliorating the lipopolysaccharide (LPS)-induced cytokine storm in human peripheral blood mononuclear cells (PBMCs) as an in-vitro cellular model. METHODS The cytotoxicity of AA30 was assessed in DPD-MSCs by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Growth factors and cytokine levels in the AA30-preconditioned DPD-MSC secretome were analysed by fluorescence-activated cell sorting (FACS) analysis. The angiogenic potential of the AA30-preconditioned DPD-MSC secretome was assessed by chick yolk-sac membrane (YSM) assay. Culture medium with 0.001% ethanol was used as vehicle control. The efficacy of the AA30-preconditioned DPD-MSC secretome in ameliorating the cytokine storm was assessed in LPS pre-treated PBMCs. The mRNA and protein expression of inflammatory markers such as IL-1β, IL-6 and IL-10 were analysed by using RT-PCR and FACS analysis respectively. RESULTS AA30 did not exhibit cytotoxicity in the concentration range of 1% to 50%. Furthermore, the AA30-preconditioned DPD-MSC secretome exhibited a significant increase in the levels of angiogenic factors, such as human angiopoietin-2, EPO and PDGF-AA, and decreased levels of cytokines, such as TNF-α, CXCL-8 and IL-6. The AA30-preconditioned DPD-MSC secretome showed augmented angiogenesis compared to vehicle controls. The DPD-MSC secretome ameliorated LPS-induced mRNA and protein expression of IL-1β, IL-6 and IL-10 in PBMCs. CONCLUSION The AA30-preconditioned DPD-MSC secretome augmented angiogenesis and ameliorated the LPS-induced cytokine storm in human PBMCs in vitro. Our data demonstrate that AA30 preconditioning enhances the therapeutic potency of MSCs and their secretome.
Collapse
Affiliation(s)
- Parth Aphale
- Dr D. Y. Patil Homeopathic Medical College and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Dharmendra Sharma
- Dr D. Y. Patil Homeopathic Medical College and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Avinash Kharat
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Supriya Kheur
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Chinmay Gawade
- Dr D. Y. Patil Homeopathic Medical College and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| | - Indumati Somasundaram
- Department of Biotechnology Engineering, Kolhapur Institute of Technology's College of Engineering, Kolhapur, India
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India
| |
Collapse
|
18
|
Gamal RM, Hazem SH, Hamed MF, Abdelaziz RR. PI3K inhibitor "alpelisib" alleviates methotrexate induced liver injury in mice and potentiates its cytotoxic effect against MDA-MB-231 triple negative breast cancer cell line. Toxicol Appl Pharmacol 2024; 488:116979. [PMID: 38797265 DOI: 10.1016/j.taap.2024.116979] [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: 03/18/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Hepatotoxicity is the main off-target effect of methotrexate (MTX) limiting its effective clinical use. Besides, MDA-MB231 breast cancer cells show chemoresistance, partly via PI3K/AKT pathway. Therefore, we investigated the ameliorative potentials of the PI3K inhibitor, alpelisib (ALP) on MTX-induced hepatotoxicity (in vivo) and the restraining potentials of ALP on MDA-MB231 chemoresistance to MTX (in vitro). Twenty-eight male BALB/c mice were divided into 4 groups. In treatment groups, mice were administered ALP (2.5 and 5 mg/kg) for 5 days and MTX (20 mg/kg) from day 2 till day 5. The results showed that ALP restored hepatic architecture, reduced immune cell infiltration (F4/80, Ly6G and MPO) and repressed the rise in liver enzymes (AST and ALT) induced by MTX. Additionally, ALP rectified the MTX-induced disruption of cellular oxidant status by boosting antioxidant defense systems (HO-1 and GSH) and repressing lipid peroxidation (MDA and 4-HNE). Finally, ALP curbed MTX-induced hepatocyte apoptosis (NF-κB and BAX) and shifted the cytokine milieu away from inflammation (IL-17, IL-22, IL-6 and IL- 10). The results of the in vitro experiments revealed that ALP alone and in combination with MTX, synergistically, reduced cancer cell viability (MTT assay), migration (wound healing assay) and their capacity to establish colonies (colony formation assay) as compared to MTX alone. RT-PCR revealed the antiproliferative (Bcl-2) and proapoptotic (BAX) potentials of ALP and ALP/MTX combination especially after 24 h. In conclusion, targeting PI3K/AKT pathway is a promising strategy in triple negative breast cancer patients by ameliorating hepatotoxicity and restraining chemoresistance to chemotherapy.
Collapse
Affiliation(s)
- Rana M Gamal
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Sara H Hazem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Mohamed F Hamed
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Rania R Abdelaziz
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
19
|
Parmar R, Pickering H, Ahn R, Rossetti M, Gjertson DW, Ruffin F, Chan LC, Fowler VG, Yeaman MR, Reed EF. Integrated transcriptomic analysis reveals immune signatures distinguishing persistent versus resolving outcomes in MRSA bacteremia. Front Immunol 2024; 15:1373553. [PMID: 38846955 PMCID: PMC11153731 DOI: 10.3389/fimmu.2024.1373553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Staphylococcus aureus bacteremia (SAB) is a life-threatening infection particularly involving methicillin-resistant S. aureus (MRSA). In contrast to resolving MRSA bacteremia (RB), persistent MRSA bacteremia (PB) blood cultures remain positive despite appropriate antibiotic treatment. Host immune responses distinguishing PB vs. RB outcomes are poorly understood. Here, integrated transcriptomic, IL-10 cytokine levels, and genomic analyses sought to identify signatures differentiating PB vs. RB outcomes. Methods Whole-blood transcriptomes of propensity-matched PB (n=28) versus RB (n=30) patients treated with vancomycin were compared in one independent training patient cohort. Gene expression (GE) modules were analyzed and prioritized relative to host IL-10 cytokine levels and DNA methyltransferase-3A (DNMT3A) genotype. Results Differential expression of T and B lymphocyte gene expression early in MRSA bacteremia discriminated RB from PB outcomes. Significant increases in effector T and B cell signaling pathways correlated with RB, lower IL-10 cytokine levels and DNMT3A heterozygous A/C genotype. Importantly, a second PB and RB patient cohort analyzed in a masked manner demonstrated high predictive accuracy of differential signatures. Discussion Collectively, the present findings indicate that human PB involves dysregulated immunity characterized by impaired T and B cell responses associated with excessive IL-10 expression in context of the DNMT3A A/A genotype. These findings reveal distinct immunologic programs in PB vs. RB outcomes, enable future studies to define mechanisms by which host and/or pathogen drive differential signatures and may accelerate prediction of PB outcomes. Such prognostic assessment of host risk could significantly enhance early anti-infective interventions to avert PB and improve patient outcomes.
Collapse
Affiliation(s)
- Rajesh Parmar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Richard Ahn
- Department of Microbiology, Immunology, & Molecular Genetics, University of California Los Angeles, Los Angeles, CA, United States
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - David W. Gjertson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University, Durham, NC, United States
| | - Liana C. Chan
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Divisions of Molecular Medicine and Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, United States
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Vance G. Fowler
- Division of Infectious Diseases, Duke University, Durham, NC, United States
| | - Michael R. Yeaman
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Divisions of Molecular Medicine and Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, United States
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
20
|
Hamdorf M, Imhof T, Bailey-Elkin B, Betz J, Theobald SJ, Simonis A, Di Cristanziano V, Gieselmann L, Dewald F, Lehmann C, Augustin M, Klein F, Alejandre Alcazar MA, Rongisch R, Fabri M, Rybniker J, Goebel H, Stetefeld J, Brachvogel B, Cursiefen C, Koch M, Bock F. The unique ORF8 protein from SARS-CoV-2 binds to human dendritic cells and induces a hyper-inflammatory cytokine storm. J Mol Cell Biol 2024; 15:mjad062. [PMID: 37891014 PMCID: PMC11181941 DOI: 10.1093/jmcb/mjad062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 02/01/2023] [Accepted: 10/26/2023] [Indexed: 10/29/2023] Open
Abstract
The novel coronavirus pandemic, first reported in December 2019, was caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 infection leads to a strong immune response and activation of antigen-presenting cells, which can elicit acute respiratory distress syndrome (ARDS) characterized by the rapid onset of widespread inflammation, the so-called cytokine storm. In response to viral infections, monocytes are recruited into the lung and subsequently differentiate into dendritic cells (DCs). DCs are critical players in the development of acute lung inflammation that causes ARDS. Here, we focus on the interaction of a specific SARS-CoV-2 open reading frame protein, ORF8, with DCs. We show that ORF8 binds to DCs, causes pre-maturation of differentiating DCs, and induces the secretion of multiple proinflammatory cytokines by these cells. In addition, we identified DC-SIGN as a possible interaction partner of ORF8 on DCs. Blockade of ORF8 leads to reduced production of IL-1β, IL-6, IL-12p70, TNF-α, MCP-1 (also named CCL2), and IL-10 by DCs. Therefore, a neutralizing antibody blocking the ORF8-mediated cytokine and chemokine response could be an improved therapeutic strategy against SARS-CoV-2.
Collapse
Affiliation(s)
- Matthias Hamdorf
- Cornea Lab Experimental Ophthalmology, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Thomas Imhof
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Institute for Experimental Dentistry and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Ben Bailey-Elkin
- Department of Microbiology, University of Manitoba, Winnipeg MB R3B 2E9 Manitoba, Canada
| | - Janina Betz
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Institute for Experimental Dentistry and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Sebastian J Theobald
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Alexander Simonis
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Veronica Di Cristanziano
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, 50935 Cologne, Germany
| | - Lutz Gieselmann
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, 50935 Cologne, Germany
| | - Felix Dewald
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, 50935 Cologne, Germany
| | - Clara Lehmann
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Max Augustin
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, 50935 Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Miguel A Alejandre Alcazar
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Department of Children and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster Stress Responses in Aging-associated Diseases, 50931 Cologne, Germany
- Institute for Lung Health (ILH), Universities of Gießen and Marburg Lung Centre, Member of the German Center for Lung Research, 35392 Gießen, Germany
| | - Robert Rongisch
- Dermatology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Mario Fabri
- Dermatology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Jan Rybniker
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Heike Goebel
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Jörg Stetefeld
- Department of Microbiology, University of Manitoba, Winnipeg MB R3B 2E9 Manitoba, Canada
| | - Bent Brachvogel
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Claus Cursiefen
- Cornea Lab Experimental Ophthalmology, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Manuel Koch
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Institute for Experimental Dentistry and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Felix Bock
- Cornea Lab Experimental Ophthalmology, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
21
|
Islam F, Habib S, Badruddza K, Rahman M, Islam MR, Sultana S, Nessa A. The Association of Cytokines IL-2, IL-6, TNF-α, IFN-γ, and IL-10 With the Disease Severity of COVID-19: A Study From Bangladesh. Cureus 2024; 16:e57610. [PMID: 38707035 PMCID: PMC11069400 DOI: 10.7759/cureus.57610] [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: 04/04/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction Clinically, the early prediction of the severity of COVID-19 is often challenging, as a dramatic change in severity can occur without warning. The severity of COVID-19 disease is associated with an increased level of inflammatory mediators, including cytokines. This study aimed to evaluate the association of the levels of cytokines interleukin (IL)-2, IL-6, tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), and IL-10 with the severity of COVID-19 in Bangladesh. Materials and methods This cross-sectional study included a total of 60 confirmed cases of COVID-19, comprising 30 severe cases (Group A) and 30 non-severe cases (Group B), and 10 healthy individuals (Group C) attending Bangabandhu Sheikh Mujib Medical University (BSMMU) from March 2021 to February 2022. The cytokine assay was performed using the human Th1/Th2/Th17 cytokine kit BD cytometric bead array (CBA) on the BD Accuri C6 Plus flow cytometer. Statistical analysis was conducted using IBM SPSS Statistics for Windows, Version 23 (Released 2015; IBM Corp., Armonk, New York). Results The mean ages of the patients in Groups A, B, and C were 60.73±5.97, 57.13±7.68, and 48.10±9.13 years, respectively, with a male predominance in all groups. The mean IL-2, IL-6, IL-10, and TNF-α levels had a positive correlation with the increased age group and male gender, although it was not statistically significant. The mean IL-6 and IFN-γ levels were significantly higher among severe cases (216.95±147.78 and 0.98±0.95 pg/mL, respectively) compared to non-severe cases (94.29±128.79 and 0.41±0.61 pg/mL, respectively) and healthy individuals (1.08±1.97 and 0.15±0.28 pg/mL, respectively). Furthermore, the anti-inflammatory cytokine IL-10 was also significantly higher among severe cases (17.92±21.87 pg/mL) compared to non-severe cases (5.38±6.73 pg/mL) and healthy individuals (1.62±1.65 pg/mL). Conclusion IL-6, IFN-γ, and IL-10 have a significant association with the severity of COVID-19 disease. Clinicians treating patients with COVID-19 can consider the level of these cytokines as biomarkers of severity.
Collapse
Affiliation(s)
- Farzana Islam
- Division of Laboratory Service, Sheikh Hasina National Institute of Burn and Plastic Surgery, Dhaka, BGD
| | - Shahriar Habib
- Department of Microbiology, Sher-E-Bangla Medical College, Barishal, BGD
| | - Khaja Badruddza
- Department of Biochemistry, Sher-E-Bangla Medical College, Barishal, BGD
| | - Mahatabur Rahman
- Department of Gastroenterology, Sheikh Russel National Gastro-Liver Institute, Dhaka, BGD
| | - Mohammad R Islam
- Department of Anesthesia, Mainamoti Medical College and Hospital, Cumilla, BGD
| | - Sharmin Sultana
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Afzalun Nessa
- Department of Virology, Bangabandhu Sheikh Mujjib Medical University, Dhaka, BGD
| |
Collapse
|
22
|
Al-Qahtani AA, Alhamlan FS, Al-Qahtani AA. Pro-Inflammatory and Anti-Inflammatory Interleukins in Infectious Diseases: A Comprehensive Review. Trop Med Infect Dis 2024; 9:13. [PMID: 38251210 PMCID: PMC10818686 DOI: 10.3390/tropicalmed9010013] [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: 10/10/2023] [Revised: 11/29/2023] [Accepted: 12/10/2023] [Indexed: 01/23/2024] Open
Abstract
Interleukins (ILs) are signaling molecules that are crucial in regulating immune responses during infectious diseases. Pro-inflammatory ILs contribute to the activation and recruitment of immune cells, whereas anti-inflammatory ILs help to suppress excessive inflammation and promote tissue repair. Here, we provide a comprehensive overview of the role of pro-inflammatory and anti-inflammatory ILs in infectious diseases, with a focus on the mechanisms underlying their effects, their diagnostic and therapeutic potential, and emerging trends in IL-based therapies.
Collapse
Affiliation(s)
- Arwa A. Al-Qahtani
- Department of Family Medicine, College of Medicine, Al-Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Fatimah S. Alhamlan
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| | - Ahmed Ali Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia;
- Department of Microbiology and Immunology, College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia
| |
Collapse
|
23
|
Liao CH, Lai TY, Lin YY, Liao YC, Lai GM, Chu TH, Wu SY, Tsai WL, Whang-Peng J, Liu F, Chiou TJ, Yao CJ. Inhibition of Polyinosinic-Polycytidylic Acid-Induced Acute Pulmonary Inflammation and NF-κB Activation in Mice by a Banana Plant Extract. Int J Med Sci 2024; 21:107-122. [PMID: 38164360 PMCID: PMC10750330 DOI: 10.7150/ijms.88748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/17/2023] [Indexed: 01/03/2024] Open
Abstract
NF-κB activation is pivotal for the excess inflammation causing the critical condition and mortality of respiratory viral infection patients. This study was aimed to evaluate the effect of a banana plant extract (BPE) on suppressing NF-κB activity and acute lung inflammatory responses in mice induced by a synthetic double-stranded RNA viral mimetic, polyinosinic-polycytidylic acid (poly (I:C)). The inflammatory responses were analyzed by immunohistochemistry and HE stains and ELISA. The NF-κB activities were detected by immunohistochemistry in vivo and immunofluorescence and Western blot in vitro. Results showed that BPE significantly decreased influx of immune cells (neutrophils, lymphocytes, and total WBC), markedly suppressed the elevation of pro-inflammatory cytokines and chemokines (IL-6, RANTES, IFN-γ, MCP-1, keratinocyte-derived chemokine, and IL-17), and restored the diminished anti-inflammatory IL-10 in the bronchoalveolar lavage fluid (BALF) of poly (I:C)-stimulated mice. Accordingly, HE staining revealed that BPE treatment alleviated poly (I:C)-induced inflammatory cell infiltration and histopathologic changes in mice lungs. Moreover, immunohistochemical analysis showed that BPE reduced the pulmonary IL-6, CD11b (macrophage marker), and nuclear NF-κB p65 staining intensities, whilst restored that of IL-10 in poly (I:C)-stimulated mice. In vitro, BPE antagonized poly(I:C)-induced elevation of IL-6, nitric oxide, reactive oxygen species, NF-κB p65 signaling, and transient activation of p38 MAPK in human lung epithelial-like A549 cells. Taken together, BPE ameliorated viral mimic poly(I:C)-induced acute pulmonary inflammation in mice, evidenced by reduced inflammatory cell infiltration and regulation of both pro- and anti-inflammatory cytokines. The mechanism of action might closely associate with NF-κB signaling inhibition.
Collapse
Affiliation(s)
- Chien-Huang Liao
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Tung-Yuan Lai
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan
- Traditional Chinese Medicine Cancer Center, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan
| | - Yu-Ying Lin
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Yi-Chun Liao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Gi-Ming Lai
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Tien-Hua Chu
- Chimera Bioscience Inc., No. 18 Siyuan St., Zhongzheng Dist., Taipei 10087, Taiwan
| | - Szu-Yao Wu
- Chimera Bioscience Inc., No. 18 Siyuan St., Zhongzheng Dist., Taipei 10087, Taiwan
| | - Wei-Lun Tsai
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Jacqueline Whang-Peng
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Frank Liu
- Department of Research and Development, Natural Well Technical Company, Guishan, Taoyuan 33377, Taiwan
| | - Tzeon-Jye Chiou
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chih-Jung Yao
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
24
|
Chirumbolo S, Pandolfi S, Franzini M, Valdenassi L. Cytokine profiles in COVID-19 patients undergoing adjunct ozone therapy: some comments. Inflammopharmacology 2023; 31:3363-3365. [PMID: 37831391 DOI: 10.1007/s10787-023-01359-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023]
Abstract
A comment to: Ghaleh HEG, Izadi M, Javanbakht M, Ghanei M, Einollahi B, Jafari NJ, Alishiri G, Aslani J, Abolghasemi H, Simonetti V, Khafaei M, Zhao S, Saadat SH, Ahmadi M, Parvin S, Vazifedoust S, Alvanagh AG. Cytokine profile and antioxidants status in the moderate and severe COVID-19 patients: a trial of ozone therapy impact as a medicinal supplement. Inflammopharmacology. 2023 Jul 12. https://doi.org/10.1007/s10787 -023-01288-9.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Engineering for Innovation Medicine, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy.
| | - Sergio Pandolfi
- Italian Society of Oxygen-Ozone Therapy (SIOOT), Bergamo, Italy
| | | | | |
Collapse
|
25
|
Assar S, Dastbaz M, Amini K, Roghani SA, Lotfi R, Taghadosi M, Kafi H, Abdan Z, Allahyari H, Rostampour R, Shahrokhvand SZ. Assessing the gene expression of the adenosine 5'-monophosphate-activated protein kinase (AMPK) and its relation with the IL-6 and IL-10 plasma levels in COVID-19 patients. Mol Biol Rep 2023; 50:9925-9933. [PMID: 37874507 DOI: 10.1007/s11033-023-08835-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/19/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Metabolic dysregulation and excessive inflammation are implicated in the pathogenesis of the highly infectious disease of coronavirus disease 2019 (COVID-19), which is caused by a newly emerging coronavirus (i.e., severe acute respiratory syndrome-coronavirus 2; SARS-CoV-2). The adenosine 5'-monophosphate-activated protein kinase (AMPK), an energy sensor regulating the metabolic pathways in diverse cells, exerts a regulatory role in the immune system. This study aims to examine the mRNA expression level of AMPK and the plasma levels of interleukin-6 (IL-6) and IL-10 cytokines in patients with different grades of COVID-19. METHODS Peripheral blood was collected from 60 patients with COVID-19 (Moderate, severe, and critical). The plasma levels of IL-6 and IL-10 were quantified by enzyme-linked immunosorbent assay (ELISA), and the mRNA expression level of AMPK was determined using real-time PCR. RESULTS The results showed that the plasma levels of IL-6 increased significantly in critical and severe patients compared to moderate cases of COVID-19 (P < 0.001). Moreover, IL-10 plasma concentrations were significantly higher in critical and severe cases than in moderate cases of COVID-19 (P < 0.01 and P < 0.05, respectively). Also, the gene expression of AMPK was meaningfully enhanced in critical patients relative to moderate and severe cases of COVID-19, in order (P < 0.001 and P < 0.01, respectively). There was a positive association between AMPK gene expression and plasma levels of IL-6 and IL-10 (P = 0.006, r = 0.348, P = 0.028, r = 0.283, respectively). CONCLUSION Increasing AMPK gene expression is likely a necessary effort of the immune system to inhibit inflammation in critical COVID-19. However, this effort seems to be inadequate, probably due to factors that induce inflammation, like erythrocyte sedimentation rate (ESR) and IL-6.
Collapse
Affiliation(s)
- Shirin Assar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Dastbaz
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Komail Amini
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Askar Roghani
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ramin Lotfi
- Clinical Research Development Center, Tohid Hospital, Kurdistan University of Medical Sciences, Sanandaj, Iran.
- Lung Diseases and Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, 6617713446, Iran.
| | - Mahdi Taghadosi
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamidreza Kafi
- Medical Department, Orchid Pharmed Company, Tehran, Iran
| | - Zahra Abdan
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hosna Allahyari
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Rezvan Rostampour
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyedeh Zahra Shahrokhvand
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
26
|
Pencheva M, Bozhkova M, Kalchev Y, Petrov S, Baldzhieva A, Kalfova T, Dichev V, Keskinova D, Genova S, Atanasova M, Murdzheva M. The Serum ACE2, CTSL, AngII, and TNFα Levels after COVID-19 and mRNA Vaccines: The Molecular Basis. Biomedicines 2023; 11:3160. [PMID: 38137381 PMCID: PMC10741205 DOI: 10.3390/biomedicines11123160] [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: 10/19/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND The SARS-CoV-2 virus as well as the COVID-19 mRNA vaccines cause an increased production of proinflammatory cytokines. AIM We investigated the relationship between ACE2, CTSL, AngII, TNFα and the serum levels of IL-6, IL-10, IL-33, IL-28A, CD40L, total IgM, IgG, IgA and absolute count of T- and B-lymphocytes in COVID-19 patients, vaccinees and healthy individuals. METHODS We measured the serum levels ACE2, AngII, CTSL, TNFα and humoral biomarkers (CD40L, IL-28A, IL-10, IL-33) by the ELISA method. Immunophenotyping of lymphocyte subpopulations was performed by flow cytometry. Total serum immunoglobulins were analyzed by the turbidimetry method. RESULTS The results established an increase in the total serum levels for ACE2, CTSL, AngII and TNFα by severely ill patients and vaccinated persons. The correlation analysis described a positive relationship between ACE2 and proinflammatory cytokines IL-33 (r = 0.539) and CD40L (r = 0.520), a positive relationship between AngII and CD40L (r = 0.504), as well as between AngII and IL-33 (r = 0.416), and a positive relationship between CTSL, total IgA (r = 0.437) and IL-28A (r = 0.592). Correlation analysis confirmed only two of the positive relationships between TNFα and IL-28A (r = 0.491) and CD40L (r = 0.458). CONCLUSIONS In summary, the findings presented in this study unveil a complex web of interactions within the immune system in response to SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- Mina Pencheva
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Martina Bozhkova
- Department of Medical Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.B.); (Y.K.); (S.P.); (A.B.); (T.K.); (M.A.); (M.M.)
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Yordan Kalchev
- Department of Medical Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.B.); (Y.K.); (S.P.); (A.B.); (T.K.); (M.A.); (M.M.)
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Steliyan Petrov
- Department of Medical Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.B.); (Y.K.); (S.P.); (A.B.); (T.K.); (M.A.); (M.M.)
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Alexandra Baldzhieva
- Department of Medical Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.B.); (Y.K.); (S.P.); (A.B.); (T.K.); (M.A.); (M.M.)
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Teodora Kalfova
- Department of Medical Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.B.); (Y.K.); (S.P.); (A.B.); (T.K.); (M.A.); (M.M.)
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Valentin Dichev
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
- Department of Medical Biology, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Donka Keskinova
- Department of Applied and Institutional Sociology, Faculty of Philosophy and History, University of Plovdiv “Paisii Hilendarski”, 4000 Plovdiv, Bulgaria;
| | - Silvia Genova
- Department of General and Clinical Pathology, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Mariya Atanasova
- Department of Medical Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.B.); (Y.K.); (S.P.); (A.B.); (T.K.); (M.A.); (M.M.)
- Laboratory of Virology, UMBAL “St. George” EAD, 4002 Plovdiv, Bulgaria
| | - Mariana Murdzheva
- Department of Medical Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.B.); (Y.K.); (S.P.); (A.B.); (T.K.); (M.A.); (M.M.)
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| |
Collapse
|
27
|
Ouyang P, Tao Y, Wei W, Li Q, Liu S, Ren Y, Huang X, Chen D, Geng Y. Spring Viremia of Carp Virus Infection Induces Carp IL-10 Expression, Both In Vitro and In Vivo. Microorganisms 2023; 11:2812. [PMID: 38004823 PMCID: PMC10673272 DOI: 10.3390/microorganisms11112812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/28/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Interleukin-10 (IL-10) is a pleiotropic cytokine with both immune enhancement and immunosuppression activities, but the main role is immunosuppression and anti-inflammatory ability. In order to use the immunosuppressive function of IL-10, many viruses, such as SARS-CoV-2, hepatitis B virus and EB virus, can evade the host's immune surveillance and clearance by increasing the expression of host IL-10. However, it has not been reported whether the aquatic animal infection virus can upregulate the expression of host IL-10 and the mechanisms are still unknown. Spring viremia of carp (SVC) is a fatal viral disease for many fish species and is caused by spring viremia of carp virus (SVCV). This disease has caused significant economic losses in the aquaculture industry worldwide. In this study, the expression of carp IL-10 with or without infection of SVCV in epithelioma papulosum cyprinid (EPC) cells, carp head kidney (cHK) primary cells and common carp tissues were analyzed using RT-PCR and ELISA. The results show that SVCV infection induced carp IL-10 mRNA and protein expression, both in vitro and in vivo. However, the upregulation of carp IL-10 by SVCV was hindered by specific inhibitors of the JAK inhibitor (CP-690550), STAT3 inhibitor (STA-21), NF-κB inhibitor (BAY11-7082) and p38 MAPK (mitogen-activated protein kinase) inhibitor (SB202190), but not JNK inhibitor (SP600125). Furthermore, the results demonstrated that JAK1, JAK2, JAK3, TYK2 and STAT5 played important roles in carp IL-10 production induced by SVCV infection. Taken together, SVCV infection significantly induced carp IL-10 expression and the upregulation trigged in JAK-STAT, NF-κB and p38MAPK pathways. To our knowledge, this is the first time that a fish infection virus upregulated the host IL-10 expression through the JAK-STAT, NF-κB and p38MAPK pathways. Altogether, fish viruses may have a similar mechanism as human or other mammalian viruses to escape host immune surveillance and clearance.
Collapse
Affiliation(s)
- Ping Ouyang
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Q.L.); (S.L.); (Y.R.); (Y.G.)
| | - Yu Tao
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Q.L.); (S.L.); (Y.R.); (Y.G.)
| | - Wenyan Wei
- Chengdu Academy of Agriculture and Forestry Sciences, Chengdu 611130, China;
| | - Qiunan Li
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Q.L.); (S.L.); (Y.R.); (Y.G.)
| | - Shuya Liu
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Q.L.); (S.L.); (Y.R.); (Y.G.)
| | - Yongqiang Ren
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Q.L.); (S.L.); (Y.R.); (Y.G.)
| | - Xiaoli Huang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.H.); (D.C.)
| | - Defang Chen
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (X.H.); (D.C.)
| | - Yi Geng
- Department of Basic Veterinary, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Q.L.); (S.L.); (Y.R.); (Y.G.)
| |
Collapse
|
28
|
Zu C, Wu S, Zhang M, Wei G, Xu H, Cui J, Chang AH, Huang H, Hu Y. A distinct cytokine network distinguishes chimeric antigen receptor T cell (CAR-T)-associated hemophagocytic lymphohistiocytosis-like toxicity (carHLH) from severe cytokine release syndrome following CAR-T therapy. Cytotherapy 2023; 25:1167-1175. [PMID: 37480884 DOI: 10.1016/j.jcyt.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/24/2023] [Accepted: 06/27/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND AIMS With the increasing application of chimeric antigen receptor (CAR)-T cell therapy in various malignancies, an extra toxicity profile has been revealed, including a severe complication resembling hemophagocytic lymphohistiocytosis (HLH), which is usually disguised by severe cytokine release syndrome (CRS). METHODS In a clinical trial in whom 99 patients received B-cell maturation antigen CAR-T cells, we identified 20 (20.20%) cases of CAR-T cell-associated HLH (carHLH), most of whom possessed a background of severe CRS (grade ≥3). The overlapping features of carHLH and severe CRS attracted us to further explore the differences between them. RESULTS We showed that carHLH can be distinguished by extreme elevation of interferon-γ, granzyme B, interleukin-1RA and interleukin-10, which can be informative in developing prevention and management strategies of this toxicity. Moreover, we developed a predictive model of carHLH with a mean area under the curve of 0.81 ± 0.07, incorporating serum lactate dehydrogenase at day 6 post-CRS and serum fibrinogen at day 3 post-CRS. CONCLUSIONS The incidence of carHLH in CAR-T recipients might be relatively higher than we previously thought. relatively higher than we previously. A cytokine network distinguished from CRS is responsible for carHLH. And corresponding cytokine-directed therapies, especially targeting IL-10, are worth trying.
Collapse
Affiliation(s)
- Cheng Zu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Zhejiang, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Shenghao Wu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hematology, The Dingli Clinical College of Wenzhou Medical University (The Second Affiliated Hospital of Shanghai University, Wenzhou Central Hospital), Wenzhou, China
| | - Mingming Zhang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Zhejiang, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Guoqing Wei
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Zhejiang, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Huijun Xu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Zhejiang, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Jiazhen Cui
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Zhejiang, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Alex H Chang
- Shanghai YaKe Biotechnology Ltd., Shanghai, China; Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Zhejiang, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China.
| | - Yongxian Hu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China; Institute of Hematology, Zhejiang University, Zhejiang, China; Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China.
| |
Collapse
|
29
|
Chiu MH, Gershkovich B, Yu IL, O'Brien ER, Deng J, McDonald B. Heat shock protein 27 in the pathogenesis of COVID-19 and non-COVID acute respiratory distress syndrome. Cell Stress Chaperones 2023; 28:877-887. [PMID: 37966617 PMCID: PMC10746647 DOI: 10.1007/s12192-023-01381-6] [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: 01/29/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 11/16/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common cause of hypoxemic respiratory failure in intensive care units that has increased dramatically as a result of the COVID-19 pandemic. In both COVID-19 and non-COVID ARDS, the pathogenesis of lung injury involves local (pulmonary) and systemic inflammation, leading to impaired gas exchange, requirement for mechanical ventilation, and a high risk of mortality. Heat shock protein 27 (HSP27) is a chaperone protein expressed in times of cell stress with roles in modulation of systemic inflammation via the NF-κB pathway. Given its important role as a modulator of inflammation, we sought to investigate the role of HSP27 and its associated auto-antibodies in ARDS caused by both SARS-CoV-2 and non-COVID etiologies. A total of 68 patients admitted to the intensive care unit with ARDS requiring mechanical ventilation were enrolled in a prospective, observational study that included 22 non-COVID-19 and 46 COVID-19 patients. Blood plasma levels of HSP27, anti-HSP27 auto-antibody (AAB), and cytokine profiles were measured on days 1 and 3 of ICU admission along with clinical outcome measures. Patients with COVID-19 ARDS displayed significantly higher levels of HSP27 in plasma, and a higher ratio of HSP27:AAB on both day 1 and day 3 of ICU admission. In patients with COVID-19, higher levels of circulating HSP27 and HSP27:AAB ratio were associated with a more severe systemic inflammatory response and adverse clinical outcomes including more severe hypoxemic respiratory failure. These findings implicate HSP27 as a marker of advanced pathogenesis of disease contributing to the dysregulated systemic inflammation and worse clinical outcomes in COVID-19 ARDS, and therefore may represent a potential therapeutic target.
Collapse
Affiliation(s)
- Michael H Chiu
- Libin Cardiovascular Institute of Alberta, Department of Cardiac Sciences, University of Calgary, Calgary, Canada.
- Department of Critical Care Medicine, University of Calgary, Calgary, Canada.
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | | | - Ian-Ling Yu
- Department of Critical Care Medicine, University of Calgary, Calgary, Canada
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Edward R O'Brien
- Libin Cardiovascular Institute of Alberta, Department of Cardiac Sciences, University of Calgary, Calgary, Canada
| | - Jingti Deng
- Libin Cardiovascular Institute of Alberta, Department of Cardiac Sciences, University of Calgary, Calgary, Canada
| | - Braedon McDonald
- Department of Critical Care Medicine, University of Calgary, Calgary, Canada
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
30
|
Brandenburg K, Ferrer-Espada R, Martinez-de-Tejada G, Nehls C, Fukuoka S, Mauss K, Weindl G, Garidel P. A Comparison between SARS-CoV-2 and Gram-Negative Bacteria-Induced Hyperinflammation and Sepsis. Int J Mol Sci 2023; 24:15169. [PMID: 37894850 PMCID: PMC10607443 DOI: 10.3390/ijms242015169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Sepsis is a life-threatening condition caused by the body's overwhelming response to an infection, such as pneumonia or urinary tract infection. It occurs when the immune system releases cytokines into the bloodstream, triggering widespread inflammation. If not treated, it can lead to organ failure and death. Unfortunately, sepsis has a high mortality rate, with studies reporting rates ranging from 20% to over 50%, depending on the severity and promptness of treatment. According to the World Health Organization (WHO), the annual death toll in the world is about 11 million. One of the main toxins responsible for inflammation induction are lipopolysaccharides (LPS, endotoxin) from Gram-negative bacteria, which rank among the most potent immunostimulants found in nature. Antibiotics are consistently prescribed as a part of anti-sepsis-therapy. However, antibiotic therapy (i) is increasingly ineffective due to resistance development and (ii) most antibiotics are unable to bind and neutralize LPS, a prerequisite to inhibit the interaction of endotoxin with its cellular receptor complex, namely Toll-like receptor 4 (TLR4)/MD-2, responsible for the intracellular cascade leading to pro-inflammatory cytokine secretion. The pandemic virus SARS-CoV-2 has infected hundreds of millions of humans worldwide since its emergence in 2019. The COVID-19 (Coronavirus disease-19) caused by this virus is associated with high lethality, particularly for elderly and immunocompromised people. As of August 2023, nearly 7 million deaths were reported worldwide due to this disease. According to some reported studies, upregulation of TLR4 and the subsequent inflammatory signaling detected in COVID-19 patients "mimics bacterial sepsis". Furthermore, the immune response to SARS-CoV-2 was described by others as "mirror image of sepsis". Similarly, the cytokine profile in sera from severe COVID-19 patients was very similar to those suffering from the acute respiratory distress syndrome (ARDS) and sepsis. Finally, the severe COVID-19 infection is frequently accompanied by bacterial co-infections, as well as by the presence of significant LPS concentrations. In the present review, we will analyze similarities and differences between COVID-19 and sepsis at the pathophysiological, epidemiological, and molecular levels.
Collapse
Affiliation(s)
- Klaus Brandenburg
- Brandenburg Antiinfektiva, c/o Forschungszentrum Borstel, Leibniz-Lungenzentrum, Parkallee 10, 23845 Borstel, Germany; (K.B.); (K.M.)
| | - Raquel Ferrer-Espada
- Department of Microbiology, University of Navarra, IdiSNA (Navarra Institute for Health Research), Irunlarrea 1, E-31008 Pamplona, Spain;
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Guillermo Martinez-de-Tejada
- Department of Microbiology, University of Navarra, IdiSNA (Navarra Institute for Health Research), Irunlarrea 1, E-31008 Pamplona, Spain;
| | - Christian Nehls
- Forschungszentrum Borstel, FG Biophysik, Parkallee 10, 23845 Borstel, Germany;
| | - Satoshi Fukuoka
- National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu 761-0395, Japan;
| | - Karl Mauss
- Brandenburg Antiinfektiva, c/o Forschungszentrum Borstel, Leibniz-Lungenzentrum, Parkallee 10, 23845 Borstel, Germany; (K.B.); (K.M.)
- Sylter Klinik Karl Mauss, Dr.-Nicolas-Strasse 3, 25980 Westerland (Sylt), Germany
| | - Günther Weindl
- Pharmazeutisches Institut, Abteilung Pharmakologie und Toxikologie, Universität Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany;
| | - Patrick Garidel
- Physikalische Chemie, Martin-Luther-Universität Halle-Wittenberg, 06108 Halle (Saale), Germany
| |
Collapse
|
31
|
Andriankaja OM, Adatorwovor R, Kantarci A, Hasturk H, Shaddox L, Levine MA. Periodontal Disease, Local and Systemic Inflammation in Puerto Ricans with Type 2 Diabetes Mellitus. Biomedicines 2023; 11:2770. [PMID: 37893143 PMCID: PMC10604368 DOI: 10.3390/biomedicines11102770] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Periodontal disease (PD) is prevalent in type 2 diabetic condition (T2DM). OBJECTIVES We assessed the associations between serum or gingival crevicular fluid (GCF) endothelial and inflammatory mediators and chronic PD among T2DM Hispanic adults. METHODS We enrolled 248 Puerto Rican residents with T2DM aged 40-65 years. The exposures included serum inflammatory mediators (IL-1b, IL-6, IL-10, and TNF-α), endothelial adhesion molecules, RANKL levels, and the GCF content of these analytes from a subset of 158 samples. The outcomes included the percent of sites with a probing pocket depth (PPD) ≥ 4 mm and clinical attachment loss ≥ 4 mm. Adjusted logistic regression models were fit to the categorized outcomes. RESULTS Increased serum IL-10 (Adj. OR: 1.10, p = 0.04), sICAM-1 (Adj. OR: 1.01; p = 0.06), and elevated serum IL-1β (Adj. OR: 1.93; p = 0.06) were statistically significant or close to being significantly associated with a percent of sites with PPD ≥ 4 mm. An increase in GCF IL-1α (Adj. OR: 1.16; p < 0.01) and IL-1β (Adj: 2.40; p = 0.02) was associated with periodontal parameters. CONCLUSIONS Our findings suggested that oral and systemic endothelial and inflammatory mediators are associated with periodontal clinical parameters among Hispanic adults with T2DM.
Collapse
Affiliation(s)
- Oelisoa M. Andriankaja
- Center for Oral Health Research (COHR), University of Kentucky College of Dentistry, Lexington, KY 40536, USA;
| | - Reuben Adatorwovor
- College of Public Health, University of Kentucky, Lexington, KY 40536, USA;
| | | | - Hatice Hasturk
- Center for Clinical and Translational Research, The Forsyth Institute, Cambridge, MA 02142, USA;
| | - Luciana Shaddox
- Center for Oral Health Research (COHR), University of Kentucky College of Dentistry, Lexington, KY 40536, USA;
| | - Michael A. Levine
- Center for Bone Health, Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| |
Collapse
|
32
|
Rajamanickam A, Nathella PK, Selvaraj N, Manoj M, Thangaraj JWV, Muthusamy SK, Chethrapilly Purushothaman GK, Bhatnagar T, Ponnaiah M, Ramasamy S, Velusamy S, Babu S. Characterization of IL-10 Family of Cytokines in Acute and Convalescent COVID-19 Individuals. J Interferon Cytokine Res 2023; 43:469-477. [PMID: 37708007 DOI: 10.1089/jir.2023.0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Cytokines are major players in orchestrating inflammation, disease pathogenesis, and severity during COVID-19. Members of the interleukin (IL)-10 family of cytokines play important roles in regulating immune responses to various inflammatory and infectious diseases. However, the role of the IL-10 family of cytokines in COVID-19 remains elusive. Hence, we determined the plasma levels of the IL-10 family of cytokines (IL-10, IL-19, IL-20, IL-22, and IL-24) in 7 groups of COVID-19 individuals, based on days since real-time reverse transcriptase-polymerase chain reaction confirmation of SARS-CoV-2 infection. Our data show that the levels of IL-10, IL-19, IL-20, IL-22, and IL-24 cytokines decreased from days 15-30 to days 61-90 and plateaued thereafter. Severe COVID-19 patients exhibit increased plasma levels of IL-10, IL-19, IL-20, IL-22, and IL-24 compared to mild patients. Thus, our study provides evidence of alterations in the plasma levels of the IL-10 family of cytokines in convalescent COVID-19 individuals.
Collapse
Affiliation(s)
- Anuradha Rajamanickam
- National Institutes of Health-International Center for Excellence in Research, Chennai, India
| | | | - Nandhini Selvaraj
- National Institutes of Health-International Center for Excellence in Research, Chennai, India
| | | | | | | | | | | | | | | | | | - Subash Babu
- National Institutes of Health-International Center for Excellence in Research, Chennai, India
| |
Collapse
|
33
|
Kim Y, Park IH, Shin J, Choi J, Jeon C, Jeon S, Shin JS, Jung H. Sublingual Dissolving Microneedle (SLDMN)-Based Vaccine for Inducing Mucosal Immunity against SARS-CoV-2. Adv Healthc Mater 2023; 12:e2300889. [PMID: 37337388 DOI: 10.1002/adhm.202300889] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/07/2023] [Indexed: 06/21/2023]
Abstract
The coronavirus pandemic has accelerated the development of next-generation vaccination technology to combat future pandemic outbreaks. Mucosal vaccination effectively protects the mucosal surfaces, the primary sites of viral entry, by inducing the secretion of immunoglobulin A (IgA) and humoral IgG. Here, a dissolving microneedle (DMN) is adopted as a mucosal vaccine delivery platform to directly penetrate the sublingual site, which is rich in antigen-presenting cells (APCs) and lymphoid tissues. The sublingual dissolving microneedle (SLDMN) vaccination platform comprised a micropillar-based compartment and a 3D-printed SLDMN applicator as a substitute for the DMN patch. The penetration efficacy of SLDMNs is assessed using in vitro optical coherence tomography (OCT) and in vivo histological analysis. The efficacy of SLDMN is also evaluated in a vaccine form using the recombinant spike (S1) protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Furthermore, SLDMN is used to challenge transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) receptors. Its effects are evaluated on antibody production, survival rate, and inflammation attenuation after infection compared to the intramuscular (IM) injections. Overall, SLDMN effectively induced mucosal immunity via IgA secretion, attenuated lung inflammation, and lowered the levels of cytokines and chemokines, which may prevent the "cytokine storm" after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Youseong Kim
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - In Ho Park
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jiwoo Shin
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jaibyung Choi
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chansol Jeon
- JUVIC, 208Ho, 272, Digital-ro, Guro-gu, Seoul, 08389, Republic of Korea
| | - Seonghun Jeon
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyungil Jung
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- JUVIC, 208Ho, 272, Digital-ro, Guro-gu, Seoul, 08389, Republic of Korea
| |
Collapse
|
34
|
Andrejkovits ÁV, Huțanu A, Susányi EJ, Negrea V, Văsieșiu AM. The Prognostic Utility of Cytokines in Hospitalized COVID-19 Patients. J Crit Care Med (Targu Mures) 2023; 9:208-217. [PMID: 37969879 PMCID: PMC10644278 DOI: 10.2478/jccm-2023-0025] [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: 07/08/2023] [Accepted: 09/14/2023] [Indexed: 11/17/2023] Open
Abstract
Introduction The severity of COVID-19 relies on several factors, but the overproduction of pro-inflammatory cytokines remains a central mechanism. The aim of this study was to investigate the predictive utility of interleukin (IL)-6, IL-8, IL-10, IL-12, tumor necrosis factor alpha (TNF-α), and interferon gamma (IFN-γ) measurement in patients with COVID-19. Material and Methods We prospectively enrolled 181 adult patients with COVID-19 admitted to the 1st Infectious Disease County Hospital Târgu Mureș from December 2020 to September 2021. Serum cytokine levels were measured and correlated with disease severity, need for oxygen therapy, intensive care unit (ICU) transfer, and outcome. Results We found significantly higher serum levels of IL-6, IL-8, and IL-10 in patients with severe COVID-19 and in those with a fatal outcome. The logistic regression analysis showed a significant predictive value for IL-8 regarding disease severity, and for IL6 and IL-10 regarding ICU transfer and fatal outcome. Conclusions Serum levels of IL-6, IL-8, and IL-10 were significantly increased in patients with COVID-19, but their predictive value regarding disease severity and the need for oxygen therapy was poor. We found IL-6 and IL-10 to have a good predictive performance regarding ICU transfer and fatal outcome.
Collapse
Affiliation(s)
- Ákos Vince Andrejkovits
- Doctoral School of Medicine and Pharmacy, I.O.S.U.D., George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
| | - Adina Huțanu
- Department of Laboratory Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
- Center for Advanced Medical and Pharmaceutical Research, Targu Mures, Romania
| | - Ervin József Susányi
- First Infectious Disease Clinic of Targu Mureș, Mureș County Clinical Hospital, Romania
| | - Valentina Negrea
- Doctoral School of Medicine and Pharmacy, I.O.S.U.D., George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
- Department of Infectious Disease, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
| | - Anca Meda Văsieșiu
- First Infectious Disease Clinic of Targu Mureș, Mureș County Clinical Hospital, Romania
- Department of Infectious Disease, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
| |
Collapse
|
35
|
Silva BSDA, Pereira T, Minuzzi LG, Padilha CS, Figueiredo C, Olean-Oliveira T, dos Santos IVM, von Ah Morano AE, Marchioto Júnior O, Ribeiro JPJ, Dos Santos VR, Seelaender M, Teixeira AA, Dos Santos RVT, Lemos VDA, Freire APCF, Dorneles GP, Marmett B, Olean-Oliveira A, Teixeira MFS, Seraphim PM, Caseiro A, Pinho RA, Islam H, Little JP, Krüger K, Rosa-Neto JC, Coelho-E-Silva MJ, Lira FS. Mild to moderate post-COVID-19 alters markers of lymphocyte activation, exhaustion, and immunometabolic responses that can be partially associated by physical activity level- an observational sub-analysis fit- COVID study. Front Immunol 2023; 14:1212745. [PMID: 37753077 PMCID: PMC10518618 DOI: 10.3389/fimmu.2023.1212745] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/10/2023] [Indexed: 09/28/2023] Open
Abstract
Aim This study aimed to evaluate if physical activity is associated with systemic and cellular immunometabolic responses, in young adults after mild-to-moderate COVID-19 infection. Methods Mild- to- moderate post-COVID-19 patients (70.50 ± 43.10 days of diagnosis; age: 29.4 (21.9- 34.9) years; BMI: 25.5 ± 4.3 kg m2 n = 20) and healthy age-matched controls (age: 29.3 (21.2 - 32.6) years; BMI: 25.4 ± 4.7 kg m2; n = 20) were evaluated. Physical activity levels (PAL), body composition, dietary habits, muscular and pulmonary function, mental health, sleep quality, metabolic parameters, immune phenotypic characterization, stimulated whole blood and PBMC culture (cytokine production), mRNA, and mitochondrial respiration in PBMCs were evaluated. Results The post-COVID-19 group exhibited lower levels of moderate to vigorous physical activity (MVPA) (p = 0.038); therefore, all study comparisons were performed with adjustment for MVPA. Post-COVID-19 impacted the pulmonary function (FEV1, FEV1%pred, FVC, and FVC %pred) compared with the control (p adjusted by MVPA (p adj) <0.05). Post-COVID-19 exhibited lower levels of serum IL-6 (p adj <0.01), whereas it showed higher serum IL-10, triglyceride, leptin, IgG, ACE activity, TNFRSF1A, and PGE2 (p adj <0.05) levels compared with controls. Post-COVID-19 presented a lower percentage of Treg cells (p adj = 0.03) and altered markers of lymphocyte activation and exhaustion (lower CD28 expression in CD8+ T cells (p adj = 0.014), whereas CD4+T cells showed higher PD1 expression (p adj = 0.037)) compared with the control group. Finally, post- COVID-19 presented an increased LPS-stimulated whole- blood IL-10 concentration (p adj <0.01). When exploring mitochondrial respiration and gene expression in PBMCs, we observed a higher LEAK state value (p adj <0.01), lower OXPHOS activity (complex I) (p adj = 0.04), and expression of the Rev-Erb-α clock mRNA after LPS stimulation in the post-COVID-19 patients than in the control (p adj <0.01). Mainly, PAL was associated with changes in IL-10, triglyceride, and leptin levels in the plasma of post-COVID-19 patients. PAL was also associated with modulation of the peripheral frequency of Treg cells and the expression of PD-1 in CD8+ T cells, although it abrogated the statistical effect in the analysis of TNF-α and IL-6 production by LPS- and PMA-stimulated PBMC of post-COVID-19 patients. Conclusion Young adults after mild-to-moderate SARS-CoV-2 infection appeared to have lower physical activity levels, which can be associated with clinical and immunometabolic responses in a complex manner.
Collapse
Affiliation(s)
- Bruna Spolador de Alencar Silva
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Telmo Pereira
- Polytechnic Institute of Coimbra, Coimbra Health School, Coimbra, Portugal
| | - Luciele Guerra Minuzzi
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Camila Souza Padilha
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Caique Figueiredo
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Tiago Olean-Oliveira
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Ivete Vera Medeiros dos Santos
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Ana Elisa von Ah Morano
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Osmar Marchioto Júnior
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - José Procópio Jabur Ribeiro
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Vanessa Ribeiro Dos Santos
- Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Marília Seelaender
- Cancer Metabolism Research Group, LIM26-HC, FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | - Ana Paula Coelho Figueira Freire
- Department of Health Sciences, Central Washington University, Ellensburg, WA, United States
- Physiotherapy Department, Universidade do Oeste Paulista (UNOESTE), Presidente Prudente, Brazil
| | - Gilson Pires Dorneles
- Laboratory of Cellular and Molecular Immunology, Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Bruna Marmett
- Laboratory of Cellular and Molecular Immunology, Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - André Olean-Oliveira
- Department of Chemistry and Biochemistry, School of Science and Technology, Universidade Estadual Paulista (UNESP), Presidente Prudente, SP, Brazil
| | - Marcos F. S. Teixeira
- Department of Chemistry and Biochemistry, School of Science and Technology, Universidade Estadual Paulista (UNESP), Presidente Prudente, SP, Brazil
| | - Patrícia M. Seraphim
- Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
| | - Armando Caseiro
- Polytechnic Institute of Coimbra, Coimbra Health School, Coimbra, Portugal
| | - Ricardo Aurino Pinho
- Graduate Program in Health Sciences, School of Medicine, Pontifícia Universidade Católica do Parana, Curitiba, Brazil
| | - Hashim Islam
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Jonathan Peter Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - José César Rosa-Neto
- Department of Cell and Developmental Biology, University of São Paulo, São Paulo, Brazil
| | - Manuel-João Coelho-E-Silva
- Faculty of Sport Sciences and Physical Education, Research Center for Sport and Physical Activity (uid/dtp/04213/2020), Universidade de Coimbra, Coimbra, Portugal
| | - Fábio Santos Lira
- Exercise and Immunometabolism Research Group, Postgraduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, Brazil
- Faculty of Sport Sciences and Physical Education, Research Center for Sport and Physical Activity (uid/dtp/04213/2020), Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
36
|
Xia Y, Yang Q, Wu SY, Wu Z, Li Q, Du J. Interferon lambda modulates proinflammatory cytokines production in PBMCs from patients with chronic kidney disease. Hum Immunol 2023; 84:464-470. [PMID: 37394297 DOI: 10.1016/j.humimm.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND CKD is a major cause of morbidity and mortality worldwide. Considerable evidence now indicates that renal inflammation plays a central role in the initiation and progression of CKD. Recent investigations have demonstrated that IFNλ plays an important role in the pathogenesis of autoimmune and inflammatory diseases. However, the association of IFNλ with CKD is still poorly understood. OBJECTIVE To analyze the correlation between IFNλ levels and pro-inflammatory cytokines, and to investigate the effect of IFNλ on PBMCs in patients with CKD. METHODS PBMCs were harvested from patients with CKD and healthy controls for measuring the expression level of inflammatory cytokines by RT-qPCR. Spearman correlation test was used to analyze correlation between IFNλ and cytokines as well as eGFR. PBMCs from healthy individuals and CKD patients were subjected to IFNλ protein stimulation. IL6, TNFα, IL10, ISG15 and MX1 mRNA level were measured by RT-PCR, STAT1 and phosphorylated STAT1 protein level were measured by Western blot. RESULTS Patients with CKD showed higher levels of IFNλ in PBMCs compared to healthy controls. IFNλ mRNA levels were associated with cytokines and eGFR. The transcription of IL6, TNFα, and IL10 was significantly increased in healthy human PBMCs after IFNλ stimulation. In addition, IFNλ acts on PBMCs by p-STAT1 and ISG15 as well as MX1. CONCLUSION High expression of IFNλ was found in CKD patients and was associated with eGFR and disease-related cytokines. More importantly, IFNλ promoted the expression of pro-inflammatory cytokines in PBMCs, suggesting a potential pro-inflammatory role of IFNλ in CKD.
Collapse
Affiliation(s)
- Yuhao Xia
- Weifang Medical University, Shandong, China; Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Qiannan Yang
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Shang Ying Wu
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Zhicheng Wu
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Qian Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Jing Du
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
37
|
Sari MI, Jusuf NK, Munir D, Putra A, Bisri T, Ilyas S, Farhat F, Muhar AM, Rusda M, Amin MM. The Role of Mesenchymal Stem Cell Secretome in the Inflammatory Mediators and the Survival Rate of Rat Model of Sepsis. Biomedicines 2023; 11:2325. [PMID: 37626822 PMCID: PMC10452511 DOI: 10.3390/biomedicines11082325] [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: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
In sepsis, simultaneously elevated levels of pro-inflammatory cytokines and interleukin (IL)-10 indicate immune response dysregulation, increasing the mortality of the host. As mesenchymal stem cell (MSC) secretome is known to have immunomodulatory effects, we aim to assess the role of MSC secretome in the inflammatory mediators (NF-κB p65 and p50, TNF-α, IL-10) and the survival rate of a rat model of sepsis. In this study, forty-eight male Rattus norvegicus rats were divided into one sham group and three groups with sepsis induction: the control group and the sepsis-induced rat groups treated with 150 μL (T1) and 300 μL (T2) of secretome. The survival rate was observed per 6 h for 48 h and plotted using the Kaplan-Meier method. Compared to the control group, T2 showed a significant decrease in the relative expression of NF-κB and the serum TNF-α level, and a significant increase in the serum IL-10 level. Meanwhile, T1 showed a significant decrease in the serum TNF-α level compared to the control group. The Kaplan-Meier Log Rank test did not show significance in the distribution of survival between T1, T2, and the control group. However, from the 18th to the 36th hour, the survival rate of T2 was lower than the survival rate of the control group and T1, with a noticeable difference between T2 and the control group, as well as T1 at the 36th hour. At the 42nd hour, the survival rate of T2 was the same as the control group and remained lower than T1. In conclusion, MSC secretome regulated the inflammatory mediators in rat model of sepsis, with a dose of 150 μL being more effective.
Collapse
Affiliation(s)
- Mutiara Indah Sari
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.R.); (M.M.A.)
| | - Nelva Karmila Jusuf
- Department of Dermatology & Venereology, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Delfitri Munir
- Department of Ear, Nose & Throat, Head & Neck, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (D.M.); (F.F.)
| | - Agung Putra
- Stem Cell and Cancer Research, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang 50112, Indonesia;
| | - Tatang Bisri
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, Universitas Jenderal Achmad Yani, Bandung 40513, Indonesia;
| | - Syafruddin Ilyas
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Farhat Farhat
- Department of Ear, Nose & Throat, Head & Neck, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (D.M.); (F.F.)
| | - Adi Muradi Muhar
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Muhammad Rusda
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.R.); (M.M.A.)
| | - Mustafa Mahmud Amin
- Philosophy Doctor in Medicine Program, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.R.); (M.M.A.)
| |
Collapse
|
38
|
van Heerden PV, Abutbul A, Naama A, Maayan S, Makram N, Nachshon A, abu Jabal K, Hershkovitz O, Binder L, Shabat Y, Reicher B, Mevorach D. Apoptotic cells for treatment of acute respiratory distress syndrome associated with COVID-19. Front Immunol 2023; 14:1242551. [PMID: 37600829 PMCID: PMC10433372 DOI: 10.3389/fimmu.2023.1242551] [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: 06/19/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Background Hyper-inflammatory immune response, a hallmark of severe COVID-19, is associated with increased mortality. Acute respiratory distress syndrome (ARDS) is a common manifestation. We undertook two phase I/II studies in five and then 16 subjects with severe/critical COVID-19 to assess the safety and preliminary efficacy of apoptotic cells (Allocetra™-OTS, Enlivex Therapeutics), a cellular immunomodulatory therapy that reprograms macrophages to reduce hyper-inflammatory response severity. Methods Eligible patients presenting to the Emergency Room with severe COVID-19 and respiratory dysfunction received one intravenous administration of Allocetra™-OTS and were monitored for adverse events (AEs) for 28 days. The primary aim was to determine the safety profile of treatment; secondary aims were recovery from ARDS, intensive care unit (ICU) and hospital length-of-stay, and mortality. Immune modulator markers were measured to elucidate the mechanism of action of Allocetra™-OTS. Results 21 patients with severe-critical COVID-19 of Gamma, Alpha and Delta variants, were treated with a single dose of apoptotic cells. 19/21 patients had mild-to-severe ARDS at presentation. Median age was 53 years, 16/21 were males, 16/21 were overweight/obese. No serious related adverse events (SAEs) were reported. All 21 study subjects survived to day 28 (end of study); 19/21 recovered completely. Comparable mortality rates at the hospital were 3.8%-8.9% for age- and gender-matched patients, and 39%-55% for critical patients. Recovering patients exhibited rapid ARDS resolution and parallel resolution of inflammation markers and elevated cytokines/chemokines. Conclusion In patients with severe/critical COVID-19 associated with ARDS, Allocetra™-OTS was safe, well-tolerated, and showed promising results for resolution of respiratory failure and inflammation. Trial registration https://clinicaltrials.gov/ct2/show/study/NCT04513470, https://clinicaltrials.gov/ct2/show/study/NCT04590053, Identifiers NCT04513470, NCT04590053.
Collapse
Affiliation(s)
| | - Avraham Abutbul
- Medical Intensive Care Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ahmad Naama
- Department of Emergency Medicine, Hadassah-Hebrew University Medical Center and Hebrew University-Hadassah Faculty of Medicine, Jerusalem, Israel
| | - Shlomo Maayan
- Infectious Diseases Division, Barzilai Medical Center, Ashkelon, Israel
| | - Nassar Makram
- Infectious Diseases Division, Barzilai Medical Center, Ashkelon, Israel
| | - Akiva Nachshon
- General Intensive Care Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Kamal abu Jabal
- Ziv Medical Center and Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | | | - Lior Binder
- Enlivex Therapeutics Ltd., Ness Ziona, Israel
| | | | | | - Dror Mevorach
- Enlivex Therapeutics Ltd., Ness Ziona, Israel
- Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- The Institute of Rheumatology-Immunology-Rheumatology, The Wohl Institute for Translational Medicine, Hadassah-Hebrew University Medical Center and Hebrew University-Hadassah Faculty of Medicine, Jerusalem, Israel
| |
Collapse
|
39
|
Sadeghizadeh M, Asadollahi E, Jahangiri B, Yadollahzadeh M, Mohajeri M, Afsharpad M, Najafi F, Rezaie N, Eskandari M, Tavakoli-Ardakani M, Feizabadi F, Masjedi MR. Promising clinical outcomes of nano-curcumin treatment as an adjunct therapy in hospitalized COVID-19 patients: A randomized, double-blinded, placebo-controlled trial. Phytother Res 2023; 37:3631-3644. [PMID: 37118944 DOI: 10.1002/ptr.7844] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 04/01/2023] [Accepted: 04/08/2023] [Indexed: 04/30/2023]
Abstract
Different immunomodulation strategies have been used to manage COVID-19 due to the complex immune-inflammatory processes involved in the pathogenesis of this infection. Curcumin with its powerful anti-inflammatory and antiviral properties could serve as a possible COVID-19 therapy. In this study, a randomized, double-blinded, placebo-controlled trial was performed to investigate the effectiveness and safety of nano-curcumin oral soft gels as a complementary therapy in moderate-severe COVID-19 patients. Hydroxychloroquine (HCQ) plus sofosbuvir was routinely administered to all 42 COVID-19 patients, who were randomly assigned to receive 140 mg of nano-curcumin or placebo for 14 days. CT scans of the chest were taken, and blood tests were run for all patients at time points of 0, 7, and 14 days. Our results indicated that C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) levels significantly decreased from baseline in the nano-curcumin-treated group on day 7. Furthermore, blood levels of D-dimer, CRP, serum ferritin, ESR, and inflammatory cytokines including IL-6, IL-8, and IL-10 decreased more significantly in the nano-curcumin-treated group after 14 days. Additionally, the nano-curcumin group showed significant improvements in chest CT scores, oxygen saturation levels, and hospitalization duration. Based on our data, oral administration of nano-curcumin may be regarded as a promising adjunct treatment for COVID-19 patients due to its ability to speed up chest clearance and recovery.
Collapse
Affiliation(s)
- Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Elahe Asadollahi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Babak Jahangiri
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Mahdi Yadollahzadeh
- Firoozgar Medical & Educational Hospital Department of Internal Medicine School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mandana Afsharpad
- Cancer Control Research Center, Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran
| | - Farhood Najafi
- Department of Resin and Additives, Institute for Color Science and Technology, Tehran, Iran
| | - Nader Rezaie
- Department of Pulmonology, Firouzgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohana Eskandari
- Firoozgar Medical & Educational Hospital Department of Internal Medicine School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maria Tavakoli-Ardakani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Feizabadi
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Masjedi
- Cancer Control Research Center, Cancer Control Foundation, Iran University of Medical Sciences, Tehran, Iran
- Department of Pulmonary Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Tobacco Control Research Center (TCRC), Iranian Anti-tobacco Association, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Nelson AM, Erdmann AA, Coe CL, Juckett MB, Morris K, Knight JM, Hematti P, Costanzo ES. Inflammatory cytokines and depression symptoms following hematopoietic cell transplantation. Brain Behav Immun 2023; 112:11-17. [PMID: 37236325 PMCID: PMC10524437 DOI: 10.1016/j.bbi.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/21/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
Increased synthesis and release of inflammatory signalling proteins is common among individuals with hematologic malignancies undergoing hematopoietic cell transplantation (HCT) due to intensive conditioning regimens and complications such as graft-versus-host-disease and infections. Prior research indicates that inflammatory responses can activate central nervous system pathways that evoke changes in mood. This study examined relationships between markers of inflammatory activity and depression symptoms following HCT. Individuals undergoing allogeneic (n = 84) and autologous (n = 155) HCT completed measures of depression symptoms pre-HCT and 1, 3, and 6 months post-HCT. Proinflammatory (IL-6, TNF-α) and regulatory (IL-10) cytokines were assessed by ELISA in peripheral blood plasma. Mixed-effects linear regression models indicated that patients with elevated IL-6 and IL-10 reported more severe depression symptoms at the post-HCT assessments. These findings were replicated when examining both allogeneic and autologous samples. Follow-up analyses clarified that relationships were strongest for neurovegetative, rather than cognitive or affective, symptoms of depression. These findings suggest that anti-inflammatory therapeutics targeting an inflammatory mediator of depression could improve quality of life of HCT recipients.
Collapse
Affiliation(s)
- Ashley M Nelson
- Department of Psychiatry, Harvard Medical School/Massachusetts General Hospital, Boston, MA, USA
| | - Alexandra A Erdmann
- Department of Pediatrics, Division of Pediatric Cardiology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher L Coe
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark B Juckett
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Keayra Morris
- Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jennifer M Knight
- Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Peiman Hematti
- Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Erin S Costanzo
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA; Department of Medicine, Division of Hematology and Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, WI, USA.
| |
Collapse
|
41
|
Montes-Cobos E, Bastos VC, Monteiro C, de Freitas JC, Fernandes HD, Constancio CS, Rodrigues DA, Gama AM, Vidal VM, Alves LS, Zalcberg-Renault L, de Lira GS, Ota VA, Caloba C, Conde L, Leitão IC, Tanuri A, Ferreira OD, Pereira RM, Vale AM, Castiñeiras TM, Kaiserlian D, Echevarria-Lima J, Bozza MT. Oligosymptomatic long-term carriers of SARS-CoV-2 display impaired innate resistance but increased high-affinity anti-spike antibodies. iScience 2023; 26:107219. [PMID: 37529320 PMCID: PMC10300054 DOI: 10.1016/j.isci.2023.107219] [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: 02/07/2023] [Revised: 05/05/2023] [Accepted: 06/22/2023] [Indexed: 08/03/2023] Open
Abstract
The vast spectrum of clinical features of COVID-19 keeps challenging scientists and clinicians. Low resistance to infection might result in long-term viral persistence, but the underlying mechanisms remain unclear. Here, we studied the immune response of immunocompetent COVID-19 patients with prolonged SARS-CoV-2 infection by immunophenotyping, cytokine and serological analysis. Despite viral loads and symptoms comparable to regular mildly symptomatic patients, long-term carriers displayed weaker systemic IFN-I responses and fewer circulating pDCs and NK cells at disease onset. Type 1 cytokines remained low, while type-3 cytokines were in turn enhanced. Of interest, we observed no defects in antigen-specific cytotoxic T cell responses, and circulating antibodies displayed higher affinity against different variants of SARS-CoV-2 Spike protein in these patients. The identification of distinct immune responses in long-term carriers adds up to our understanding of essential host protective mechanisms to ensure tissue damage control despite prolonged viral infection.
Collapse
Affiliation(s)
- Elena Montes-Cobos
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victoria C. Bastos
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarice Monteiro
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João C.R. de Freitas
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heiny D.P. Fernandes
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarice S. Constancio
- Laboratório de Imunologia Básica e Aplicada, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle A.S. Rodrigues
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andreza M.D.S. Gama
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinicius M. Vidal
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia S. Alves
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laura Zalcberg-Renault
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme S. de Lira
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Victor A. Ota
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina Caloba
- Laboratório de Imunologia Molecular, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isabela C. Leitão
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Amilcar Tanuri
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Orlando D.C. Ferreira
- Laboratório de Virologia Molecular, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata M. Pereira
- Laboratório de Imunologia Molecular, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André M. Vale
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Terezinha M. Castiñeiras
- Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dominique Kaiserlian
- INSERM U1060, Université Claude Bernard Lyon 1, Centre hospitalier Lyon-Sud, Pierre-Benite, France
| | - Juliana Echevarria-Lima
- Laboratório de Imunologia Básica e Aplicada, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Mopuru R. IL-10 Dysregulation in Bipolar Disorder: Potential Mechanisms and Treatment Implications. Indian J Psychol Med 2023; 45:439. [PMID: 37483581 PMCID: PMC10357918 DOI: 10.1177/02537176231154270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Affiliation(s)
- Ranesh Mopuru
- Centre for Fundamental Research and Creative Education, Bengaluru, Karnataka, India
| |
Collapse
|
43
|
Avram I, Pelinescu D, Gatea F, Ionescu R, Barcan A, Rosca R, Zanfirescu A, Vamanu E. Boletus edulis Extract-A New Modulator of Dysbiotic Microbiota. Life (Basel) 2023; 13:1481. [PMID: 37511858 PMCID: PMC10381576 DOI: 10.3390/life13071481] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
The regular administration of antibiotics is a public concern due to the prejudices of large population groups and the high frequency with which antimicrobial products are prescribed. The current study aimed to evaluate the in vitro effect of a new extract from Boletus edulis (BEE) on the human microbiota. One of the disadvantages of this extensive use is the disruption of the human microbiota, leading to potential negative health consequences. The in vitro evaluation of BEE consisted in determining its cytotoxicity, influence on the concentration of four types of cytokines (IL-6, IL-10, IL-1β, TNFα), and capacity to modulate the human microbiota after administering antibiotics. The latter was assessed by microbiome analysis and the evaluation of short-chain fatty acid synthesis (SCFAs). Simultaneously, the content of total polyphenols, the antioxidant capacity, and the compositional analysis of the extract (individual polyphenols composition) were determined. The results showed that BEE modulates the microbial pattern and reduces inflammatory progression. The data demonstrated antioxidant properties correlated with the increase in synthesizing some biomarkers, such as SCFAs, which mitigated antibiotic-induced dysbiosis without using probiotic products.
Collapse
Affiliation(s)
- Ionela Avram
- Department of Genetics, University of Bucharest, 36-46 Bd. M. Kogalniceanu, 5th District, 050107 Bucharest, Romania
| | - Diana Pelinescu
- Department of Genetics, University of Bucharest, 36-46 Bd. M. Kogalniceanu, 5th District, 050107 Bucharest, Romania
| | - Florentina Gatea
- Centre of Bioanalysis, National Institute for Biological Sciences, 296 Spl. Independentei, 060031 Bucharest, Romania
| | - Robertina Ionescu
- Department of Genetics, University of Bucharest, 36-46 Bd. M. Kogalniceanu, 5th District, 050107 Bucharest, Romania
| | - Alexandru Barcan
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania
| | - Razvan Rosca
- Anoom Laboratories SRL, București, 28 Vintila Mihaileanu Sector 1, 024023 Bucharest, Romania
| | - Anca Zanfirescu
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania
| |
Collapse
|
44
|
Chang WJ, Jenkins LC, Humburg P, Wasinger V, Walton DM, Schabrun SM. Systemic pro- and anti-inflammatory profiles in acute non-specific low back pain: An exploratory longitudinal study of the relationship to six-month outcome. PLoS One 2023; 18:e0287192. [PMID: 37384753 PMCID: PMC10309993 DOI: 10.1371/journal.pone.0287192] [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: 10/13/2022] [Accepted: 06/01/2023] [Indexed: 07/01/2023] Open
Abstract
OBJECTIVES Pro-inflammatory molecules are thought to underpin the development of chronic low back pain (LBP). Although research has begun to explore the association between pro-inflammatory molecules in acute LBP and long-term outcome, no study has explored the role of anti-inflammatory molecules. We aimed to explore whether levels of systemic pro- and anti-inflammatory molecules 1) changed over a period of six months from the onset of acute LBP; 2) differed between people who were recovered (N = 11) and unrecovered (N = 24) from their episode of LBP at six months; 3) baseline psychological factors were related to inflammatory molecule serum concentrations at baseline, three and six months. METHODS We retrospectively included participants with acute LBP included from a larger prospective trial and examined blood samples for the measurement of pro- and anti-inflammatory molecules and measures of pain, disability, and psychological factors at baseline, three and six months. RESULTS The serum concentrations of pro- and anti-inflammatory molecules did not differ over time when compared between participants who recovered and those who did not recover at six-month follow-up. At three months, the unrecovered group had higher interleukin (IL)-8 and IL-10 serum concentrations than the recovered group. Baseline psychological factors were not related to inflammatory molecules at any time point. DISCUSSION This exploratory study showed that levels of systemic inflammatory molecules did not change over the course of LBP, irrespective of whether people were recovered or unrecovered at six months. There was no relationship between acute-stage psychological factors and systemic inflammatory molecules. Further investigation is needed to elucidate the contribution of pro- and anti-inflammatory molecules to long-term LBP outcome.
Collapse
Affiliation(s)
- Wei-Ju Chang
- Centre for Pain IMPACT, Neuroscience Research Australia (NeuRA), Randwick, New South Wales, Australia
- School of Health Sciences, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| | - Luke C. Jenkins
- Centre for Pain IMPACT, Neuroscience Research Australia (NeuRA), Randwick, New South Wales, Australia
- School of Health Sciences, Western Sydney University, Penrith, New South Wales, Australia
| | - Peter Humburg
- Stats Central, Mark Wainwright Analytical Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - Valerie Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW Sydney, Kensington, New South Wales, Australia
| | - David M. Walton
- School of Physical Therapy, University of Western Ontario, London, Ontario, Canada
| | - Siobhan M. Schabrun
- Centre for Pain IMPACT, Neuroscience Research Australia (NeuRA), Randwick, New South Wales, Australia
- School of Physical Therapy, University of Western Ontario, London, Ontario, Canada
- The Gray Centre for Mobility and Activity, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
45
|
Gamer J, Van Booven DJ, Zarnowski O, Arango S, Elias M, Kurian A, Joseph A, Perez M, Collado F, Klimas N, Oltra E, Nathanson L. Sex-Dependent Transcriptional Changes in Response to Stress in Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Pilot Project. Int J Mol Sci 2023; 24:10255. [PMID: 37373402 PMCID: PMC10299261 DOI: 10.3390/ijms241210255] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/23/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex, multi-symptom illness characterized by debilitating fatigue and post-exertional malaise (PEM). Numerous studies have reported sex differences at the epidemiological, cellular, and molecular levels between male and female ME/CFS patients. To gain further insight into these sex-dependent changes, we evaluated differential gene expression by RNA-sequencing (RNA-Seq) in 33 ME/CFS patients (20 female, 13 male) and 34 matched healthy controls (20 female and 14 male) before, during, and after an exercise challenge intended to provoke PEM. Our findings revealed that pathways related to immune-cell signaling (including IL-12) and natural killer cell cytotoxicity were activated as a result of exertion in the male ME/CFS cohort, while female ME/CFS patients did not show significant enough changes in gene expression to meet the criteria for the differential expression. Functional analysis during recovery from an exercise challenge showed that male ME/CFS patients had distinct changes in the regulation of specific cytokine signals (including IL-1β). Meanwhile, female ME/CFS patients had significant alterations in gene networks related to cell stress, response to herpes viruses, and NF-κβ signaling. The functional pathways and differentially expressed genes highlighted in this pilot project provide insight into the sex-specific pathophysiology of ME/CFS.
Collapse
Affiliation(s)
- Jackson Gamer
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.G.); (N.K.)
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
| | - Derek J. Van Booven
- Dr. J.P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Oskar Zarnowski
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
| | - Sebastian Arango
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
| | - Mark Elias
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
| | - Asha Kurian
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
| | - Andrew Joseph
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
| | - Melanie Perez
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
| | - Fanny Collado
- Department of Veterans Affairs, Miami VA Healthcare System, Geriatric Research Education and Clinical Center (GRECC), Miami, FL 33125, USA;
- South Florida Veterans Affairs Foundation for Research and Education Inc., Fort Lauderdale, FL 33125, USA
| | - Nancy Klimas
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.G.); (N.K.)
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
- Department of Veterans Affairs, Miami VA Healthcare System, Geriatric Research Education and Clinical Center (GRECC), Miami, FL 33125, USA;
- South Florida Veterans Affairs Foundation for Research and Education Inc., Fort Lauderdale, FL 33125, USA
| | - Elisa Oltra
- School Medicine, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain;
| | - Lubov Nathanson
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (J.G.); (N.K.)
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (O.Z.); (S.A.); (M.E.); (A.K.); (A.J.); (M.P.)
| |
Collapse
|
46
|
Csobonyeiova M, Smolinska V, Harsanyi S, Ivantysyn M, Klein M. The Immunomodulatory Role of Cell-Free Approaches in SARS-CoV-2-Induced Cytokine Storm-A Powerful Therapeutic Tool for COVID-19 Patients. Biomedicines 2023; 11:1736. [PMID: 37371831 DOI: 10.3390/biomedicines11061736] [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: 05/23/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Currently, there is still no effective and definitive cure for the coronavirus disease 2019 (COVID-19) caused by the infection of the novel highly contagious severe acute respiratory syndrome virus (SARS-CoV-2), whose sudden outbreak was recorded for the first time in China in late December 2019. Soon after, COVID-19 affected not only the vast majority of China's population but the whole world and caused a global health public crisis as a new pandemic. It is well known that viral infection can cause acute respiratory distress syndrome (ARDS) and, in severe cases, can even be lethal. Behind the inflammatory process lies the so-called cytokine storm (CS), which activates various inflammatory cytokines that damage numerous organ tissues. Since the first outbreak of SARS-CoV-2, various research groups have been intensively trying to investigate the best treatment options; however, only limited outcomes have been achieved. One of the most promising strategies represents using either stem cells, such as mesenchymal stem cells (MSCs)/induced pluripotent stem cells (iPSCs), or, more recently, using cell-free approaches involving conditioned media (CMs) and their content, such as extracellular vesicles (EVs) (e.g., exosomes or miRNAs) derived from stem cells. As key mediators of intracellular communication, exosomes carry a cocktail of different molecules with anti-inflammatory effects and immunomodulatory capacity. Our comprehensive review outlines the complex inflammatory process responsible for the CS, summarizes the present results of cell-free-based pre-clinical and clinical studies for COVID-19 treatment, and discusses their future perspectives for therapeutic applications.
Collapse
Affiliation(s)
- Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Apel, Dunajská 52, 811 08 Bratislava, Slovakia
- Regenmed Ltd., Medená 29, 811 08 Bratislava, Slovakia
| | - Veronika Smolinska
- Regenmed Ltd., Medená 29, 811 08 Bratislava, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stefan Harsanyi
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | | | - Martin Klein
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
47
|
Carlini V, Noonan DM, Abdalalem E, Goletti D, Sansone C, Calabrone L, Albini A. The multifaceted nature of IL-10: regulation, role in immunological homeostasis and its relevance to cancer, COVID-19 and post-COVID conditions. Front Immunol 2023; 14:1161067. [PMID: 37359549 PMCID: PMC10287165 DOI: 10.3389/fimmu.2023.1161067] [Citation(s) in RCA: 116] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Interleukin-10 (IL-10) is a pleiotropic cytokine that has a fundamental role in modulating inflammation and in maintaining cell homeostasis. It primarily acts as an anti-inflammatory cytokine, protecting the body from an uncontrolled immune response, mostly through the Jak1/Tyk2 and STAT3 signaling pathway. On the other hand, IL-10 can also have immunostimulating functions under certain conditions. Given the pivotal role of IL-10 in immune modulation, this cytokine could have relevant implications in pathologies characterized by hyperinflammatory state, such as cancer, or infectious diseases as in the case of COVID-19 and Post-COVID-19 syndrome. Recent evidence proposed IL-10 as a predictor of severity and mortality for patients with acute or post-acute SARS-CoV-2 infection. In this context, IL-10 can act as an endogenous danger signal, released by tissues undergoing damage in an attempt to protect the organism from harmful hyperinflammation. Pharmacological strategies aimed to potentiate or restore IL-10 immunomodulatory action may represent novel promising avenues to counteract cytokine storm arising from hyperinflammation and effectively mitigate severe complications. Natural bioactive compounds, derived from terrestrial or marine photosynthetic organisms and able to increase IL-10 expression, could represent a useful prevention strategy to curb inflammation through IL-10 elevation and will be discussed here. However, the multifaceted nature of IL-10 has to be taken into account in the attempts to modulate its levels.
Collapse
Affiliation(s)
- Valentina Carlini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Douglas M. Noonan
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Eslam Abdalalem
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Clementina Sansone
- Stazione Zoologica Anton Dohrn, Istituto Nazionale di Biologia, Ecologia e Biotecnologie Marine, Napoli, Italy
| | - Luana Calabrone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Adriana Albini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) European Institute of Oncology IEO-, Milan, Italy
| |
Collapse
|
48
|
Biswas B, Roy S, Banerjee I, Jana S, Bhattacharjee B, Chakraborty S, Mondal A, Goswami R. IL-10/IL-6 ratio from nasal & oral swab samples, acts as an inflammatory indicator for COVID-19 patients infected with the delta variant. Heliyon 2023; 9:e16985. [PMID: 37292329 PMCID: PMC10238278 DOI: 10.1016/j.heliyon.2023.e16985] [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: 10/26/2022] [Revised: 05/25/2023] [Accepted: 06/02/2023] [Indexed: 06/10/2023] Open
Abstract
Background Hyper-inflammatory immune response of SARS-CoV-2 is often characterized by the release of multiple pro-inflammatory cytokines with an impact on the expression of numerous other interleukins (ILs). However, from oral and nasal swab samples the specific quantitative association of the different IL-markers with the disease progression and its relationship with the status of vaccination remains unclear. Materials and methods Patients' combined oral and nasal swab samples were collected from both non-vaccinated and double-vaccinated individuals with high (Ct value < 25) and low (Ct value > 30) viral loads, along with uninfected donors. None of the patients were critically ill, or needed ICU support. The expression of different cytokines (IL6, IL10, IL1B, IFNG) and mucin (MUC5AC, MUC1) markers were assessed between different groups by qRT-PCR. The important cytokine markers differentiating between vaccinated and non-vaccinated patients were identified by PCA. Conclusion IL6 expression was higher in non-vaccinated COVID-19 patients infected with delta-variant irrespective of their viral-load compared to uninfected individuals. However, in double-vaccinated patients, only in high viral-load patients (Ct value < 25), IL6 expression increased. In high viral-load patients, irrespective to their vaccination status, IL10 expression was lower compared to the uninfected control group. Surprisingly, IL10 expression was lower in double-vaccinated patients with Ct value > 30. IL1B, and IFNG expression remained unaltered in uninfected and infected individuals. However, MUC5AC expression was lower in non-vaccinated patients with Ct value < 25 compared to control group. Our study unveiled that IL10/IL6 ratio can be used as a biomarker for COVID-19 patients upon proper establishment of it in a clinical setting.
Collapse
Affiliation(s)
| | | | | | - Subhasis Jana
- Purba Medinipur District Hospital, West Bengal, India
| | | | | | | | | |
Collapse
|
49
|
Ribeiro DM, Lopes PA, Pinto RMA, Pestana JM, Costa MM, Alfaia CM, Mourato MP, de Almeida AM, Freire JPB, Prates JAM. Dietary Ulva lactuca and CAZyme supplementation improve serum biochemical profile and hepatic composition of weaned piglets. Sci Rep 2023; 13:8784. [PMID: 37258799 DOI: 10.1038/s41598-023-36008-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/27/2023] [Indexed: 06/02/2023] Open
Abstract
Ulva lactuca is a seaweed with antinutritional cell wall for monogastrics. Carbohydrate-Active enZymes (CAZymes) supplementation can potentially cause its disruption. This study evaluates four diets: Ctrl-control diet; UL-control + 7% U. lactuca (wild caught, powdered form); ULR-UL + 0.005% Rovabio® Excel AP; ULU-UL + 0.01% ulvan lyase on piglets' haematologic and serologic profiles, hepatic lipids and minerals. White blood cells and lymphocytes reached the highest values in piglets fed UL compared to control, and to control and ULR; respectively (P < 0.05). IgG levels were boosted by seaweed incorporation compared to control (P = 0.015). The glycaemic homeostasis was assured by the seaweed inclusion. Dietary seaweed decreased serum lipids (P < 0.001), with the exception of ULU, due to HDL-cholesterol increase (P < 0.001). Cortisol was decreased in ULR and ULU (P < 0.001). No systemic inflammation was observed (P > 0.05). While hepatic n-3 PUFA increased in piglets fed with seaweed diets due to increment of beneficial 22:5n-3 and 22:6n-3 fatty acids (P < 0.05), the opposite occurred for n-6 PUFA, PUFA/SFA and n-6/n-3 ratios (P < 0.05). Hepatic pigments were unchanged (P > 0.05). ULR reduced α-tocopherol levels (P = 0.036) and increased serum potassium levels (P < 0.001) compared to control. Seaweed contributed to overcome piglets' weaning stress, with some benefits of including CAZyme supplementation.
Collapse
Affiliation(s)
- David M Ribeiro
- LEAF - Linking Landscape, Environment, Agriculture and Food Research Center, Associated Laboratory TERRA, Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017, Lisboa, Portugal
| | - Paula A Lopes
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477, Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Lisboa, Portugal
| | - Rui M A Pinto
- iMED.UL, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisboa, Portugal
- JCS, Laboratório de Análises Clínicas Dr. Joaquim Chaves, Avenida General Norton de MatosMiraflores, 1495-148, Algés, Portugal
| | - José M Pestana
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477, Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Lisboa, Portugal
| | - Mónica M Costa
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477, Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Lisboa, Portugal
| | - Cristina M Alfaia
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477, Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Lisboa, Portugal
| | - Miguel P Mourato
- LEAF - Linking Landscape, Environment, Agriculture and Food Research Center, Associated Laboratory TERRA, Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017, Lisboa, Portugal
| | - André M de Almeida
- LEAF - Linking Landscape, Environment, Agriculture and Food Research Center, Associated Laboratory TERRA, Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017, Lisboa, Portugal
| | - João P B Freire
- LEAF - Linking Landscape, Environment, Agriculture and Food Research Center, Associated Laboratory TERRA, Instituto Superior de Agronomia, Universidade de Lisboa, Tapada da Ajuda, 1349-017, Lisboa, Portugal
| | - José A M Prates
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477, Lisboa, Portugal.
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Lisboa, Portugal.
| |
Collapse
|
50
|
Hwang IC, Valeriano VD, Song JH, Pereira M, Oh JK, Han K, Engstrand L, Kang DK. Mucosal immunization with lactiplantibacillus plantarum-displaying recombinant SARS-CoV-2 epitopes on the surface induces humoral and mucosal immune responses in mice. Microb Cell Fact 2023; 22:96. [PMID: 37161468 PMCID: PMC10169176 DOI: 10.1186/s12934-023-02100-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND The use of probiotic lactic acid bacteria as a mucosal vaccine vector is considered a promising alternative compared to the use of other microorganisms because of its "Generally Regarded as Safe" status, its potential adjuvant properties, and its tolerogenicity to the host. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease (COVID-19), is highly transmissible and pathogenic. This study aimed to determine the potential of Lactiplantibacillus plantarum expressing SARS-CoV-2 epitopes as a mucosal vaccine against SARS-CoV-2. RESULTS In this study, the possible antigenic determinants of the spike (S1-1, S1-2, S1-3, and S1-4), membrane (ME1 and ME2), and envelope (E) proteins of SARS-CoV-2 were predicted, and recombinant L. plantarum strains surface-displaying these epitopes were constructed. Subsequently, the immune responses induced by these recombinant strains were compared in vitro and in vivo. Most surface-displayed epitopes induced pro-inflammatory cytokines [tumor necrosis factor alpha (TNF-α and interleukin (IL)-6] and anti-inflammatory cytokines (IL-10) in lipopolysaccharide-induced RAW 264.7, with the highest anti-inflammatory to pro-inflammatory cytokine ratio in the S1-1 and S1-2 groups, followed by that in the S1-3 group. When orally administered of recombinant L. plantarum expressing SARS-CoV-2 epitopes in mice, all epitopes most increased the expression of IL-4, along with induced levels of TNF-α, interferon-gamma, and IL-10, specifically in spike protein groups. Thus, the surface expression of epitopes from the spike S1 protein in L. plantarum showed potential immunoregulatory effects, suggesting its ability to potentially circumvent hyperinflammatory states relevant to monocyte/macrophage cell activation. At 35 days post immunization (dpi), serum IgG levels showed a marked increase in the S1-1, S1-2, and S1-3 groups. Fecal IgA levels increased significantly from 21 dpi in all the antigen groups, but the boosting effect after 35 dpi was explicitly observed in the S1-1, S1-2, and S1-3 groups. Thus, the oral administration of SARS-CoV-2 antigens into mice induced significant humoral and mucosal immune responses. CONCLUSION This study suggests that L. plantarum is a potential vector that can effectively deliver SARS-CoV-2 epitopes to intestinal mucosal sites and could serve as a novel approach for SARS-CoV-2 mucosal vaccine development.
Collapse
Affiliation(s)
- In-Chan Hwang
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea
| | - Valerie Diane Valeriano
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Ji Hoon Song
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea
| | - Marcela Pereira
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Ju Kyoung Oh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Kyudong Han
- Department of Microbiology, College of Science and Technology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Lars Engstrand
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|